1
|
Arellano MYG, VanHeest M, Emmadi S, Abdul-Hafez A, Ibrahim SA, Thiruvenkataramani RP, Teleb RS, Omar H, Kesaraju T, Mohamed T, Madhukar BV, Omar SA. Role of Mesenchymal Stem/Stromal Cells (MSCs) and MSC-Derived Extracellular Vesicles (EVs) in Prevention of Telomere Length Shortening, Cellular Senescence, and Accelerated Biological Aging. Bioengineering (Basel) 2024; 11:524. [PMID: 38927760 PMCID: PMC11200821 DOI: 10.3390/bioengineering11060524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 06/28/2024] Open
Abstract
Biological aging is defined as a progressive decline in tissue function that eventually results in cell death. Accelerated biologic aging results when the telomere length is shortened prematurely secondary to damage from biological or environmental stressors, leading to a defective reparative mechanism. Stem cells therapy may have a potential role in influencing (counteract/ameliorate) biological aging and maintaining the function of the organism. Mesenchymal stem cells, also called mesenchymal stromal cells (MSCs) are multipotent stem cells of mesodermal origin that can differentiate into other types of cells, such as adipocytes, chondrocytes, and osteocytes. MSCs influence resident cells through the secretion of paracrine bioactive components such as cytokines and extracellular vesicles (EVs). This review examines the changes in telomere length, cellular senescence, and normal biological age, as well as the factors contributing to telomere shortening and accelerated biological aging. The role of MSCs-especially those derived from gestational tissues-in prevention of telomere shortening (TS) and accelerated biological aging is explored. In addition, the strategies to prevent MSC senescence and improve the antiaging therapeutic application of MSCs and MSC-derived EVs in influencing telomere length and cellular senescence are reviewed.
Collapse
Affiliation(s)
- Myrna Y. Gonzalez Arellano
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Regional Neonatal Intensive Care Unit, Sparrow Hospital, Lansing, MI 48912, USA
| | - Matthew VanHeest
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
| | - Sravya Emmadi
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
| | - Amal Abdul-Hafez
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
| | - Sherif Abdelfattah Ibrahim
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Histology and Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Ranga P. Thiruvenkataramani
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Regional Neonatal Intensive Care Unit, Sparrow Hospital, Lansing, MI 48912, USA
| | - Rasha S. Teleb
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Department of Pediatrics and Neonatology, Qena Faculty of Medicine, South Valley University, Qena 83523, Egypt
| | - Hady Omar
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
| | - Tulasi Kesaraju
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
| | - Tarek Mohamed
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Regional Neonatal Intensive Care Unit, Sparrow Hospital, Lansing, MI 48912, USA
| | - Burra V. Madhukar
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
| | - Said A. Omar
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Regional Neonatal Intensive Care Unit, Sparrow Hospital, Lansing, MI 48912, USA
| |
Collapse
|
2
|
Cord Blood Plasma and Placental Mesenchymal Stem Cells-Derived Exosomes Increase Ex Vivo Expansion of Human Cord Blood Hematopoietic Stem Cells While Maintaining Their Stemness. Cells 2023; 12:cells12020250. [PMID: 36672185 PMCID: PMC9857343 DOI: 10.3390/cells12020250] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/23/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have been used for ex vivo expansion of umbilical cord blood (UCB) hematopoietic stem cells (HSCs) to maintain their primitive characters and long-term reconstitution abilities during transplantation. Therapeutic effects of MSCs mainly rely on paracrine mechanisms, including secretion of exosomes (Exos). The objective of this study was to examine the effect of cord blood plasma (CBP)-derived Exos (CBP Exos) and Placental MSCs-derived Exos (MSCs Exos) on the expansion of UCB HSCs to increase their numbers and keep their primitive characteristics. METHODS CD34+ cells were isolated from UCB, cultured for 10 days, and the expanded HSCs were sub-cultured in semisolid methylcellulose media for primitive colony forming units (CFUs) assay. MSCs were cultured from placental chorionic plates. RESULTS CBP Exos and MSCs Exos compared with the control group significantly increased the number of total nucleated cells (TNCs), invitro expansion of CD34+ cells, primitive subpopulations of CD34+38+ and CD34+38-Lin- cells (p < 0.001). The expanded cells showed a significantly higher number of total CFUs in the Exos groups (p < 0.01). CONCLUSION CBP- and placental-derived exosomes are associated with significant ex vivo expansion of UCB HSCs, while maintaining their primitive characters and may eliminate the need for transplantation of an additional unit of UCB.
Collapse
|
3
|
Bruschi M, Vanzolini T, Sahu N, Balduini A, Magnani M, Fraternale A. Functionalized 3D scaffolds for engineering the hematopoietic niche. Front Bioeng Biotechnol 2022; 10:968086. [PMID: 36061428 PMCID: PMC9428512 DOI: 10.3389/fbioe.2022.968086] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/13/2022] [Indexed: 11/16/2022] Open
Abstract
Hematopoietic stem cells (HSCs) reside in a subzone of the bone marrow (BM) defined as the hematopoietic niche where, via the interplay of differentiation and self-renewal, they can give rise to immune and blood cells. Artificial hematopoietic niches were firstly developed in 2D in vitro cultures but the limited expansion potential and stemness maintenance induced the optimization of these systems to avoid the total loss of the natural tissue complexity. The next steps were adopted by engineering different materials such as hydrogels, fibrous structures with natural or synthetic polymers, ceramics, etc. to produce a 3D substrate better resembling that of BM. Cytokines, soluble factors, adhesion molecules, extracellular matrix (ECM) components, and the secretome of other niche-resident cells play a fundamental role in controlling and regulating HSC commitment. To provide biochemical cues, co-cultures, and feeder-layers, as well as natural or synthetic molecules were utilized. This review gathers key elements employed for the functionalization of a 3D scaffold that demonstrated to promote HSC growth and differentiation ranging from 1) biophysical cues, i.e., material, topography, stiffness, oxygen tension, and fluid shear stress to 2) biochemical hints favored by the presence of ECM elements, feeder cell layers, and redox scavengers. Particular focus is given to the 3D systems to recreate megakaryocyte products, to be applied for blood cell production, whereas HSC clinical application in such 3D constructs was limited so far to BM diseases testing.
Collapse
Affiliation(s)
- Michela Bruschi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
- *Correspondence: Michela Bruschi,
| | - Tania Vanzolini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Neety Sahu
- Department of Orthopedic Surgery, School of Medicine, Stanford University, Stanford, CA, United States
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | | |
Collapse
|
4
|
Vilabril S, Nadine S, Neves CMSS, Correia CR, Freire MG, Coutinho JAP, Oliveira MB, Mano JF. One-Step All-Aqueous Interfacial Assembly of Robust Membranes for Long-Term Encapsulation and Culture of Adherent Stem/Stromal Cells. Adv Healthc Mater 2021; 10:e2100266. [PMID: 33764007 DOI: 10.1002/adhm.202100266] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Indexed: 02/06/2023]
Abstract
The therapeutic effectiveness and biological relevance of technologies based on adherent cells depend on platforms that enable long-term culture in controlled environments. Liquid-core capsules have been suggested as semipermeable moieties with spatial homogeneity due to the high mobility of all components in their core. The lack of cell-adhesive sites in liquid-core structures often hampers their use as platforms for stem cell-based technologies for long-term survival and cell-directed self-organization. Here, the one-step fast formation of robust polymeric capsules formed by interfacial complexation of oppositely charged polyelectrolytes in an all-aqueous environment, compatible with the simultaneous encapsulation of mesenchymal stem/stromal cells (MSCs) and microcarriers, is described. The adhesion of umbilical cord MSCs to polymeric microcarriers enables their aggregation and culture for more than 21 days in capsules prepared either manually by dropwise addition, or by scalable electrohydrodynamic atomization, generating robust and stable capsules. Cell aggregation and secretion overtime can be tailored by providing cells with static or dynamic (bioreactor) environments.
Collapse
Affiliation(s)
- Sara Vilabril
- CICECO ‐ Aveiro Institute of Materials Department of Chemistry University of Aveiro Aveiro 3810‐193 Portugal
| | - Sara Nadine
- CICECO ‐ Aveiro Institute of Materials Department of Chemistry University of Aveiro Aveiro 3810‐193 Portugal
| | - Catarina M. S. S. Neves
- CICECO ‐ Aveiro Institute of Materials Department of Chemistry University of Aveiro Aveiro 3810‐193 Portugal
| | - Clara R. Correia
- CICECO ‐ Aveiro Institute of Materials Department of Chemistry University of Aveiro Aveiro 3810‐193 Portugal
| | - Mara G. Freire
- CICECO ‐ Aveiro Institute of Materials Department of Chemistry University of Aveiro Aveiro 3810‐193 Portugal
| | - João A. P. Coutinho
- CICECO ‐ Aveiro Institute of Materials Department of Chemistry University of Aveiro Aveiro 3810‐193 Portugal
| | - Mariana B. Oliveira
- CICECO ‐ Aveiro Institute of Materials Department of Chemistry University of Aveiro Aveiro 3810‐193 Portugal
| | - João F. Mano
- CICECO ‐ Aveiro Institute of Materials Department of Chemistry University of Aveiro Aveiro 3810‐193 Portugal
| |
Collapse
|
5
|
Lerman MJ, Smith BT, Gerald AG, Santoro M, Fookes JA, Mikos AG, Fisher JP. Aminated 3D Printed Polystyrene Maintains Stem Cell Proliferation and Osteogenic Differentiation. Tissue Eng Part C Methods 2020; 26:118-131. [PMID: 31971874 PMCID: PMC7041340 DOI: 10.1089/ten.tec.2019.0217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/18/2019] [Indexed: 12/14/2022] Open
Abstract
As 3D printing becomes more common and the technique is used to build culture platforms, it is imperative to develop surface treatments for specific responses. The advantages of aminating and oxidizing polystyrene (PS) for human mesenchymal stem cell (hMSC) proliferation and osteogenic differentiation are investigated. We find that ammonia (NH3) plasma incorporates amines while oxygen plasma adds carbonyl and carboxylate groups. Across 2D, 3D, and 3D dynamic culture, we find that the NH3- treated surfaces encouraged cell proliferation. Our results show that the NH3-treated scaffold was the only treatment allowing dynamic proliferation of hMSCs with little evidence of osteogenic differentiation. With osteogenic media, particularly in 3D culture, we find the NH3 treatment encouraged greater and earlier expression of RUNX2 and ALP. The NH3-treated PS scaffolds support hMSC proliferation without spontaneous osteogenic differentiation in static and dynamic culture. This work provides an opportunity for further investigations into shear profiling and coculture within the developed culture system toward developing a bone marrow niche model.
Collapse
Affiliation(s)
- Max J. Lerman
- Department of Materials Science and Engineering, University of Maryland, College Park, Maryland
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland
| | - Brandon T. Smith
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland
- Department of Bioengineering, MS-142 BioScience Research Collaborative, Rice University, Houston, Texas
| | - Anushka G. Gerald
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Marco Santoro
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - James A. Fookes
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Antonios G. Mikos
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland
- Department of Bioengineering, MS-142 BioScience Research Collaborative, Rice University, Houston, Texas
| | - John P. Fisher
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| |
Collapse
|
6
|
Li Q, Zhao D, Chen Q, Luo M, Huang J, Yang C, Wang F, Li W, Liu T. Wharton's jelly mesenchymal stem cell-based or umbilical vein endothelial cell-based serum-free coculture with cytokines supports the ex vivo expansion/maintenance of cord blood hematopoietic stem/progenitor cells. Stem Cell Res Ther 2019; 10:376. [PMID: 31806004 PMCID: PMC6894464 DOI: 10.1186/s13287-019-1502-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 11/14/2019] [Accepted: 11/20/2019] [Indexed: 02/05/2023] Open
Abstract
Background The umbilical cord blood (UCB) has been widely accepted as an alternative source of hematopoietic stem/progenitor cells (HSPCs) for transplantation, and its use in adults is still restricted because of low absolute numbers. To overcome this obstacle, expansion of UCB-HSPCs under feeder cell-based coculture is a promising possibility. In this study, we explored UCB-CD34+ cells ex vivo expansion using Wharton’s jelly mesenchymal stem cells (WJ-MSCs) or umbilical vein endothelial cells (UVECs) as feeder layer-based serum-free coculture system with a cocktail of cytokines. Methods UCB-CD34+ cells were cultured in five different coculture conditions composed of umbilical cord stromal cells (WJ-MSCs or UVECs) with or without a cocktail of cytokines (SCF, FLT3L, and TPO). The cultured cells were harvested at day 10 and analyzed for phenotypes and functionalities, including total nuclear cells (TNCs), CD34+ cells, CD34+CD38− cells, colony-forming unit (CFU) for committed progenitors, and long-term culture initiating cells (LTC-ICs) for HSPCs. Results Our work showed the numbers of TNC cells, CD34+ cells, and CD34+CD38− cells were expanded under five coculture conditions, and the feeder layer-based cocultures further promoted the expansion. The numbers of colonies of CFU-GM, CFU-E/BFU-E, and CFU-GEMM in the cocultures with cytokines were significantly higher than their counterparts at day 0 (p < 0.05), while no significant difference (p > 0.05) in those without the addition of cytokines. The numbers of LTC-ICs were increased both under the WJ-MSCs and UVECs with cytokine cocultures, but only in the UVECs group showed a significant difference (p < 0.05), and were decreased under conditions without cytokine (p < 0.05). Conclusion Our data demonstrate that both WJ-MSCs and UVECs as feeder layer could efficiently support the expansion of UCB-CD34+ cells in synergy with SCF, FLT3L, and TPO under serum-free culture condition. The UVECs combined with the 3GF cytokine cocktail could maintain the growth of LTC-ICs derived from UCB-CD34+ cells and even expand to some extent.
Collapse
Affiliation(s)
- Qiuyang Li
- Department of Hematology, Hematology Research Laboratory, West China Hospital of Sichuan University, #37 Guo Xue Xiang Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Dewan Zhao
- Department of Hematology, Hematology Research Laboratory, West China Hospital of Sichuan University, #37 Guo Xue Xiang Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Qiang Chen
- Sichuan Cord Blood Stem Cell Bank, Chengdu, Sichuan, People's Republic of China
| | - Maowen Luo
- Sichuan Cord Blood Stem Cell Bank, Chengdu, Sichuan, People's Republic of China
| | - Jingcao Huang
- Department of Hematology, Hematology Research Laboratory, West China Hospital of Sichuan University, #37 Guo Xue Xiang Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Cao Yang
- Sichuan Cord Blood Stem Cell Bank, Chengdu, Sichuan, People's Republic of China
| | - Fangfang Wang
- Department of Hematology, Hematology Research Laboratory, West China Hospital of Sichuan University, #37 Guo Xue Xiang Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Wenxian Li
- Sichuan Cord Blood Stem Cell Bank, Chengdu, Sichuan, People's Republic of China
| | - Ting Liu
- Department of Hematology, Hematology Research Laboratory, West China Hospital of Sichuan University, #37 Guo Xue Xiang Street, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
7
|
Adipose-derived stromal cell immunosuppression of T cells is enhanced under "physiological" hypoxia. Tissue Cell 2019; 63:101320. [PMID: 32223948 DOI: 10.1016/j.tice.2019.101320] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/12/2019] [Accepted: 12/02/2019] [Indexed: 01/10/2023]
Abstract
Multipotent mesenchymal stromal cells (MSCs) are characterized by immunomodulatory properties along with the high proliferative and paracrine activity, as well as multilineage potency. The effects of MSCs on the T cell adaptive immunity are of a special interest. Low O2 level (1-7 %) is known to be typical for the putative site of the MSC - T cell interactions. A comparative evaluation of the effects of adipose tissue derived MSC (ASCs) on the mitogen-stimulated T cells at the ambient (20 %) and tissue-related (5 %) O2 levels demonstrated reduced T cell activation by the HLA-DR expression, decreased pro-inflammatory and increased anti-inflammatory cytokine production in co-culture, inhibited T cell proliferation, with the effects increased at hypoxia. T cell interactions with ASCs resulted in the up-regulation of PDCD1, Foxp3, and TGFβ1 known to play an important role in the immune response suppression, and in the down-regulation of genes involved in the inflammatory reaction (IL2, IFNG). These changes were significantly increased under hypoxia. At the same time, neither ASCs nor the reduced O2 level had negative effects on the viability of T cells.
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW Herein, we seek to describe the current and future role of ex-vivo expansion of cord blood hematopoietic stem cells. RECENT FINDINGS As this field is only in its infancy, there have been many challenges identified. Decreased number of stem cells contained in a cord blood unit and early differentiation of stem cells once expanded have been two overarching challenges faced by the field. Many recent techniques have focused on the properties of the microenvironment and targetable cellular pathways as novel approaches to circumvent these challenges. SUMMARY Novel discoveries have led to the development of approaches that will increase hematopoietic stem cell yield and will improve engraftment in patients receiving cord blood hematopoietic stem cell transplantation. As a result, patients receiving cord blood hematopoietic stem cell transplantationcontinue to have improved outcomes.
Collapse
|
9
|
Human multipotent hematopoietic progenitor cell expansion is neither supported in endothelial and endothelial/mesenchymal co-cultures nor in NSG mice. Sci Rep 2019; 9:12914. [PMID: 31501490 PMCID: PMC6733927 DOI: 10.1038/s41598-019-49221-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 08/12/2019] [Indexed: 01/22/2023] Open
Abstract
Endothelial and mesenchymal stromal cells (ECs/MSCs) are crucial components of hematopoietic bone marrow stem cell niches. Both cell types appear to be required to support the maintenance and expansion of multipotent hematopoietic cells, i.e. hematopoietic stem cells (HSCs) and multipotent progenitors (MPPs). With the aim to exploit niche cell properties for experimental and potential clinical applications, we analyzed the potential of primary ECs alone and in combination with MSCs to support the ex vivo expansion/maintenance of human hematopoietic stem and progenitor cells (HSPCs). Even though a massive expansion of total CD34+ HSPCs was observed, none of the tested culture conditions supported the expansion or maintenance of multipotent HSPCs. Instead, mainly lympho-myeloid primed progenitors (LMPPs) were expanded. Similarly, following transplantation into immunocompromised mice the percentage of multipotent HSPCs within the engrafted HSPC population was significantly decreased compared to the original graft. Consistent with the in vitro findings, a bias towards lympho-myeloid lineage potentials was observed. In our conditions, neither classical co-cultures of HSPCs with primary ECs or MSCs, even in combination, nor the xenograft environment in immunocompromised mice efficiently support the expansion of multipotent HSPCs. Instead, enhanced expansion and a consistent bias towards lympho-myeloid committed LMPPs were observed.
Collapse
|
10
|
Gornostaeva AN, Andreeva ER, Bobyleva PI, Buravkova LB. Interaction of allogeneic adipose tissue-derived stromal cells and unstimulated immune cells in vitro: the impact of cell-to-cell contact and hypoxia in the local milieu. Cytotechnology 2018; 70:299-312. [PMID: 28975481 PMCID: PMC5809659 DOI: 10.1007/s10616-017-0144-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 09/07/2017] [Indexed: 01/22/2023] Open
Abstract
Multipotent mesenchymal stem cells (MSCs) are an attractive tool for cell therapy and regenerative medicine. Being applied in vivo, allogeneic MSCs are faced with both activated and unstimulated immune cells. The effects of MSCs on activated immune cells are well described and are mainly suppressive. Less is known about the interaction of MSCs with unstimulated immune cells. We evaluated the contribution of tissue-related O2 level ("physiological" hypoxia-5% O2) and cell-to-cell contact to the interaction between allogeneic adipose tissue-derived MSCs (ASCs) and unstimulated peripheral blood mononuclear cells (PBMCs). Under both O2 levels, ASCs affected the immune response by elevating the proportion of CD69+ T cells and modifying the functional activity of unstimulated PBMCs, providing a significant reduction of ROS level and activation of lysosome compartment. "Physiological" hypoxia partially attenuated the ASC modulation of PBMC function, reducing CD69+ cell activation and more significantly supressing ROS. In direct co-culture, the ASC effects were more pronounced. PBMC viability was preferentially maintained, and the lymphocyte subset ratio was altered in favour of B cells. Our findings demonstrate that allogeneic ASCs do not enhance the activation of unstimulated immune cells and can provide supportive functions. The "hypoxic" phenotype of ASCs may be more "desirable" for the interaction with allogeneic immune cells that may be required in cell therapy protocols.
Collapse
Affiliation(s)
- Aleksandra N Gornostaeva
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye shosse, 76a, Moscow, Russia, 123007
| | - Elena R Andreeva
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye shosse, 76a, Moscow, Russia, 123007.
| | - Polina I Bobyleva
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye shosse, 76a, Moscow, Russia, 123007
| | - Ludmila B Buravkova
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye shosse, 76a, Moscow, Russia, 123007
| |
Collapse
|
11
|
Lau SX, Leong YY, Ng WH, Ng AWP, Ismail IS, Yusoff NM, Ramasamy R, Tan JJ. Human mesenchymal stem cells promote CD34 + hematopoietic stem cell proliferation with preserved red blood cell differentiation capacity. Cell Biol Int 2017; 41:697-704. [PMID: 28403524 DOI: 10.1002/cbin.10774] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 03/08/2017] [Indexed: 11/11/2022]
Abstract
Studies showed that co-transplantation of mesenchymal stem cells (MSCs) and cord blood-derived CD34+ hematopoietic stem cells (HSCs) offered greater therapeutic effects but little is known regarding the effects of human Wharton's jelly derived MSCs on HSC expansion and red blood cell (RBC) generation in vitro. This study aimed to investigate the effects of MSCs on HSC expansion and differentiation. HSCs were co-cultured with MSCs or with 10% MSCs-derived conditioned medium, with HSCs cultured under standard medium served as a control. Cell expansion rates, number of mononuclear cell post-expansion and number of enucleated cells post-differentiation were evaluated. HSCs showed superior proliferation in the presence of MSC with mean expansion rate of 3.5 × 108 ± 1.8 × 107 after day 7 compared to the conditioned medium and the control group (8.9 × 107 ± 1.1 × 108 and 7.0 × 107 ± 3.3 × 106 respectively, P < 0.001). Although no significant differences in RBC differentiation were observed between groups at passage IV, the number of enucleated cell was greater compared to earlier passages, indicating successful RBC differentiation. Cord blood-derived CD34+ HSCs can be greatly expanded by co-culturing with MSCs without affecting the RBC differentiation capability, suggesting the importance of direct MSC-HSCs contact in HSC expansion and RBC differentiation.
Collapse
Affiliation(s)
- Show Xuan Lau
- Advanced Medical and Dental Institute, UniversitiSains Malaysia, Bertam 13200 Kepala Batas, Penang, Malaysia
| | - Yin Yee Leong
- Advanced Medical and Dental Institute, UniversitiSains Malaysia, Bertam 13200 Kepala Batas, Penang, Malaysia
| | - Wai Hoe Ng
- Advanced Medical and Dental Institute, UniversitiSains Malaysia, Bertam 13200 Kepala Batas, Penang, Malaysia
| | - Albert Wee Po Ng
- Advanced Medical and Dental Institute, UniversitiSains Malaysia, Bertam 13200 Kepala Batas, Penang, Malaysia
| | - Ida Shazrina Ismail
- Advanced Medical and Dental Institute, UniversitiSains Malaysia, Bertam 13200 Kepala Batas, Penang, Malaysia
| | - Narazah Mohd Yusoff
- Advanced Medical and Dental Institute, UniversitiSains Malaysia, Bertam 13200 Kepala Batas, Penang, Malaysia
| | - Rajesh Ramasamy
- Immunology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Selangor Darul Ehsan, Malaysia
| | - Jun Jie Tan
- Advanced Medical and Dental Institute, UniversitiSains Malaysia, Bertam 13200 Kepala Batas, Penang, Malaysia
| |
Collapse
|
12
|
Wharton’s Jelly Mesenchymal Stromal Cells as a Feeder Layer for the Ex Vivo Expansion of Hematopoietic Stem and Progenitor Cells: a Review. Stem Cell Rev Rep 2016; 13:35-49. [DOI: 10.1007/s12015-016-9702-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
13
|
Yamazoe H, Ichikawa T, Hagihara Y, Iwasaki Y. Generation of a patterned co-culture system composed of adherent cells and immobilized nonadherent cells. Acta Biomater 2016; 31:231-240. [PMID: 26685756 DOI: 10.1016/j.actbio.2015.12.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 11/25/2015] [Accepted: 12/10/2015] [Indexed: 01/31/2023]
Abstract
Patterned co-culture is a promising technique used for fundamental investigation of cell-cell communication and tissue engineering approaches. However, conventional methods are inapplicable to nonadherent cells. In this study, we aimed to establish a patterned co-culture system composed of adherent and nonadherent cells. Nonadherent cells were immobilized on a substrate using a cell membrane anchoring reagent conjugated to a protein, in order to incorporate them into the co-culture system. Cross-linked albumin film, which has unique surface properties capable of regulating protein adsorption, was used to control their spatial localization. The utility of our approach was demonstrated through the fabrication of a patterned co-culture consisting of micropatterned neuroblastoma cells surrounded by immobilized myeloid cells. Furthermore, we also created a co-culture system composed of cancer cells and immobilized monocytes. We observed that monocytes enhanced the drug sensitivity of cancer cells and its influence was limited to cancer cells located near the monocytes. Therefore, the incorporation of nonadherent cells into a patterned co-culture system is useful for creating culture systems containing immune cells, as well as investigating the influence of these immune cells on cancer drug sensitivity. STATEMENT OF SIGNIFICANCE Various methods have been proposed for creating patterned co-culture systems, in which multiple cell types are attached to a substrate with a desired pattern. However, conventional methods, including our previous report published in Acta Biomaterialia (2010, 6, 526-533), are unsuitable for nonadherent cells. Here, we developed a novel method that incorporates nonadherent cells into the co-culture system, which allows us to precisely manipulate and study microenvironments containing nonadherent and adherent cells. Using this technique, we demonstrated that monocytes (nonadherent cells) could enhance the drug sensitivity of cancer cells and that their influence had a limited effective range. Thus, our technique is useful for recreating complex tissues in order to investigate cellular interactions involving nonadherent cells.
Collapse
Affiliation(s)
- Hironori Yamazoe
- National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka 563-8577, Japan.
| | - Takashi Ichikawa
- National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka 563-8577, Japan; Department of Chemistry and Materials Engineering, Faculty of Chemistry, Materials, and Bioengineering, Kansai University, 3-3-35 Yamate-cho, Suita-shi, Osaka 564-8680, Japan
| | - Yoshihisa Hagihara
- National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka 563-8577, Japan
| | - Yasuhiko Iwasaki
- Department of Chemistry and Materials Engineering, Faculty of Chemistry, Materials, and Bioengineering, Kansai University, 3-3-35 Yamate-cho, Suita-shi, Osaka 564-8680, Japan
| |
Collapse
|
14
|
van der Garde M, van Pel M, Millán Rivero JE, de Graaf-Dijkstra A, Slot MC, Kleinveld Y, Watt SM, Roelofs H, Zwaginga JJ. Direct Comparison of Wharton's Jelly and Bone Marrow-Derived Mesenchymal Stromal Cells to Enhance Engraftment of Cord Blood CD34(+) Transplants. Stem Cells Dev 2015; 24:2649-59. [PMID: 26414086 DOI: 10.1089/scd.2015.0138] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cotransplantation of CD34(+) hematopoietic stem and progenitor cells (HSPCs) with mesenchymal stromal cells (MSCs) enhances HSPC engraftment. For these applications, MSCs are mostly obtained from bone marrow (BM). However, MSCs can also be isolated from the Wharton's jelly (WJ) of the human umbilical cord. This source, regarded to be a waste product, enables a relatively low-cost MSC acquisition without any burden to the donor. In this study, we evaluated the ability of WJ MSCs to enhance HSPC engraftment. First, we compared cultured human WJ MSCs with human BM-derived MSCs (BM MSCs) for in vitro marker expression, immunomodulatory capacity, and differentiation into three mesenchymal lineages. Although we confirmed that WJ MSCs have a more restricted differentiation capacity, both WJ MSCs and BM MSCs expressed similar levels of surface markers and exhibited similar immune inhibitory capacities. Most importantly, cotransplantation of either WJ MSCs or BM MSCs with CB CD34(+) cells into NOD SCID mice showed similar enhanced recovery of human platelets and CD45(+) cells in the peripheral blood and a 3-fold higher engraftment in the BM, blood, and spleen 6 weeks after transplantation when compared to transplantation of CD34(+) cells alone. Upon coincubation, both MSC sources increased the expression of adhesion molecules on CD34(+) cells, although stromal cell-derived factor-1 (SDF-1)-induced migration of CD34(+) cells remained unaltered. Interestingly, there was an increase in CFU-GEMM when CB CD34(+) cells were cultured on monolayers of WJ MSCs in the presence of exogenous thrombopoietin, and an increase in BFU-E when BM MSCs replaced WJ MSCs in such cultures. Our results suggest that WJ MSC is likely to be a practical alternative for BM MSC to enhance CB CD34(+) cell engraftment.
Collapse
Affiliation(s)
- Mark van der Garde
- 1 Jon J van Rood Center for Clinical Transfusion Research , Sanquin Blood Supply Foundation, Leiden, the Netherlands .,2 Department of Immunohematology and Blood Transfusion, Leiden University Medical Center , Leiden, the Netherlands .,3 Stem Cell Research, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford , and NHS Blood and Transplant Oxford, Oxford, United Kingdom
| | - Melissa van Pel
- 2 Department of Immunohematology and Blood Transfusion, Leiden University Medical Center , Leiden, the Netherlands
| | - Jose Eduardo Millán Rivero
- 1 Jon J van Rood Center for Clinical Transfusion Research , Sanquin Blood Supply Foundation, Leiden, the Netherlands .,2 Department of Immunohematology and Blood Transfusion, Leiden University Medical Center , Leiden, the Netherlands
| | - Alice de Graaf-Dijkstra
- 1 Jon J van Rood Center for Clinical Transfusion Research , Sanquin Blood Supply Foundation, Leiden, the Netherlands
| | - Manon C Slot
- 1 Jon J van Rood Center for Clinical Transfusion Research , Sanquin Blood Supply Foundation, Leiden, the Netherlands
| | - Yoshiko Kleinveld
- 1 Jon J van Rood Center for Clinical Transfusion Research , Sanquin Blood Supply Foundation, Leiden, the Netherlands
| | - Suzanne M Watt
- 3 Stem Cell Research, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford , and NHS Blood and Transplant Oxford, Oxford, United Kingdom
| | - Helene Roelofs
- 2 Department of Immunohematology and Blood Transfusion, Leiden University Medical Center , Leiden, the Netherlands
| | - Jaap Jan Zwaginga
- 1 Jon J van Rood Center for Clinical Transfusion Research , Sanquin Blood Supply Foundation, Leiden, the Netherlands .,2 Department of Immunohematology and Blood Transfusion, Leiden University Medical Center , Leiden, the Netherlands
| |
Collapse
|
15
|
Andreeva ER, Andrianova IV, Sotnezova EV, Buravkov SV, Bobyleva PI, Romanov YA, Buravkova LB. Human adipose-tissue derived stromal cells in combination with hypoxia effectively support ex vivo expansion of cord blood haematopoietic progenitors. PLoS One 2015; 10:e0124939. [PMID: 25919031 PMCID: PMC4412539 DOI: 10.1371/journal.pone.0124939] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 03/20/2015] [Indexed: 12/12/2022] Open
Abstract
The optimisation of haematopoietic stem and progenitor cell expansion is on demand in modern cell therapy. In this work, haematopoietic stem/progenitor cells (HSPCs) have been selected from unmanipulated cord blood mononuclear cells (cbMNCs) due to adhesion to human adipose-tissue derived stromal cells (ASCs) under standard (20%) and tissue-related (5%) oxygen. ASCs efficiently maintained viability and supported further HSPC expansion at 20% and 5% O2. During co-culture with ASCs, a new floating population of differently committed HSPCs (HSPCs-1) grew. This suspension was enriched with СD34+ cells up to 6 (20% O2) and 8 (5% O2) times. Functional analysis of HSPCs-1 revealed cobble-stone area forming cells (CAFCs) and lineage-restricted colony-forming cells (CFCs). The number of CFCs was 1.6 times higher at tissue-related O2, than in standard cultivation (20% O2). This increase was related to a rise in the number of multipotent precursors - BFU-E, CFU-GEMM and CFU-GM. These changes were at least partly ensured by the increased concentration of MCP-1 and IL-8 at 5% O2. In summary, our data demonstrated that human ASCs enables the selection of functionally active HSPCs from unfractionated cbMNCs, the further expansion of which without exogenous cytokines provides enrichment with CD34+ cells. ASCs efficiently support the viability and proliferation of cord blood haematopoietic progenitors of different commitment at standard and tissue-related O2 levels at the expense of direct and paracrine cell-to-cell interactions.
Collapse
Affiliation(s)
- Elena R. Andreeva
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
- * E-mail:
| | - Irina V. Andrianova
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
- Institute of Experimental Cardiology, Cardiology Research Center, Moscow, Russia
| | - Elena V. Sotnezova
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | | | - Polina I. Bobyleva
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Yury A. Romanov
- Institute of Experimental Cardiology, Cardiology Research Center, Moscow, Russia
| | | |
Collapse
|
16
|
Jadalannagari S, Aljitawi OS. Ectodermal Differentiation of Wharton's Jelly Mesenchymal Stem Cells for Tissue Engineering and Regenerative Medicine Applications. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:314-22. [PMID: 25517045 DOI: 10.1089/ten.teb.2014.0404] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSCs) from Wharton's jelly (WJ) of the human umbilical cord are perinatal stem cells that have self-renewal ability, extended proliferation potential, immunosuppressive properties, and are accordingly excellent candidates for tissue engineering. These MSCs are unique, easily accessible, and a noncontroversial cell source of regeneration in medicine. Wharton's jelly mesenchymal stem cells (WJMSCs) are multipotent and capable of multilineage differentiation into cells like adipocytes, bone, cartilage, and skeletal muscle upon exposure to appropriate conditions. The ectoderm is one of the three primary germ layers found in the very early embryo that differentiates into the epidermis, nervous system (spine, peripheral nerves, brain), and exocrine glands (mammary, sweat, salivary, and lacrimal glands). Accumulating evidence shows that MSCs obtained from WJ have an ectodermal differentiation potential. The current review examines this differentiation potential of WJMSC into the hair follicle, skin, neurons, and sweat glands along with discussing the potential utilization of such differentiation in regenerative medicine.
Collapse
Affiliation(s)
| | - Omar S Aljitawi
- 1Department of Bioengineering, University of Kansas, Lawrence, Kansas.,2Department of Hematology/Oncology, Blood and Marrow Transplant Program, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
17
|
The assessment of the in vivo to in vitro cellular transition of human umbilical cord multipotent stromal cells. Placenta 2014; 36:232-9. [PMID: 25524058 DOI: 10.1016/j.placenta.2014.11.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Revised: 11/26/2014] [Accepted: 11/28/2014] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Human umbilical cord stroma is a rich source of primitive multipotent stromal cells (hUC-MSCs). However, the methods for hUC-MSC isolation and propagation remain controversial and vary among laboratories. Our group previously demonstrated that two cell types emerge upon enzymatic isolation of hUC-MSCs, which subsequently undergo a transition towards a fibroblastoid phenotype in later passages. The aim of this study was to further analyse cultured hUC-MSCs by evaluating the cytoskeletal and cell adhesion proteins and by comparing the remodelling of those proteins in umbilical cord sections to determine the cell alterations due to enzymatic and explant methods. METHODS Tissue sections and cultured cells isolated by enzymatic or explant methods were analysed morphologically and by labelling cytokeratin, vimentin, alpha-smooth muscle actin, E-cadherin and N-cadherin profiles. RESULTS The present observations confirmed that wide, flat cells (type-1) share myofibroblastic features, appear exclusively in enzymatically isolated early cultures; gradually diminish or are replaced by fibroblastoid cells (type-2) in later passages. In contrast, the explant method does not result in the existence of type-1 cells in vitro. Among the tested CK subtypes, CK18 expression is upregulated, whereas CK19 expression is downregulated upon culturing after both protocols. Vimentin and α-SMA, as the major intermediate filaments of hUC-MSCs were found unaltered throughout the culturing period regardless of the cell isolation technique used. DISCUSSION The data presented confirm and further elucidate the previously observed phenotypic change in hUC-MSCs as illustrated by alterations in structural proteins during enzymatic isolation and subsequent culturing of cells compared with in situ equivalents.
Collapse
|
18
|
Mesenchymal stromal cells derived from human umbilical cord tissues: primitive cells with potential for clinical and tissue engineering applications. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2014; 123:29-54. [PMID: 20012739 DOI: 10.1007/10_2009_15] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem or stromal cells (MSCs) have a high potential for cell-based therapies as well as for tissue engineering applications. Since Friedenstein first isolated stem or precursor cells from the human bone marrow (BM) stroma that were capable of osteogenesis, BM is currently the most common source for MSCs. However, BM presents several disadvantages, namely low frequency of MSCs, high donor-dependent variations in quality, and painful invasive intervention. Thus, tremendous research efforts have been observed during recent years to find alternative sources for MSCs.In this context, the human umbilical cord (UC) has gained more and more attention. Since the UC is discarded after birth, the cells are easily accessible without ethical concerns. This postnatal organ was found to be rich in primitive stromal cells showing typical characteristics of bone-marrow MSCs (BMSCs), e.g., they grow as plastic-adherent cells with a fibroblastic morphology, express a set of typical surface markers, and can be directly differentiated at least along mesodermal lineages. Compared to BM, the UC tissue bears a higher frequency of stromal cells with a higher in vitro expansion potential. Furthermore, immune-privileged and immune-modulatory properties are reported for UC-derived cells, which open highly interesting perspectives for clinical applications.
Collapse
|
19
|
Bal G, Kamhieh-Milz J, Sterzer V, Al-Samman M, Debski J, Klein O, Kamhieh-Milz S, Bhakdi S, Salama A. Proteomic Profiling of Secreted Proteins for the Hematopoietic Support of Interleukin-Stimulated Human Umbilical Vein Endothelial Cells. Cell Transplant 2013; 22:1185-99. [DOI: 10.3727/096368912x657288] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Human umbilical cord vein endothelial cells (HUVECs) secrete a number of factors that greatly impact the proliferation and differentiation of hematopoietic stem and progenitor cells (HSPCs). These factors remain largely unknown. Here, we report on the most comprehensive proteomic profiling of the HUVEC secretome and identified 827 different secreted proteins. Two hundred and thirty-one proteins were found in all conditions, whereas 369 proteins were identified only under proinflammatory conditions following IL-1β, IL-3, and IL-6 stimulation. Thirteen proteins including complement factor b (CFb) were identified only under IL-1β and IL-3 conditions and may potentially represent HSPC proliferation factors. The combination of bioinformatics and gene ontology annotations indicates the role of the complement system and its activation. Furthermore, CFb was found to be transcriptionally strongly upregulated. Addition of complement component 5b-9 (C5b-9) monoclonal antibody to the stem cell expansion assay was capable of significantly reducing their proliferation. This study suggests a complement-mediated cross-talk between endothelial cells and HSPCs under proinflammatory conditions.
Collapse
Affiliation(s)
- Gürkan Bal
- Institute for Transfusion Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Julian Kamhieh-Milz
- Institute for Transfusion Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Viktor Sterzer
- Institute for Transfusion Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Muhammad Al-Samman
- Institute for Transfusion Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Janusz Debski
- Institute of Biochemistry and Biophysics, Polish Academy of Science, Warsaw, Poland
| | - Oliver Klein
- Berlin-Brandenburg Center for Regenerative Therapies, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Sundrela Kamhieh-Milz
- Institute for Transfusion Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Sucharit Bhakdi
- Institute of Medical Microbiology and Hygiene, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Abdulgabar Salama
- Institute for Transfusion Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
20
|
Comparative Evaluation of Human Mesenchymal Stem Cells of Fetal (Wharton's Jelly) and Adult (Adipose Tissue) Origin during Prolonged In Vitro Expansion: Considerations for Cytotherapy. Stem Cells Int 2013; 2013:246134. [PMID: 23533440 PMCID: PMC3603673 DOI: 10.1155/2013/246134] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 11/28/2012] [Accepted: 11/28/2012] [Indexed: 01/20/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are somatic cells with a dual capacity for self-renewal and differentiation, and diverse therapeutic applicability, both experimentally and in the clinic. These cells can be isolated from various human tissues that may differ anatomically or developmentally with relative ease. Heterogeneity due to biological origin or in vitro manipulation is, nevertheless, considerable and may equate to differences in qualitative and quantitative characteristics which can prove crucial for successful therapeutic use. With this in mind, in the present study we have evaluated the proliferation kinetics and phenotypic characteristics of MSCs derived from two abundant sources, that is, fetal umbilical cord matrix (Wharton's jelly) and adult adipose tissue (termed WJSC and ADSC, resp.) during prolonged in vitro expansion, a process necessary for obtaining cell numbers sufficient for clinical application. Our results show that WJSC are derived with relatively high efficiency and bear a substantially increased proliferation capacity whilst largely sustaining the expression of typical immunophenotypic markers, whereas ADSC exhibit a reduced proliferation potential showing typical signs of senescence at an early stage. By combining kinetic with phenotypic data we identify culture thresholds up to which both cell types maintain their stem properties, and we discuss the practical implications of their differences.
Collapse
|
21
|
The Therapeutic Potential, Challenges and Future Clinical Directions of Stem Cells from the Wharton’s Jelly of the Human Umbilical Cord. Stem Cell Rev Rep 2012; 9:226-40. [DOI: 10.1007/s12015-012-9418-z] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
22
|
Mesenchymal stem cells promote a primitive phenotype CD34+c-kit+ in human cord blood-derived hematopoietic stem cells during ex vivo expansion. Cell Mol Biol Lett 2012; 18:11-33. [PMID: 23104253 PMCID: PMC6275752 DOI: 10.2478/s11658-012-0036-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 10/22/2012] [Indexed: 12/30/2022] Open
Abstract
The purpose of this study was to evaluate the influence of bone marrow-mesenchymal stem cells (BM-MSC) and exogenously added cytokines on the proliferation, primitive cell subpopulation maintenance (including the c-kit+ marker) and clonogenic capacity of hematopoietic stem cells (HSC). BM-MSC were collected from volunteer donors, isolated and characterized. Umbilical cord blood (UCB) samples were collected from healthy full-term deliveries. UCB-CD34+ cells were cultured in the presence or absence of BM-MSC and/or cytokines for 3 and 7 days. CD34+ cell proliferation was evaluated using the CSFE method and cell phenotype was determined by CD34, c-kit, CD33, CD38, HLA-DR, cyCD22 and cyCD3 detection. Cell clonogenic ability was also assessed. Exogenously added SCF, TPO and FLT3L increased CD34+ cell proliferation in the presence or absence of BM-MSC, but with concomitant cell differentiation. Without any added cytokines, BM-MSC are able to increase the percentage of primitive progenitors as evaluated by c-kit expression and CFU-GEMM increase. Interestingly, this latter effect was dependent on both cell-cell interactions and secreted factors. A 7-day co-culture period will be optimal for obtaining an increased primitive HSC level. Including c-kit as a marker for primitive phenotype evaluation has shown the relevance of BM-MSC and their secreted factors on UCB-HSC stemness function. This effect could be dissociated from that of the addition of exogenous cytokines, which induced cellular differentiation instead.
Collapse
|
23
|
Hammoud M, Vlaski M, Duchez P, Chevaleyre J, Lafarge X, Boiron JM, Praloran V, Brunet De La Grange P, Ivanovic Z. Combination of low O(2) concentration and mesenchymal stromal cells during culture of cord blood CD34(+) cells improves the maintenance and proliferative capacity of hematopoietic stem cells. J Cell Physiol 2012; 227:2750-8. [PMID: 21913190 DOI: 10.1002/jcp.23019] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The physiological approach suggests that an environment associating the mesenchymal stromal cells (MSC) and low O(2) concentration would be most favorable for the maintenance of hematopoietic stem cells (HSCs) in course of ex vivo expansion of hematopoietic grafts. To test this hypothesis, we performed a co-culture of cord blood CD34(+) cells with or without MSC in presence of cytokines for 10 days at 20%, 5%, and 1.5% O(2) and assessed the impact on total cells, CD34(+) cells, committed progenitors (colony-forming cells-CFC) and stem cells activity (pre-CFC and Scid repopulating cells-SRC). Not surprisingly, the expansion of total cells, CD34(+) cells, and CFC was higher in co-culture and at 20% O(2) compared to simple culture and low O(2) concentrations, respectively. However, co-culture at low O(2) concentrations provided CD34(+) cell and CFC amplification similar to classical culture at 20% O(2) . Interestingly, low O(2) concentrations ensured a better pre-CFC and SRC preservation/expansion in co-culture. Indeed, SRC activity in co-culture at 1.5% O(2) was higher than in freshly isolated CD34(+) cells. Interleukin-6 production by MSC at physiologically low O(2) concentrations might be one of the factors mediating this effect. Our data demonstrate that association of co-culture and low O(2) concentration not only induces sufficient expansion of committed progenitors (with respect to the classical culture), but also ensures a better maintenance/expansion of hematopoietic stem cells (HSCs), pointing to the oxygenation as a physiological regulatory factor but also as a cell engineering tool.
Collapse
Affiliation(s)
- Mohammad Hammoud
- Aquitaine-Limousin Branch of French Blood Institute (Etablissement Français du Sang, Aquitaine-Limousin, EFS-AL), Bordeaux, France
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Mitterberger MC, Lechner S, Mattesich M, Kaiser A, Probst D, Wenger N, Pierer G, Zwerschke W. DLK1(PREF1) is a negative regulator of adipogenesis in CD105⁺/CD90⁺/CD34⁺/CD31⁻/FABP4⁻ adipose-derived stromal cells from subcutaneous abdominal fat pats of adult women. Stem Cell Res 2012; 9:35-48. [PMID: 22640926 DOI: 10.1016/j.scr.2012.04.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 04/04/2012] [Accepted: 04/06/2012] [Indexed: 12/20/2022] Open
Abstract
The main physiological function of adipose-derived stromal/progenitor cells (ASC) is to differentiate into adipocytes. ASC are most likely localized at perivascular sites in adipose tissues and retain the capacity to differentiate into multiple cell types. Although cell surface markers for ASC have been described, there is no complete consensus on the antigen expression pattern that will precisely define these cells. DLK1(PREF1) is an established marker for mouse adipocyte progenitors which inhibits adipogenesis. This suggests that DLK1(PREF1) could be a useful marker to characterize human ASC. The DLK1(PREF1) status of human ASC is however unknown. In the present study we isolated ASC from the heterogeneous stromal vascular fraction of subcutaneous abdominal fat pats of adult women. These cells were selected by their plastic adherence and expanded to passage 5. The ASC were characterized as relatively homogenous cell population with the capacity to differentiate in vitro into adipocytes, chondrocytes, and osteoblasts and the immunophenotype CD105⁺/CD90⁺/CD34⁺/CD31⁻/FABP4⁻. The ASC were positive for DLK1(PREF1) which was well expressed in proliferating and density arrested cells but downregulated in the course of adipogenic differentiation. To investigate whether DLK1(PREF1) plays a role in the regulation of adipogenesis in these cells RNAi-mediated knockdown experiments were conducted. Knockdown of DLK1(PREF1) in differentiating ASC resulted in a significant increase of the expression of the adipogenic key regulator PPARγ2 and of the terminal adipogenic differentiation marker FABP4. We conclude that DLK1(PREF1) is well expressed in human ASC and acts as a negative regulator of adipogenesis. Moreover, DLK1(PREF1) could be a functional marker contributing to the characterization of human ASC.
Collapse
Affiliation(s)
- Maria C Mitterberger
- Department of Cell Metabolism and Differentiation Research, Institute for Biomedical Aging Research of the Austrian Academy of Sciences, Rennweg 10, A-6020 Innsbruck, Austria
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Xia X, Ji T, Chen P, Li X, Fang Y, Gao Q, Liao S, You L, Xu H, Ma Q, Wu P, Hu W, Wu M, Cao L, Li K, Weng Y, Han Z, Wei J, Liu R, Wang S, Xu G, Wang D, Zhou J, Ma D. Mesenchymal stem cells as carriers and amplifiers in CRAd delivery to tumors. Mol Cancer 2011; 10:134. [PMID: 22054049 PMCID: PMC3215929 DOI: 10.1186/1476-4598-10-134] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 11/03/2011] [Indexed: 12/13/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) have been considered to be the attractive vehicles for delivering therapeutic agents toward various tumor diseases. This study was to explore the distribution pattern, kinetic delivery of adenovirus, and therapeutic efficacy of the MSC loading of E1A mutant conditionally replicative adenovirus Adv-Stat3(-) which selectively replicated and expressed high levels of anti-sense Stat3 complementary DNA in breast cancer and melanoma cells. Methods We assessed the release ability of conditionally replicative adenovirus (CRAd) from MSC using crystal violet staining, TCID50 assay, and quantitative PCR. In vitro killing competence of MSCs carrying Adv-Stat3(-) toward breast cancer and melanoma was performed using co-culture system of transwell plates. We examined tumor tropism of MSC by Prussian blue staining and immunofluorescence. In vivo killing competence of MSCs carrying Adv-Stat3(-) toward breast tumor was analyzed by comparison of tumor volumes and survival periods. Results Adv-Stat3(-) amplified in MSCs and were released 4 days after infection. MSCs carrying Adv-Stat3(-) caused viral amplification, depletion of Stat3 and its downstream proteins, and led to significant apoptosis in breast cancer and melanoma cell lines. In vivo experiments confirmed the preferential localization of MSCs in the tumor periphery 24 hours after tail vein injection, and this localization was mainly detected in the tumor parenchyma after 72 hours. Intravenous injection of MSCs carrying Adv-Stat3(-) suppressed the Stat3 pathway, down-regulated Ki67 expression, and recruited CD11b-positive cells in the local tumor, inhibiting tumor growth and increasing the survival of tumor-bearing mice. Conclusions These results indicate that MSCs migrate to the tumor site in a time-dependent manner and could be an effective platform for the targeted delivery of CRAd and the amplification of tumor killing effects.
Collapse
Affiliation(s)
- Xi Xia
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Arufe MC, Fuente ADL, Fuentes I, Toro FJD, Blanco FJ. Umbilical cord as a mesenchymal stem cell source for treating joint pathologies. World J Orthop 2011; 2:43-50. [PMID: 22474635 PMCID: PMC3302041 DOI: 10.5312/wjo.v2.i6.43] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Revised: 06/01/2011] [Accepted: 06/08/2011] [Indexed: 02/06/2023] Open
Abstract
Articular cartilage disorders and injuries often result in life-long chronic pain and compromised quality of life. Regrettably, the regeneration of articular cartilage is a continuing challenge for biomedical research. One of the most promising therapeutic approaches is cell-based tissue engineering, which provides a healthy population of cells to the injured site but requires differentiated chondrocytes from an uninjured site. The use of healthy chondrocytes has been found to have limitations. A promising alternative cell population is mesenchymal stem cells (MSCs), known to possess excellent proliferation potential and proven capability for differentiation into chondrocytes. The “immunosuppressive” property of human MSCs makes them an important candidate for allogeneic cell therapy. The use of allogeneic MSCs to repair large defects may prove to be an alternative to current autologous and allogeneic tissue-grafting procedures. An allogeneic cell-based approach would enable MSCs to be isolated from any donor, expanded and cryopreserved in allogeneic MSC banks, providing a readily available source of progenitors for cell replacement therapy. These possibilities have spawned the current exponential growth in stem cell research in pharmaceutical and biotechnology communities. Our objective in this review is to summarize the knowledge about MSCs from umbilical cord stroma and focus mainly on their applications for joint pathologies.
Collapse
|
27
|
Celebi B, Mantovani D, Pineault N. Irradiated Mesenchymal Stem Cells improve the ex vivo expansion of Hematopoietic Progenitors by partly mimicking the bone marrow endosteal environment. J Immunol Methods 2011; 370:93-103. [PMID: 21699899 DOI: 10.1016/j.jim.2011.06.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 04/19/2011] [Accepted: 06/03/2011] [Indexed: 12/12/2022]
Abstract
Mesenchymal Stem Cells (MSCs) regulate the growth and differentiation of Hematopoietic Progenitor cells (HPCs) through the release of soluble factors or through their differentiation into osteoblasts. We recently demonstrated that expansion of megakaryocyte (MK) progenitors ex vivo had reached a plateau when CD34(+) cells were grown with two optimized cytokine cocktails developed for the growth of MK. Hence, we sought to determine whether co-culture of CD34(+) cells with Bone Marrow (BM) MSCs could further increase the expansion of myeloid and MK progenitors. First, we tested the impact of cell-cell contact and pre-irradiation treatment of the MSCs to identify the condition that best supports HPC expansion. This screen revealed that HPC expansions were generally greater in the non-contact conditions, and that pre-irradiation of the MSCs appeared to be of added benefits. Improved expansion of both myeloid and MK progenitors in co-culture with irradiated MSCs without contact was subsequently confirmed. Next, cytokine array profiling was carried out to investigate why irradiation promoted progenitor expansion. This revealed that the levels of as many as 33 factors were potentially altered. ELISA confirmed the significant up regulation of NT-3 and IGFBP-2. Since, these factors are known to be released by and important for osteogenic and endothelial cells, we investigated and confirmed that irradiation of MSCs induced their rapid differentiation into osteogenic-like cells, but not into endothelial-like cells. Supporting this finding, expansions of myeloid and MK progenitors were increased when CD34(+) cells were co-culture with MSCs-derived osteoblasts. Altogether, these results indicate that the improved expansion of HPCs obtained with irradiated MSCs is due in part to their differentiation into osteoblast-like cells, thereby recreating an endosteal-like environment that provides improved support for HPCs expansion.
Collapse
Affiliation(s)
- Betül Celebi
- Hema-Quebec, Research & Development Department, Quebec City, PQ, Canada, G1V 5C3
| | | | | |
Collapse
|
28
|
De Bruyn C, Najar M, Raicevic G, Meuleman N, Pieters K, Stamatopoulos B, Delforge A, Bron D, Lagneaux L. A rapid, simple, and reproducible method for the isolation of mesenchymal stromal cells from Wharton's jelly without enzymatic treatment. Stem Cells Dev 2010; 20:547-57. [PMID: 20923277 DOI: 10.1089/scd.2010.0260] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The co-infusion of mesenchymal stromal cells (MSCs) with hematopoietic stem cells could improve the hematopoietic engraftment after cord blood transplant. Adult bone marrow is the major source of MSCs for cell therapy. However, bone marrow aspiration involves an invasive procedure and, in the case of a cord blood transplant, requires the use of a third party. The umbilical cord matrix, called Wharton's jelly (WJ), was previously shown to be a valuable source of MSCs. However, the process of cell separation is not standardized and needs to be optimized. In this study, we focused on the efficiency of the isolation procedure and expansion of cells from WJ MSCs isolated from human full-term umbilical cords. MSCs were isolated from the WJ without enzyme digestion or dissection. The procedure was based only on the plastic adhesion capacities of MSCs. Briefly, umbilical cord segments of 5-10 cm were cut longitudinally and plated with the WJ onto a plastic surface for 5 days in an appropriate culture medium. After removing the cord segment, the culture was pursued until subconfluency. The number of cells and their phenotypes, clonogenic capacities, differentiation capacities, immunomodulation, and hematopoietic supportive functions were evaluated. Using this method, we were able to isolate MSCs from all human umbilical cords analyzed (n = 50). We obtained a mean of 1.4 × 10(8) cells at the second passage and >7 × 10(9) cells at the third. The expanded cells expressed characteristic markers and presented typical functional properties of MSCs such as differentiation capacities, immunologic properties, and hematopoietic supportive functions. In conclusion, we have established a simple, rapid, and reproducible protocol to isolate abundant MSCs from short segments of umbilical cords.
Collapse
Affiliation(s)
- Cécile De Bruyn
- Service d'Hématologie Expérimentale-Laboratoire de Thérapie Cellulaire Clinique, Université Libre de Bruxelles-Institut Jules Bordet, Bruxelles, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Gómez-Aristizábal A, Keating A, Davies JE. Mesenchymal stromal cells as supportive cells for hepatocytes. Mol Ther 2009; 17:1504-8. [PMID: 19584815 PMCID: PMC2835270 DOI: 10.1038/mt.2009.158] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Accepted: 06/16/2009] [Indexed: 02/07/2023] Open
Abstract
Hepatocytes and hematopoietic stem cells (HSCs) appear to share many of the same requirements for their survival, functionality, and proliferation. This may be due to a shared location during fetal development. Moreover, hepatocytes and HSCs are unable to function, or even survive, without stromal cell support. Bone marrow-derived mesenchymal stromal cells (MSCs) support the proliferation and functionality, not only of HSCs, but also of hepatocytes. Although knowledge of the mechanisms underlying HSCs' support is far more advanced than for hepatocytes, data suggest that many agents important for HSCs also maintain the normal hepatocyte phenotype in vitro. Thus, it is possible that new techniques for the maintenance and expansion of HSCs may also be useful for hepatocytes. Bone marrow-derived MSCs are easily cultured and expanded in vitro, and some data suggest that they are immunoregulatory as well as relatively nonimmunogenic. These observations suggest that allogeneic MSCs may be useful not only in supporting hepatocyte growth and proliferation but also in modulating immune responses such as stellate cell activation.
Collapse
|
30
|
Bonanno G, Mariotti A, Procoli A, Corallo M, Scambia G, Pierelli L, Rutella S. Interleukin-21 induces the differentiation of human umbilical cord blood CD34-lineage- cells into pseudomature lytic NK cells. BMC Immunol 2009; 10:46. [PMID: 19712464 PMCID: PMC2743656 DOI: 10.1186/1471-2172-10-46] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2009] [Accepted: 08/27/2009] [Indexed: 12/28/2022] Open
Abstract
Background Umbilical cord blood (UCB) is enriched with transplantable CD34+ cells. In addition to CD34-expressing haematopoietic stem cells (HSC), human UCB contains a rare population of CD34-lineage- cells endowed with the ability to differentiate along the T/NK pathway in response to interleukin (IL)-15 and a stromal cell support. IL-21 is a crucial regulator of NK cell function, whose influence on IL-15-induced differentiation of CD34-lineage- cells has not been investigated previously. The present study was designed and conducted to address whether IL-21 might replace the stromal cell requirements and foster the IL-15-induced NK differentiation of human UCB CD34-lineage- cells. Results CD34-lineage- cells were maintained in liquid culture with Flt3-L and SCF, with the addition of IL-15 and IL-21, either alone or in combination. Cultures were established in the absence of feeder cells or serum supplementation. Cytokine-treated cells were used to evaluate cell surface phenotype, expression of molecular determinants of lymphoid/NK cell differentiation, secretion of IFN-γ, GM-CSF, TNF-α and CCL3/MIP-1α, and cytolytic activity against NK-sensitive tumour cell targets. CD34-lineage- cells proliferated vigorously in response to IL-15 and IL-21 but not to IL-21 alone, and up-regulated phosphorylated Stat1 and Stat3 proteins. CD34-lineage- cells expanded by IL-21 in combination with IL-15 acquired lymphoid morphology and killer-cell immunoglobulin-like receptor (KIR)-CD56+CD16-/+ phenotype, consistent with pseudo-mature NK cells. IL-21/IL-15-differentiated cells expressed high levels of mRNA for Bcl-2, GATA-3 and Id2, a master switch required for NK-cell development, and harboured un-rearranged TCRγ genes. From a functional standpoint, IL-21/IL-15-treated cells secreted copious amounts of IFN-γ, GM-CSF and CCL3/MIP-1α, and expressed cell surface CD107a upon contact with NK-sensitive tumour targets, a measure of exocytosis of NK secretory granules. Conclusion This study underpins a novel role for IL-21 in the differentiation of pseudo-mature lytic NK cells in a synergistic context with IL-15, and identifies a potential strategy to expand functional NK cells for immunotherapy.
Collapse
Affiliation(s)
- Giuseppina Bonanno
- Department of Gynaecology, Catholic University Medical School, Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|
31
|
Walenda T, Bork S, Horn P, Wein F, Saffrich R, Diehlmann A, Eckstein V, Ho AD, Wagner W. Co-culture with mesenchymal stromal cells increases proliferation and maintenance of haematopoietic progenitor cells. J Cell Mol Med 2009; 14:337-50. [PMID: 19432817 PMCID: PMC3837622 DOI: 10.1111/j.1582-4934.2009.00776.x] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stromal cells (MSC) have been suggested to provide a suitable cellular environment for in vitro expansion of haematopoietic stem and progenitor cells (HPC) from umbilical cord blood. In this study, we have simultaneously analysed the cell division history and immunophenotypic differentiation of HPC by using cell division tracking with carboxyfluorescein diacetate N-succinimidyl ester (CFSE). Co-culture with MSC greatly enhanced proliferation of human HPC, especially of the more primitive CD34(+)CD38(-) fraction. Without co-culture CD34 and CD133 expressions decreased after several cell divisions, whereas CD38 expression was up-regulated after some cell divisions and then diminished in fast proliferating cells. Co-culture with MSC maintained a primitive immunophenotype (CD34(+), CD133(+) and CD38(-)) for more population doublings, whereas up-regulation of differentiation markers (CD13, CD45 and CD56) in HPC was delayed to higher numbers of cell divisions. Especially MSC of early cell passages maintained CD34 expression in HPC over more cell divisions, whereas MSC of higher passages further enhanced their proliferation rate. Inhibition of mitogen-activated protein kinase 1 (MAPK1) impaired proliferation and differentiation of HPC, but not maintenance of long-term culture initiating cells. siRNA knockdown of N-cadherin and VCAM1 in feeder layer cells increased the fraction of slow dividing HPC, whereas knockdown of integrin beta 1 (ITGB1) and CD44 impaired their differentiation. In conclusion, MSC support proliferation as well as self-renewal of HPC with primitive immunophenotype. The use of early passages of MSC and genetic manipulation of proteins involved in HPC-MSC interaction might further enhance cord blood expansion on MSC.
Collapse
Affiliation(s)
- Thomas Walenda
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Flores-Guzmán P, Flores-Figueroa E, Montesinos JJ, Martínez-Jaramillo G, Fernández-Sánchez V, Valencia-Plata I, Alarcón-Santos G, Mayani H. Individual and combined effects of mesenchymal stromal cells and recombinant stimulatory cytokines on the in vitro growth of primitive hematopoietic cells from human umbilical cord blood. Cytotherapy 2009; 11:886-96. [DOI: 10.3109/14653240903180076] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|