1
|
Branco A, Rayabaram J, Miranda CC, Fernandes-Platzgummer A, Fernandes TG, Sajja S, da Silva CL, Vemuri MC. Advances in ex vivo expansion of hematopoietic stem and progenitor cells for clinical applications. Front Bioeng Biotechnol 2024; 12:1380950. [PMID: 38846805 PMCID: PMC11153805 DOI: 10.3389/fbioe.2024.1380950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/25/2024] [Indexed: 06/09/2024] Open
Abstract
As caretakers of the hematopoietic system, hematopoietic stem cells assure a lifelong supply of differentiated populations that are responsible for critical bodily functions, including oxygen transport, immunological protection and coagulation. Due to the far-reaching influence of the hematopoietic system, hematological disorders typically have a significant impact on the lives of individuals, even becoming fatal. Hematopoietic cell transplantation was the first effective therapeutic avenue to treat such hematological diseases. Since then, key use and manipulation of hematopoietic stem cells for treatments has been aspired to fully take advantage of such an important cell population. Limited knowledge on hematopoietic stem cell behavior has motivated in-depth research into their biology. Efforts were able to uncover their native environment and characteristics during development and adult stages. Several signaling pathways at a cellular level have been mapped, providing insight into their machinery. Important dynamics of hematopoietic stem cell maintenance were begun to be understood with improved comprehension of their metabolism and progressive aging. These advances have provided a solid platform for the development of innovative strategies for the manipulation of hematopoietic stem cells. Specifically, expansion of the hematopoietic stem cell pool has triggered immense interest, gaining momentum. A wide range of approaches have sprouted, leading to a variety of expansion systems, from simpler small molecule-based strategies to complex biomimetic scaffolds. The recent approval of Omisirge, the first expanded hematopoietic stem and progenitor cell product, whose expansion platform is one of the earliest, is predictive of further successes that might arise soon. In order to guarantee the quality of these ex vivo manipulated cells, robust assays that measure cell function or potency need to be developed. Whether targeting hematopoietic engraftment, immunological differentiation potential or malignancy clearance, hematopoietic stem cells and their derivatives need efficient scaling of their therapeutic potency. In this review, we comprehensively view hematopoietic stem cells as therapeutic assets, going from fundamental to translational.
Collapse
Affiliation(s)
- André Branco
- Department of Bioengineering and Institute for Bioengineering and Biosciences (iBB), Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Janakiram Rayabaram
- Protein and Cell Analysis, Biosciences Division, Invitrogen Bioservices, Thermo Fisher Scientific, Bangalore, India
| | - Cláudia C. Miranda
- Department of Bioengineering and Institute for Bioengineering and Biosciences (iBB), Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- AccelBio, Collaborative Laboratory to Foster Translation and Drug Discovery, Cantanhede, Portugal
| | - Ana Fernandes-Platzgummer
- Department of Bioengineering and Institute for Bioengineering and Biosciences (iBB), Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Tiago G. Fernandes
- Department of Bioengineering and Institute for Bioengineering and Biosciences (iBB), Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Suchitra Sajja
- Protein and Cell Analysis, Biosciences Division, Invitrogen Bioservices, Thermo Fisher Scientific, Bangalore, India
| | - Cláudia L. da Silva
- Department of Bioengineering and Institute for Bioengineering and Biosciences (iBB), Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | | |
Collapse
|
2
|
Glaser DE, Curtis MB, Sariano PA, Rollins ZA, Shergill BS, Anand A, Deely AM, Shirure VS, Anderson L, Lowen JM, Ng NR, Weilbaecher K, Link DC, George SC. Organ-on-a-chip model of vascularized human bone marrow niches. Biomaterials 2022; 280:121245. [PMID: 34810038 PMCID: PMC10658812 DOI: 10.1016/j.biomaterials.2021.121245] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 11/01/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022]
Abstract
Bone marrow niches (endosteal and perivascular) play important roles in both normal bone marrow function and pathological processes such as cancer cell dormancy. Unraveling the mechanisms underlying these events in humans has been severely limited by models that cannot dissect dynamic events at the niche level. Utilizing microfluidic and stem cell technologies, we present a 3D in vitro model of human bone marrow that contains both the perivascular and endosteal niches, complete with dynamic, perfusable vascular networks. We demonstrate that our model can replicate in vivo bone marrow function, including maintenance and differentiation of CD34+ hematopoietic stem/progenitor cells, egress of neutrophils (CD66b+), and niche-specific responses to doxorubicin and granulocyte-colony stimulating factor. Our platform provides opportunities to accelerate current understanding of human bone marrow function and drug response with high spatial and temporal resolution.
Collapse
Affiliation(s)
- Drew E Glaser
- Department of Biomedical Engineering, University of California, Davis, 451 E Health Sciences Dr, GBSF 2303, Davis, CA 95616, USA
| | - Matthew B Curtis
- Department of Biomedical Engineering, University of California, Davis, 451 E Health Sciences Dr, GBSF 2303, Davis, CA 95616, USA
| | - Peter A Sariano
- Department of Biomedical Engineering, University of California, Davis, 451 E Health Sciences Dr, GBSF 2303, Davis, CA 95616, USA
| | - Zachary A Rollins
- Department of Chemical Engineering, University of California, Davis, 1 Shields Ave, Bainer 3106, Davis, CA 95616, USA
| | - Bhupinder S Shergill
- Department of Biomedical Engineering, University of California, Davis, 451 E Health Sciences Dr, GBSF 2303, Davis, CA 95616, USA
| | - Aravind Anand
- Department of Biomedical Engineering, University of California, Davis, 451 E Health Sciences Dr, GBSF 2303, Davis, CA 95616, USA
| | - Alyssa M Deely
- Department of Biomedical Engineering, University of California, Davis, 451 E Health Sciences Dr, GBSF 2303, Davis, CA 95616, USA
| | - Venktesh S Shirure
- Department of Biomedical Engineering, University of California, Davis, 451 E Health Sciences Dr, GBSF 2303, Davis, CA 95616, USA
| | - Leif Anderson
- Department of Biomedical Engineering, University of California, Davis, 451 E Health Sciences Dr, GBSF 2303, Davis, CA 95616, USA
| | - Jeremy M Lowen
- Department of Biomedical Engineering, University of California, Davis, 451 E Health Sciences Dr, GBSF 2303, Davis, CA 95616, USA
| | - Natalie R Ng
- Department of Biomedical Engineering, Washington University in St. Louis, 1 Brookings Dr, Campus Box 1100, St Louis, MO 63130, USA
| | - Katherine Weilbaecher
- Department of Medicine, Washington University in St. Louis, 660 S Euclid Ave, Campus Box 8066, St. Louis, MO 63110, USA
| | - Daniel C Link
- Department of Medicine, Washington University in St. Louis, 660 S Euclid Ave, Campus Box 8066, St. Louis, MO 63110, USA
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, 451 E Health Sciences Dr, GBSF 2303, Davis, CA 95616, USA.
| |
Collapse
|
3
|
Orticelli V, Papait A, Vertua E, Bonassi Signoroni P, Romele P, Di Pietro L, Magatti M, Teofili L, Silini AR, Parolini O. Human amniotic mesenchymal stromal cells support the ex vivo expansion of cord blood hematopoietic stem cells. Stem Cells Transl Med 2021; 10:1516-1529. [PMID: 34327849 PMCID: PMC8550705 DOI: 10.1002/sctm.21-0130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/03/2021] [Accepted: 06/06/2021] [Indexed: 12/29/2022] Open
Abstract
Currently, more than 30 000 allogeneic hematopoietic stem cell (HSC) transplantations have been performed for the treatment of hematological and nonhematological diseases using HSC from umbilical cord blood (CB). However, the wide utilization of CB as a source of HSC is limited by the low number of cells recovered. One strategy to expand ex vivo CB‐HSC is represented by the use of bone marrow mesenchymal stromal cells (BM‐MSCs) as a feeder to enhance HSC proliferation while maintaining HSC stemness. Indeed, BM‐MSCs have been recognized as one of the most relevant players in the HSC niche. Thus, it has been hypothesized that they can support the ex vivo expansion of HSC by mimicking the physiological microenvironment present in the hematopoietic niche. Due to the role of placenta in supporting fetal hematopoiesis, MSC derived from the amniotic membrane (hAMSC) of human term placenta could represent an interesting alternative to BM‐MSC as a feeder layer to enhance the proliferation and maintain HSC stemness. Therefore, in this study we investigated if hAMSC could support the ex vivo expansion of HSC and progenitor cells. The capacity of hAMSCs to support the ex vivo expansion of CB‐HSC was evaluated in comparison to the control condition represented by the CB‐CD34+ cells without a feeder layer. The coculture was performed at two different CD34+:MSC ratios (1:2 and 1:8) in both cell‐to‐cell contact and transwell setting. After 7 days, the cells were collected and analyzed for phenotype and functionality. Our results suggest that hAMSCs represent a valuable alternative to BM‐MSC to support: (a) the ex vivo expansion of CB‐HSC in both contact and transwell systems, (b) the colony forming unit ability, and (c) long‐term culture initiating cells ability. Overall, these findings may contribute to address the unmet need of high HSC content in CB units available for transplantation.
Collapse
Affiliation(s)
- Valentina Orticelli
- Dipartimento di Scienze della vita e sanità pubblica, Università Cattolica del Sacro Cuore, Rome, Italy.,IRCCS Fondazione Policlinico Universitario "Agostino Gemelli", Rome, Italy
| | - Andrea Papait
- Dipartimento di Scienze della vita e sanità pubblica, Università Cattolica del Sacro Cuore, Rome, Italy.,Centro di Ricerca E. Menni, Fondazione Poliambulanza, Brescia, Italy
| | - Elsa Vertua
- Centro di Ricerca E. Menni, Fondazione Poliambulanza, Brescia, Italy
| | | | - Pietro Romele
- Centro di Ricerca E. Menni, Fondazione Poliambulanza, Brescia, Italy
| | - Lorena Di Pietro
- Dipartimento di Scienze della vita e sanità pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marta Magatti
- Centro di Ricerca E. Menni, Fondazione Poliambulanza, Brescia, Italy
| | - Luciana Teofili
- IRCCS Fondazione Policlinico Universitario "Agostino Gemelli", Rome, Italy
| | | | - Ornella Parolini
- Dipartimento di Scienze della vita e sanità pubblica, Università Cattolica del Sacro Cuore, Rome, Italy.,IRCCS Fondazione Policlinico Universitario "Agostino Gemelli", Rome, Italy
| |
Collapse
|
4
|
Bucar S, Branco ADDM, Mata MF, Milhano JC, Caramalho Í, Cabral JMS, Fernandes-Platzgummer A, da Silva CL. Influence of the mesenchymal stromal cell source on the hematopoietic supportive capacity of umbilical cord blood-derived CD34 +-enriched cells. Stem Cell Res Ther 2021; 12:399. [PMID: 34256848 PMCID: PMC8278708 DOI: 10.1186/s13287-021-02474-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/24/2021] [Indexed: 12/18/2022] Open
Abstract
Background Umbilical cord blood (UCB) is a clinically relevant alternative source of hematopoietic stem/progenitor cells (HSPC). To overcome the low cell number per UCB unit, ex vivo expansion of UCB HSPC in co-culture with mesenchymal stromal cells (MSC) has been established. Bone marrow (BM)-derived MSC have been the standard choice, but the use of MSC from alternative sources, less invasive and discardable, could ease clinical translation of an expanded CD34+ cell product. Here, we compare the capacity of BM-, umbilical cord matrix (UCM)-, and adipose tissue (AT)-derived MSC, expanded with/without xenogeneic components, to expand/maintain UCB CD34+-enriched cells ex vivo. Methods UCB CD34+-enriched cells were isolated from cryopreserved mononuclear cells and cultured for 7 days over an established feeder layer (FL) of BM-, UCM-, or AT-derived MSC, previously expanded using fetal bovine serum (FBS) or fibrinogen-depleted human platelet lysate (HPL) supplemented medium. UCB cells were cultured in serum-free medium supplemented with SCF/TPO/FLT3-L/bFGF. Fold increase in total nucleated cells (TNC) as well as immunophenotype and clonogenic potential (cobblestone area-forming cells and colony-forming unit assays) of the expanded hematopoietic cells were assessed. Results MSC from all sources effectively supported UCB HSPC expansion/maintenance ex vivo, with expansion factors (in TNC) superior to 50x, 70x, and 80x in UCM-, BM-, and AT-derived MSC co-cultures, respectively. Specifically, AT-derived MSC co-culture resulted in expanded cells with similar phenotypic profile compared to BM-derived MSC, but resulting in higher total cell numbers. Importantly, a subpopulation of more primitive cells (CD34+CD90+) was maintained in all co-cultures. In addition, the presence of a MSC FL was essential to maintain and expand a subpopulation of progenitor T cells (CD34+CD7+). The use of HPL to expand MSC prior to co-culture establishment did not influence the expansion potential of UCB cells. Conclusions AT represents a promising alternative to BM as a source of MSC for co-culture protocols to expand/maintain HSPC ex vivo. On the other hand, UCM-derived MSC demonstrated inferior hematopoietic supportive capacity compared to MSC from adult tissues. Despite HPL being considered an alternative to FBS for clinical-scale manufacturing of MSC, further studies are needed to determine its impact on the hematopoietic supportive capacity of these cells.
Collapse
Affiliation(s)
- Sara Bucar
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - André Dargen de Matos Branco
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Márcia F Mata
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - João Coutinho Milhano
- Hospital São Francisco Xavier, Centro Hospitalar de Lisboa Ocidental, Lisboa, Portugal
| | | | - Joaquim M S Cabral
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Ana Fernandes-Platzgummer
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal. .,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
5
|
Miyoshi H, Abo K, Hosoya D, Matsuo K, Utsumi Y. Effects of mouse fetal liver cell culture density on hematopoietic cell expansion in three-dimensional cocultures with stromal cells. Int J Artif Organs 2021; 45:103-112. [PMID: 33611956 DOI: 10.1177/0391398821996377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE An effective ex vivo expansion system of primitive hematopoietic cells (HCs) is required for wider application of hematopoietic stem cell transplantation. In this study, we examined effects of culture density on mouse fetal liver cells (FLCs) used as an HC source for the expansion of primitive HCs in three-dimensional (3D) cocultures with two kinds of mouse stromal cell lines (OP9 or C3H10T1/2). MATERIALS AND METHODS FLCs were seeded at different densities (1, 2, and 10 × 107 cells/cm3) into porous polymer scaffolds with or without stromal cell layers and HCs were expanded in the cultures for 2 weeks without exogenous cytokines. RESULTS Differential effects of culture density on HC expansion were observed between cocultures and solitary FLC controls. In stromal cell cocultures, high expansion of HCs was achieved when FLCs were seeded at low densities. In contrast, the expansion in the controls was enhanced with increasing culture densities. With respect to expansion of primitive HCs existing in the FLCs, cocultures with C3H10T1/2 cells were superior to those with OP9 cells with a 29.3-fold expansion for c-kit+ hematopoietic progenitor cells and 8.3-fold expansion for CD34+ hematopoietic stem cells. In the controls, HC expansion was lower than in any cocultures, demonstrating the advantages of coculturing for HC expansion. CONCLUSION Stromal cell lines are useful in expanding primitive HCs derived from FLCs in 3D cocultures. Culture density is a pivotal factor for the effective expansion of primitive HCs and this effect differs by culture condition.
Collapse
Affiliation(s)
- Hirotoshi Miyoshi
- Department of Biomedical Engineering, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kenji Abo
- Department of Biomedical Engineering, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Daiki Hosoya
- Department of Biomedical Engineering, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazuyuki Matsuo
- Department of Biomedical Engineering, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yoshio Utsumi
- Department of Biomedical Engineering, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
6
|
Aqmasheh S, Shamsasenjan K, Khalaf Adeli E, Movassaghpourakbari A, Akbarzadehlaleh P, Pashoutan Sarvar D, Timari H. Effect of Mesenchymal Stem Cell-derived Microvesicles on Megakaryocytic Differentiation of CD34 + Hematopoietic Stem Cells. Adv Pharm Bull 2020; 10:315-322. [PMID: 32373502 PMCID: PMC7191234 DOI: 10.34172/apb.2020.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/22/2019] [Accepted: 09/30/2019] [Indexed: 01/13/2023] Open
Abstract
Purpose: Mesenchymal stem cells (MSCs) release hematopoietic cytokines, growth factors, and Microvesicles (MVs) supporting the hematopoietic stem cells (HSCs). MVs released from various cells, playing a crucial role in biological functions of their parental cells. MSC-derived MVs contain microRNAs and proteins with key roles in the regulation of hematopoiesis. Umbilical cord blood (UCB) is a source for transplantation but the long-term recovery of platelets is a main problem. Therefore, we intend to show that MSC-MVs are able to improve the differentiation of UCB-derived CD34+ cells to megakaryocyte lineage. Methods: In this descriptive study, MSCs were cultured in DMEM to collect the culture supernatant, which was ultracentrifuged for the isolation of MVs. HSCs were isolated from UCB using MACS method and cultured in IMDM supplemented with cytokines and MVs in three different conditions. Megakaryocyte differentiation was evaluated through the expression of specific markers and genes after 72 hours, and the data was analyzed by t test (P<0.05). Results: The expression of specific megakaryocyte markers (CD41 and CD61) in the presence of different concentrations of MSC-MVs did not show any significant difference. Also, the expression of specific genes of megakaryocyte lineage was compared with control group. The expression of GATA2 and c-Mpl was significantly increased, GATA1 was not significantly decreased, and FLI1 was significantly decreased. Conclusion: MSC-MVs could improve the expression of specific megakaryocyte genes; however, there was no significant expression of CD markers. Further studies, including the evaluation of late stages of megakaryocyte differentiation, are required to evaluate platelet production and shedding
Collapse
Affiliation(s)
- Sara Aqmasheh
- Umbilical Cord Blood Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasenjan
- Umbilical Cord Blood Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Khalaf Adeli
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | | | - Parvin Akbarzadehlaleh
- Department of Pharmaceutical Biotechnology, Tabriz University of Medical Science, Tabriz, Iran
| | | | - Hamzeh Timari
- Umbilical Cord Blood Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
7
|
Budgude P, Kale V, Vaidya A. Mesenchymal stromal cell‐derived extracellular vesicles as cell‐free biologics for the ex vivo expansion of hematopoietic stem cells. Cell Biol Int 2020; 44:1078-1102. [DOI: 10.1002/cbin.11313] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 01/31/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Pallavi Budgude
- Symbiosis Centre for Stem Cell ResearchSymbiosis International (Deemed University) Pune 412115 India
| | - Vaijayanti Kale
- Symbiosis Centre for Stem Cell ResearchSymbiosis International (Deemed University) Pune 412115 India
| | - Anuradha Vaidya
- Symbiosis Centre for Stem Cell ResearchSymbiosis International (Deemed University) Pune 412115 India
- Symbiosis School of Biological SciencesSymbiosis International (Deemed University) Pune 412115 India
| |
Collapse
|
8
|
PDGFB-expressing mesenchymal stem cells improve human hematopoietic stem cell engraftment in immunodeficient mice. Bone Marrow Transplant 2019; 55:1029-1040. [PMID: 31804621 PMCID: PMC7269905 DOI: 10.1038/s41409-019-0766-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 11/13/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022]
Abstract
The bone marrow (BM) niche regulates multiple hematopoietic stem cell (HSC) processes. Clinical treatment for hematological malignancies by HSC transplantation often requires preconditioning via total body irradiation, which severely and irreversibly impairs the BM niche and HSC regeneration. Novel strategies are needed to enhance HSC regeneration in irradiated BM. We compared the effects of EGF, FGF2, and PDGFB on HSC regeneration using human mesenchymal stem cells (MSCs) that were transduced with these factors via lentiviral vectors. Among the above niche factors tested, MSCs transduced with PDGFB (PDGFB-MSCs) most significantly improved human HSC engraftment in immunodeficient mice. PDGFB-MSC-treated BM enhanced transplanted human HSC self-renewal in secondary transplantations more efficiently than GFP-transduced MSCs (GFP-MSCs). Gene set enrichment analysis showed increased antiapoptotic signaling in PDGFB-MSCs compared with GFP-MSCs. PDGFB-MSCs exhibited enhanced survival and expansion after transplantation, resulting in an enlarged humanized niche cell pool that provide a better humanized microenvironment to facilitate superior engraftment and proliferation of human hematopoietic cells. Our studies demonstrate the efficacy of PDGFB-MSCs in supporting human HSC engraftment.
Collapse
|
9
|
Ajami M, Soleimani M, Abroun S, Atashi A. Comparison of cord blood CD34 + stem cell expansion in coculture with mesenchymal stem cells overexpressing SDF‐1 and soluble /membrane isoforms of SCF. J Cell Biochem 2019; 120:15297-15309. [DOI: 10.1002/jcb.28797] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/30/2019] [Accepted: 02/04/2019] [Indexed: 01/09/2023]
Affiliation(s)
- Mansoureh Ajami
- Department of Hematology and Blood Banking, Faculty of Medical Sciences Tarbiat Modares University Tehran Iran
| | - Masoud Soleimani
- Department of Hematology and Blood Banking, Faculty of Medical Sciences Tarbiat Modares University Tehran Iran
| | - Saeid Abroun
- Department of Hematology and Blood Banking, Faculty of Medical Sciences Tarbiat Modares University Tehran Iran
| | - Amir Atashi
- Department of Hematology and Blood Banking, Faculty of Medical Sciences Tarbiat Modares University Tehran Iran
- Stem Cell and Tissue Engineering Research Center Shahroud University of Medical Sciences Shahroud Iran
| |
Collapse
|
10
|
Li D, Chiu G, Lipe B, Hopkins RA, Lillis J, Ashton JM, Paul S, Aljitawi OS. Decellularized Wharton jelly matrix: a biomimetic scaffold for ex vivo hematopoietic stem cell culture. Blood Adv 2019; 3:1011-1026. [PMID: 30940636 PMCID: PMC6457237 DOI: 10.1182/bloodadvances.2018019315] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 02/10/2019] [Indexed: 12/13/2022] Open
Abstract
Hematopoietic stem progenitor cells (HSPCs) reside in the bone marrow (BM) hematopoietic "niche," a special 3-dimensional (3D) microenvironment that regulates HSPC self-renewal and multipotency. In this study, we evaluated a novel 3D in vitro culture system that uses components of the BM hematopoietic niche to expand umbilical cord blood (UCB) CD34+ cells. We developed this model using decellularized Wharton jelly matrix (DWJM) as an extracellular matrix (ECM) scaffold and human BM mesenchymal stromal cells (MSCs) as supporting niche cells. To assess the efficacy of this model in expanding CD34+ cells, we analyzed UCB CD34+ cells, following culture in DWJM, for proliferation, viability, self-renewal, multilineage differentiation, and transmigration capability. We found that DWJM significantly expanded UCB HSPC subset. It promoted UCB CD34+ cell quiescence, while maintaining their viability, differentiation potential with megakaryocytic differentiation bias, and clonogenic capacity. DWJM induced an increase in the frequency of c-kit+ cells, a population with enhanced self-renewal ability, and in CXCR4 expression in CD34+ cells, which enhanced their transmigration capability. The presence of BM MSCs in DWJM, however, impaired UCB CD34+ cell transmigration and suppressed CXCR4 expression. Transcriptome analysis indicated that DWJM upregulates a set of genes that are specifically involved in megakaryocytic differentiation, cell mobility, and BM homing. Collectively, our results indicate that the DWJM-based 3D culture system is a novel in vitro model that supports the proliferation of UCB CD34+ cells with enhanced transmigration potential, while maintaining their differentiation potential. Our findings shed light on the interplay between DWJM and BM MSCs in supporting the ex vivo culture of human UCB CD34+ cells for use in clinical transplantation.
Collapse
Affiliation(s)
- Dandan Li
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Grace Chiu
- Hematology/Oncology and Bone Marrow Transplant Program, Department of Medicine, University of Rochester Medical Center, Rochester, NY
| | - Brea Lipe
- Hematology/Oncology and Bone Marrow Transplant Program, Department of Medicine, University of Rochester Medical Center, Rochester, NY
| | - Richard A Hopkins
- Cardiac Surgery Research Laboratories, Children's Mercy Hospital and Clinics, Kansas City, MO; and
| | - Jacquelyn Lillis
- Genomics Research Center, University of Rochester Medical Center, Rochester, NY
| | - John M Ashton
- Genomics Research Center, University of Rochester Medical Center, Rochester, NY
| | - Soumen Paul
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Omar S Aljitawi
- Hematology/Oncology and Bone Marrow Transplant Program, Department of Medicine, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
11
|
Xu C, Chen J, Li L, Pu X, Chu X, Wang X, Li M, Lu Y, Zheng X. Promotion of chondrogenic differentiation of mesenchymal stem cells by copper: Implications for new cartilage repair biomaterials. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 93:106-114. [PMID: 30274037 DOI: 10.1016/j.msec.2018.07.074] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 07/14/2018] [Accepted: 07/25/2018] [Indexed: 12/31/2022]
Abstract
Copper (Cu) has drawn considerable attention in the design of biomaterials due to its multifunction, such as antibacterial property, osteogenic and angiogenic ability. However, the effect of Cu on chondrogenic differentiation of mesenchymal stem cells (MSCs) and its potential for cartilage repair biomaterials has been rarely studied. Here, we report that Cu can significantly enhance chondrogensis of MSCs. Specifically, in vitro studies showed that Cu could promote MSCs cytoskeleton change, extracellular glycosaminoglycan (GAG) deposition and the chrodrogenic genes (Sox9, Aggrecan, and Col-2) up-regulation. Furthermore, we prepared a Cu-containing alginate (Alg) porous scaffold to assess the chondroinductivity of Cu in vivo. In eight weeks, we found that Alg/Cu scaffolds could induce better formation of new cartilage tissue compared to the pure Alg scaffolds fabricated by the same procedure but without adding Cu. These encouraging results indicate that Cu can bring considerable benefits to the development and application of cartilage repair biomaterials.
Collapse
Affiliation(s)
- Changkui Xu
- Department of Orthopaedic, Guangzhou Overseas Chinese Hospital, No. 613, West Whampoa Road, Tianhe District, Guangzhou 510630, Guangdong, China; Department of Orthopaedic, Foshan Sanshui District People's Hospital, No. 16, West Guanghai Road, Sanshui District, Foshan 528100, Guangdong, China; Guangdong Key Lab of Orthopaedic Technology and Implant Materials, Key Laboratory of Trauma & Tissue Repair of Tropical Area of PLA, Department of Orthopaedic, General Hospital of Guangzhou Military Command of PLA, No. 111, Liuhua Road, Yuexiu District, Guangzhou 510010, Guangdong, China
| | - Jiarong Chen
- Guangdong Key Lab of Orthopaedic Technology and Implant Materials, Key Laboratory of Trauma & Tissue Repair of Tropical Area of PLA, Department of Orthopaedic, General Hospital of Guangzhou Military Command of PLA, No. 111, Liuhua Road, Yuexiu District, Guangzhou 510010, Guangdong, China
| | - Lihua Li
- Guangdong Key Lab of Orthopaedic Technology and Implant Materials, Key Laboratory of Trauma & Tissue Repair of Tropical Area of PLA, Department of Orthopaedic, General Hospital of Guangzhou Military Command of PLA, No. 111, Liuhua Road, Yuexiu District, Guangzhou 510010, Guangdong, China
| | - Xiaobing Pu
- Department of Orthopaedic, No.4 West China Teaching Hospital of Sichuan University, No. 18, South Renmin Road, Wuhou District, Chengdu 610041, Sichuan, China
| | - Xiao Chu
- Guangdong Key Lab of Orthopaedic Technology and Implant Materials, Key Laboratory of Trauma & Tissue Repair of Tropical Area of PLA, Department of Orthopaedic, General Hospital of Guangzhou Military Command of PLA, No. 111, Liuhua Road, Yuexiu District, Guangzhou 510010, Guangdong, China
| | - Xiaolan Wang
- Guangdong Key Lab of Orthopaedic Technology and Implant Materials, Key Laboratory of Trauma & Tissue Repair of Tropical Area of PLA, Department of Orthopaedic, General Hospital of Guangzhou Military Command of PLA, No. 111, Liuhua Road, Yuexiu District, Guangzhou 510010, Guangdong, China
| | - Mei Li
- Guangdong Key Lab of Orthopaedic Technology and Implant Materials, Key Laboratory of Trauma & Tissue Repair of Tropical Area of PLA, Department of Orthopaedic, General Hospital of Guangzhou Military Command of PLA, No. 111, Liuhua Road, Yuexiu District, Guangzhou 510010, Guangdong, China
| | - Yao Lu
- Guangdong Key Lab of Orthopaedic Technology and Implant Materials, Key Laboratory of Trauma & Tissue Repair of Tropical Area of PLA, Department of Orthopaedic, General Hospital of Guangzhou Military Command of PLA, No. 111, Liuhua Road, Yuexiu District, Guangzhou 510010, Guangdong, China; Department of Orthopaedic, Zhujiang Hospital, Southern Medical University, No. 253, Gongye Road, Haizhu District, Guangzhou 510282, Guangdong, China.
| | - Xiaofei Zheng
- Department of Orthopaedic, Guangzhou Overseas Chinese Hospital, No. 613, West Whampoa Road, Tianhe District, Guangzhou 510630, Guangdong, China.
| |
Collapse
|
12
|
Ghasemzadeh M, Hosseini E, Ahmadi M, Kamalizad M, Amirizadeh N. Comparable osteogenic capacity of mesenchymal stem or stromal cells derived from human amnion membrane and bone marrow. Cytotechnology 2018; 70:729-739. [PMID: 29305674 DOI: 10.1007/s10616-017-0177-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 12/02/2017] [Indexed: 12/15/2022] Open
Abstract
So far, substantial attentions have been attracted to the application of mesenchymal stem or stromal cells (MSCs) in different therapeutic approaches. Although human bone marrow is commonly considered as a major source for MSCs, having an invasive collection method, ethical consideration and donor availability create a challenge for scientists, leading them to explore better alternative sources for MSCs. The study presented here aimed to characterize and compare osteogenic capacity of MSCs obtained from the amnion membrane (AM) with those originated from BM. Cells isolated from AMs and BMs were cultured in DMEM-low glucose supplemented with FBS, penicillin and streptomycin. After 24 h of incubation, cells adhered to the plastic surface of the flasks were allowed to proliferate for more days. A sub-confluent culture of cells was trypsinized and re-cultured. The MSCs were characterized by the expression of specific markers with flow cytometry. The osteogenic differentiation of MSCs was also validated by alkaline phosphatase and alizarian red S staining. Our results showed comparable expression of MSCs specific markers for both MSC sources (AM and BM). We also showed the optimum osteogenic differentiation of MSCs from both sources whereas hAM-MSCs revealed higher proliferation rate. We found no essential immunophenotypic differences between MSCs originated from bone marrow and amnion membrane while their differentiations into osteoblastic linage were also comparable. This was in addition to the higher proliferation rate observed for hAM-MSCs which suggests hAM as an easily accessible and reliable source of MSCs applicable for bone engineering, regenerative medicine or other therapeutic approaches.
Collapse
Affiliation(s)
- Mehran Ghasemzadeh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization Building, Hemmat Express Way, Next to the Milad Tower, Tehran, 14665-1157, Iran
| | - Ehteramolsadat Hosseini
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization Building, Hemmat Express Way, Next to the Milad Tower, Tehran, 14665-1157, Iran.
| | - Mohammadhossein Ahmadi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization Building, Hemmat Express Way, Next to the Milad Tower, Tehran, 14665-1157, Iran
| | - Maedeh Kamalizad
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization Building, Hemmat Express Way, Next to the Milad Tower, Tehran, 14665-1157, Iran
| | - Naser Amirizadeh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization Building, Hemmat Express Way, Next to the Milad Tower, Tehran, 14665-1157, Iran
| |
Collapse
|
13
|
Timari H, Shamsasenjan K, Movassaghpour A, Akbarzadehlaleh P, Pashoutan Sarvar D, Aqmasheh S. The Effect of Mesenchymal Stem Cell-Derived Extracellular Vesicles on Hematopoietic Stem Cells Fate. Adv Pharm Bull 2017; 7:531-546. [PMID: 29399543 PMCID: PMC5788208 DOI: 10.15171/apb.2017.065] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/25/2017] [Accepted: 11/28/2017] [Indexed: 12/16/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are multipotent stem cells, with self-renewal ability as well as ability to generate all blood cells. Mesenchymal stem cells (MSCs) are multipotent stem cells, with self-renewal ability, and capable of differentiating into a variety of cell types. MSCs have supporting effects on hematopoiesis; through direct intercellular communications as well as secreting cytokines, chemokines, and extracellular vesicles (EVs). Recent investigations demonstrated that some biological functions and effects of MSCs are mediated by their EVs. MSC-EVs are the cell membrane and endosomal membrane compartments, which are important mediators in the intercellular communications. MSC-EVs contain some of the molecules such as proteins, mRNA, siRNA, and miRNA from their parental cells. MSC-EVs are able to inhibit tumor, repair damaged tissue, and modulate immune system responses. MSC-EVs compared to their parental cells, may have the specific safety advantages such as the lower potential to trigger immune system responses and limited side effects. Recently some studies demonstrated the effect of MSC-EVs on the expansion, differentiation, and clinical applications of HSCs such as improvement of hematopoietic stem cell transplantation (HSCT) and inhibition of graft versus host disease (GVHD). HSCT may be the only therapeutic choice for patients who suffer from malignant and non-malignant hematological disorders. However, there are several severe side effects such GVHD that restricts the successfulness of HSCT. In this review, we will discuss the most important effects of MSCs and MSC-EVs on the improvement of HSCT, inhibition and treatment of GVHD, as well as, on the expansion of HSCs.
Collapse
Affiliation(s)
- Hamze Timari
- Stem Cell Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasenjan
- Stem Cell Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aliakbar Movassaghpour
- Hematology Oncology Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Akbarzadehlaleh
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Sara Aqmasheh
- Stem Cell Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
14
|
Expression of Surface Molecules in Human Mesenchymal Stromal Cells Co-Cultured with Nucleated Umbilical Cord Blood Cells. Bull Exp Biol Med 2017; 162:578-582. [DOI: 10.1007/s10517-017-3662-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Indexed: 12/20/2022]
|
15
|
Perucca S, Di Palma A, Piccaluga PP, Gemelli C, Zoratti E, Bassi G, Giacopuzzi E, Lojacono A, Borsani G, Tagliafico E, Scupoli MT, Bernardi S, Zanaglio C, Cattina F, Cancelli V, Malagola M, Krampera M, Marini M, Almici C, Ferrari S, Russo D. Mesenchymal stromal cells (MSCs) induce ex vivo proliferation and erythroid commitment of cord blood haematopoietic stem cells (CB-CD34+ cells). PLoS One 2017; 12:e0172430. [PMID: 28231331 PMCID: PMC5322933 DOI: 10.1371/journal.pone.0172430] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 02/03/2017] [Indexed: 12/30/2022] Open
Abstract
A human bone marrow-derived mesenchymal stromal cell (MSCs) and cord blood-derived CD34+ stem cell co-culture system was set up in order to evaluate the proliferative and differentiative effects induced by MSCs on CD34+ stem cells, and the reciprocal influences on gene expression profiles. After 10 days of co-culture, non-adherent (SN-fraction) and adherent (AD-fraction) CD34+ stem cells were collected and analysed separately. In the presence of MSCs, a significant increase in CD34+ cell number was observed (fold increase = 14.68), mostly in the SN-fraction (fold increase = 13.20). This was combined with a significant increase in CD34+ cell differentiation towards the BFU-E colonies and with a decrease in the CFU-GM. These observations were confirmed by microarray analysis. Through gene set enrichment analysis (GSEA), we noted a significant enrichment in genes involved in heme metabolism (e.g. LAMP2, CLCN3, BMP2K), mitotic spindle formation and proliferation (e.g. PALLD, SOS1, CCNA1) and TGF-beta signalling (e.g. ID1) and a down-modulation of genes participating in myeloid and lymphoid differentiation (e.g. PCGF2) in the co-cultured CD34+ stem cells. On the other hand, a significant enrichment in genes involved in oxygen-level response (e.g. TNFAIP3, SLC2A3, KLF6) and angiogenesis (e.g. VEGFA, IGF1, ID1) was found in the co-cultured MSCs. Taken together, our results suggest that MSCs can exert a priming effect on CD34+ stem cells, regulating their proliferation and erythroid differentiation. In turn, CD34+ stem cells seem to be able to polarise the BM-niche towards the vascular compartment by modulating molecular pathways related to hypoxia and angiogenesis.
Collapse
Affiliation(s)
- Simone Perucca
- Unit of Blood Diseases and Stem Cells Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
- Laboratorio CREA (Centro di Ricerca Emato-oncologica AIL), ASST Spedali Civili of Brescia, Brescia, Italy
| | - Andrea Di Palma
- Unit of Blood Diseases and Stem Cells Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
- Laboratorio CREA (Centro di Ricerca Emato-oncologica AIL), ASST Spedali Civili of Brescia, Brescia, Italy
| | - Pier Paolo Piccaluga
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), S. Orsola-Malpighi Hospital, Bologna University School of Medicine, Bologna, Italy
- Section of Genomics and Personalized Medicine, Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy
| | - Claudia Gemelli
- Parco Scientifico e Tecnologico Materiali Innovativi e Ricerca Applicata del Mirandolese, Modena, Italy
| | - Elisa Zoratti
- Applied Research on Cancer-Network (ARC-NET), University of Verona, Verona, Italy
| | - Giulio Bassi
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Edoardo Giacopuzzi
- Unit of Biology and Genetics, Department of Molecular and Translational Medicine (DMTM), University of Brescia, Brescia, Italy
| | - Andrea Lojacono
- U.O. of Obstetrics and Gynecology I, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Giuseppe Borsani
- Unit of Biology and Genetics, Department of Molecular and Translational Medicine (DMTM), University of Brescia, Brescia, Italy
| | - Enrico Tagliafico
- Centro di Ricerche Genomiche, Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, Modena, Italy
| | - Maria Teresa Scupoli
- Interdepartmental Laboratory of Medical Research (LURM), University of Verona, Verona, Italy
| | - Simona Bernardi
- Unit of Blood Diseases and Stem Cells Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
- Laboratorio CREA (Centro di Ricerca Emato-oncologica AIL), ASST Spedali Civili of Brescia, Brescia, Italy
| | - Camilla Zanaglio
- Unit of Blood Diseases and Stem Cells Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
- Laboratorio CREA (Centro di Ricerca Emato-oncologica AIL), ASST Spedali Civili of Brescia, Brescia, Italy
| | - Federica Cattina
- Unit of Blood Diseases and Stem Cells Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Valeria Cancelli
- Unit of Blood Diseases and Stem Cells Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Michele Malagola
- Unit of Blood Diseases and Stem Cells Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Mauro Krampera
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Mirella Marini
- Department of Transfusion Medicine, Laboratory for Stem Cells Manipulation and Cryopreservation, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Camillo Almici
- Department of Transfusion Medicine, Laboratory for Stem Cells Manipulation and Cryopreservation, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Sergio Ferrari
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Domenico Russo
- Unit of Blood Diseases and Stem Cells Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
- * E-mail:
| |
Collapse
|
16
|
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: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
17
|
Yun JW, Ahn JH, Kwon E, Kim SH, Kim H, Jang JJ, Kim WH, Kim JH, Han SY, Kim JT, Kim JH, Kim W, Ku SY, Do BR, Kang BC. Human umbilical cord-derived mesenchymal stem cells in acute liver injury: Hepatoprotective efficacy, subchronic toxicity, tumorigenicity, and biodistribution. Regul Toxicol Pharmacol 2016; 81:437-447. [PMID: 27693706 DOI: 10.1016/j.yrtph.2016.09.029] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 09/26/2016] [Accepted: 09/27/2016] [Indexed: 12/21/2022]
Abstract
Umbilical cord-derived mesenchymal stem cells (UC-MSCs) therapy might be an alternative to liver transplantation for acute or chronic liver injury. The aim of this study was to evaluate the efficacy of human UC-MSCs on carbon tetrachloride (CCl4)-induced acute liver injury. In addition, its toxicity, tumorigenicity, and biodistribution were determined. Significant hepatoprotective effects of hUC-MSCs with decreased levels of hepatocellular necrosis and lobular neutrophilic infiltration were found. Regarding the safety of hUC-MSCs, no serious hUC-MSCs-related changes (body weight, food/water consumption, clinical symptom, urinalysis, hematology, clinical chemistry, organ weight, and histopathology) were observed in a 13-week subchronic toxicity study. In a 26-week tumorigenicity study, no mice developed tumor related to hUC-MSCs transplantation up to 1 × 108 cells/kg. In particular, human mitochondrial sequence detection revealed that most hUC-MSCs were cleared from the major organs of the mice at 13 weeks after transplantation. There was no systemic toxicity or neoplastic finding either. Taken together, these results suggested that hUC-MSCs have great potential for future clinical treatment of acute liver disease.
Collapse
Affiliation(s)
- Jun-Won Yun
- Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jae Hun Ahn
- Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Euna Kwon
- Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seung-Hyun Kim
- Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hanna Kim
- Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Ja-June Jang
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Woo Ho Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ji Hyang Kim
- Biotechnology Research Institute, Hurim BioCell Co. Ltd., Seoul, Republic of Korea
| | - Su-Youne Han
- Biotechnology Research Institute, Hurim BioCell Co. Ltd., Seoul, Republic of Korea
| | - Jin Tac Kim
- Biotechnology Research Institute, Hurim BioCell Co. Ltd., Seoul, Republic of Korea
| | - Jong-Hoon Kim
- Laboratory of Stem Cell Biology, Division of Biotechnology, Department of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Wookhwan Kim
- Department of General Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Seung-Yup Ku
- Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Byung-Rok Do
- Biotechnology Research Institute, Hurim BioCell Co. Ltd., Seoul, Republic of Korea.
| | - Byeong-Cheol Kang
- Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea; Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea; Biomedical Center for Animal Resource and Development, Seoul National University College of Medicine, Seoul, Republic of Korea; Designed Animal and Transplantation Research Institute, Institute of GreenBio Science Technology, Seoul National University, Pyeongchang-gun, Gangwon-do, Republic of Korea.
| |
Collapse
|
18
|
Psatha N, Karponi G, Yannaki E. Optimizing autologous cell grafts to improve stem cell gene therapy. Exp Hematol 2016; 44:528-39. [PMID: 27106799 DOI: 10.1016/j.exphem.2016.04.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 04/06/2016] [Accepted: 04/08/2016] [Indexed: 10/21/2022]
Abstract
Over the past decade, stem cell gene therapy has achieved unprecedented curative outcomes for several genetic disorders. Despite the unequivocal success, clinical gene therapy still faces challenges. Genetically engineered hematopoietic stem cells are particularly vulnerable to attenuation of their repopulating capacity once exposed to culture conditions, ultimately leading to low engraftment levels posttransplant. This becomes of particular importance when transduction rates are low or/and competitive transplant conditions are generated by reduced-intensity conditioning in the absence of a selective advantage of the transduced over the unmodified cells. These limitations could partially be overcome by introducing megadoses of genetically modified CD34(+) cells into conditioned patients or by transplanting hematopoietic stem cells hematopoietic stem cells with high engrafting and repopulating potential. On the basis of the lessons gained from cord blood transplantation, we summarize the most promising approaches to date of increasing either the numbers of hematopoietic stem cells for transplantation or/and their engraftability, as a platform toward the optimization of engineered stem cell grafts.
Collapse
Affiliation(s)
- Nikoletta Psatha
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece; Department of Medicine, University of Washington, Seattle, WA
| | - Garyfalia Karponi
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece
| | - Evangelia Yannaki
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece; Department of Medicine, University of Washington, Seattle, WA.
| |
Collapse
|
19
|
Zou Q, Wu M, Zhong L, Fan Z, Zhang B, Chen Q, Ma F. Development of a Xeno-Free Feeder-Layer System from Human Umbilical Cord Mesenchymal Stem Cells for Prolonged Expansion of Human Induced Pluripotent Stem Cells in Culture. PLoS One 2016; 11:e0149023. [PMID: 26882313 PMCID: PMC4755601 DOI: 10.1371/journal.pone.0149023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 01/26/2016] [Indexed: 02/06/2023] Open
Abstract
Various feeder layers have been extensively applied to support the prolonged growth of human pluripotent stem cells (hPSCs) for in vitro cultures. Among them, mouse embryonic fibroblast (MEF) and mouse fibroblast cell line (SNL) are most commonly used feeder cells for hPSCs culture. However, these feeder layers from animal usually cause immunogenic contaminations, which compromises the potential of hPSCs in clinical applications. In the present study, we tested human umbilical cord mesenchymal stem cells (hUC-MSCs) as a potent xeno-free feeder system for maintaining human induced pluripotent stem cells (hiPSCs). The hUC-MSCs showed characteristics of MSCs in xeno-free culture condition. On the mitomycin-treated hUC-MSCs feeder, hiPSCs maintained the features of undifferentiated human embryonic stem cells (hESCs), such as low efficiency of spontaneous differentiation, stable expression of stemness markers, maintenance of normal karyotypes, in vitro pluripotency and in vivo ability to form teratomas, even after a prolonged culture of more than 30 passages. Our study indicates that the xeno-free culture system may be a good candidate for growth and expansion of hiPSCs as the stepping stone for stem cell research to further develop better and safer stem cells.
Collapse
Affiliation(s)
- Qing Zou
- Research Center for Stem Cell and Regenerative Medicine, Sichuan Neo-life Stem Cell Biotech INC., Chengdu, Sichuan, China
- Center for Stem Cell Research & Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, Sichuan, China
| | - Mingjun Wu
- Research Center for Stem Cell and Regenerative Medicine, Sichuan Neo-life Stem Cell Biotech INC., Chengdu, Sichuan, China
| | - Liwu Zhong
- Research Center for Stem Cell and Regenerative Medicine, Sichuan Neo-life Stem Cell Biotech INC., Chengdu, Sichuan, China
| | - Zhaoxin Fan
- Research Center for Stem Cell and Regenerative Medicine, Sichuan Neo-life Stem Cell Biotech INC., Chengdu, Sichuan, China
| | - Bo Zhang
- Research Center for Stem Cell and Regenerative Medicine, Sichuan Neo-life Stem Cell Biotech INC., Chengdu, Sichuan, China
| | - Qiang Chen
- Research Center for Stem Cell and Regenerative Medicine, Sichuan Neo-life Stem Cell Biotech INC., Chengdu, Sichuan, China
- Center for Stem Cell Research & Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, Sichuan, China
- * E-mail: (FM); (QC)
| | - Feng Ma
- Research Center for Stem Cell and Regenerative Medicine, Sichuan Neo-life Stem Cell Biotech INC., Chengdu, Sichuan, China
- Center for Stem Cell Research & Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, Sichuan, China
- State Key Laboratory of Experimental Hematology, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- * E-mail: (FM); (QC)
| |
Collapse
|
20
|
Sorokina T, Shipounova I, Bigildeev A, Petinati N, Drize N, Turkina A, Chelysheva E, Shukhov O, Kuzmina L, Parovichnikova E, Savchenko V. The ability of multipotent mesenchymal stromal cells from the bone marrow of patients with leukemia to maintain normal hematopoietic progenitor cells. Eur J Haematol 2016; 97:245-52. [PMID: 26643284 DOI: 10.1111/ejh.12713] [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] [Accepted: 11/25/2015] [Indexed: 02/05/2023]
Abstract
BACKGROUND The development of leukemia impairs normal hematopoiesis and marrow stromal microenvironment. The aim of the investigation was to study the ability of multipotent mesenchymal stromal cells (MSCs) derived from the bone marrow of patients with leukemia to maintain normal hematopoietic progenitor cells. METHODS MSCs were obtained from the bone marrow of 14 patients with acute lymphoblastic (ALL), 25 with myeloid (AML), and 15 with chronic myeloid (CML) leukemia. As a control, MSCs from 22 healthy donors were used. The incidence of cobblestone area forming cells (CAFC 7-8 d) in the bone marrow of healthy donor cultivated on the supportive layer of patients MSCs was measured. RESULTS The ability of MSCs from AML and ALL patients at the moment of diagnosis to maintain normal CAFC was significantly decreased when compared to donors. After chemotherapy, the restoration of ALL patients' MSCs functions was slower than that of AML. CML MSCs maintained CAFC better than donors' at the moment of diagnosis and this ability increased with treatment. CONCLUSIONS The ability of patients' MSCs to maintain normal hematopoietic progenitor cells was shown to change in comparison with MSCs from healthy donors and depended on nosology. During treatment, the functional capacity of patients' MSCs had been partially restored.
Collapse
Affiliation(s)
| | | | | | | | - Nina Drize
- National Research Center for Hematology, Moscow, Russia
| | - Anna Turkina
- National Research Center for Hematology, Moscow, Russia
| | | | - Oleg Shukhov
- National Research Center for Hematology, Moscow, Russia
| | | | | | | |
Collapse
|
21
|
Isolation, characterisation and comparative analysis of human umbilical cord vein perivascular cells and cord blood mesenchymal stem cells. Cell Tissue Bank 2015; 17:345-52. [PMID: 26679930 DOI: 10.1007/s10561-015-9542-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 12/15/2015] [Indexed: 01/05/2023]
Abstract
Perivascular cells are known to be ancestors of mesenchymal stem cells (MSCs) and can be obtained from heart, skin, bone marrow, eye, placenta and umbilical cord (UC). However detailed characterization of perivascular cells around the human UC vein and comparative analysis of them with MSCs haven't been done yet. In this study, our aim is to isolate perivascular cells from human UC vein and characterize them versus UC blood MSCs (UCB-MSCs). For this purpose, perivascular cells around the UC vein were isolated enzymatically and then purified with magnetic activated cell sorting (MACS) method using CD146 Microbead Kit respectively. MSCs were isolated from UCB by Ficoll density gradient solution. Perivascular cells and UCB-MSCs were characterized by osteogenic and adipogenic differentiation procedures, flow cytometric analysis [CD146, CD105, CD31, CD34, CD45 and alpha-smooth muscle actin (α-SMA)], and immunofluorescent staining (MAP1B and Tenascin C). Alizarin red and Oil red O staining results showed that perivascular cells and MSCs had osteogenic and adipogenic differentiation capacity. However, osteogenic differentiation capacity of perivascular cells were found to be less than UCB-MSCs. According to flow cytometric analysis, CD146 expression of perivascular cells were appeared to be 4.8-fold higher than UCB-MSCs. Expression of α-SMA, MAP1B and Tenascin-C from perivascular cells was determined by flow cytometry analysis and immunfluorescent staining. The results appear to support the fact that perivascular cells are the ancestors of MSCs in vascular area. They may be used as alternative cells to MSCs in the field of vascular tissue engineering.
Collapse
|
22
|
Kadekar D, Kale V, Limaye L. Differential ability of MSCs isolated from placenta and cord as feeders for supporting ex vivo expansion of umbilical cord blood derived CD34(+) cells. Stem Cell Res Ther 2015; 6:201. [PMID: 26481144 PMCID: PMC4617445 DOI: 10.1186/s13287-015-0194-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 07/15/2015] [Accepted: 09/28/2015] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Ex vivo expansion of umbilical cord blood (UCB) is attempted to increase cell numbers to overcome the limitation of cell dose. Presently, suspension cultures or feeder mediated co-cultures are performed for expansion of hematopoietic stem cells (HSCs). Mesenchymal stem cells (MSCs) have proved to be efficient feeders for the maintenance of HSCs. Here, we have established MSCs-HSCs co-culture system with MSCs isolated from less invasive and ethically acceptable sources like umbilical cord tissue (C-MSCs) and placenta (P-MSCs). MSCs derived from these tissues are often compared with bone marrow derived MSCs (BM-MSCs) which are considered as a gold standard. However, so far none of the studies have directly compared C-MSCs with P-MSCs as feeders for ex vivo expansion of HSCs. Thus, we for the first time performed a systematic comparison of hematopoietic supportive capability of C and P-MSCs using paired samples. METHODS UCB-derived CD34(+) cells were isolated and co-cultured on irradiated C and P-MSCs for 10 days. C-MSCs and P-MSCs were isolated from the same donor. The cultures comprised of serum-free medium supplemented with 25 ng/ml each of SCF, TPO, Flt-3 L and IL-6. After 10 days cells were collected and analyzed for phenotype and functionality. RESULTS C-MSCs and P-MSCs were found to be morphologically and phenotypically similar but exhibited differential ability to support ex vivo hematopoiesis. Cells expanded on P-MSCs showed higher percentage of primitive cells (CD34(+)CD38(-)), CFU (Colony forming unit) content and LTC-IC (Long term culture initiating cells) ability. CD34(+) cells expanded on P-MSCs also exhibited better in vitro adhesion to fibronectin and migration towards SDF-1α and enhanced NOD/SCID repopulation ability, as compared to those grown on C-MSCs. P-MSCs were found to be closer to BM-MSCs in their ability to expand HSCs. P-MSCs supported expansion of functionally superior HSCs by virtue of reduction in apoptosis of primitive HSCs, higher Wnt and Notch activity, HGF secretion and cell-cell contact. On the other hand, C-MSCs facilitated expansion of progenitors (CD34(+)CD38(+)) and differentiated (CD34(-)CD38(+)) cells by secretion of IL1-α, β, MCP-2, 3 and MIP-3α. CONCLUSIONS P-MSCs were found to be better feeders for ex vivo maintenance of primitive HSCs with higher engraftment potential than the cells expanded with C-MSCs as feeders.
Collapse
Affiliation(s)
- Darshana Kadekar
- Stem Cell Laboratory, National Centre for Cell Science, University of Pune Campus, Ganeshkhind, Pune, 411007, Maharashtra, India.
| | - Vaijayanti Kale
- Stem Cell Laboratory, National Centre for Cell Science, University of Pune Campus, Ganeshkhind, Pune, 411007, Maharashtra, India.
| | - Lalita Limaye
- Stem Cell Laboratory, National Centre for Cell Science, University of Pune Campus, Ganeshkhind, Pune, 411007, Maharashtra, India.
| |
Collapse
|
23
|
|
24
|
Alcayaga-Miranda F, Cuenca J, Luz-Crawford P, Aguila-Díaz C, Fernandez A, Figueroa FE, Khoury M. Characterization of menstrual stem cells: angiogenic effect, migration and hematopoietic stem cell support in comparison with bone marrow mesenchymal stem cells. Stem Cell Res Ther 2015; 6:32. [PMID: 25889741 PMCID: PMC4404686 DOI: 10.1186/s13287-015-0013-5] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 02/20/2015] [Accepted: 02/20/2015] [Indexed: 12/24/2022] Open
Abstract
Introduction Stem cells isolated from menstrual fluid (MenSCs) exhibit mesenchymal stem cell (MSCs)-like properties including multi-lineage differentiation capacity. Besides, menstrual fluid has important advantages over other sources for the isolation of MSCs, including ease of access and repeated sampling in a noninvasive manner. Such attributes allow the rapid culture of MenSCs in numbers that are sufficient for therapeutical doses, at lower cell passages. Methods In this study, we advance the characterization of MenSC populations in comparison to bone marrow derived mesenchymal stem cells (BM-MSCs) with regards to proliferation, lineage differentiation, migration potential, secretion profile and angiogenic properties in vitro and in a matrigel plug assay in mice. We additionally tested their ability to support hematopoietic stem cell (HSC) expansion in vitro. Results The phenotypic analysis of MenSCs revealed a profile largely similar to the BM-MSCs with the exception of a higher expression of the adhesion molecule CD49a (alpha1-integrin). Furthermore, the fibroblast colony forming units (CFU-F) from MenSCs yielded a 2 to 4 fold higher frequency of progenitors and their in vitro migration capacity was superior to BM-MSCs. In addition, MenSCs evidenced a superior paracrine response to hypoxic conditions as evidenced by the secretion of vascular endothelial growth factor and basic fibroblast growth factor and also improved angiogenic effect of conditioned media on endothelial cells. Furthermore, MenSCs were able to induce angiogenesis in a matrigel plug assay in vivo. Thus, an 8-fold increase in hemoglobin content was observed in implanted plugs containing MenSCs compared to BM-MSCs. Finally, we demonstrated, for the first time, the capacity of MenSCs to support the ex-vivo expansion of HSCs, since higher expansion rates of the CD34 + CD133+ population as well as higher numbers of early progenitor (CFU-GEMM) colonies were observed in comparison to the BM source. Conclusions We present evidence showing superiority of MenSCs with respect to several functional aspects, in comparison with BM-MSCs. However, the impact of such properties in their use as adult-derived stem cells for regenerative3 medicine remains to be clarified. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0013-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Francisca Alcayaga-Miranda
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile. .,Cells for Cells, Santiago, Chile.
| | - Jimena Cuenca
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile. .,Cells for Cells, Santiago, Chile.
| | | | | | | | - Fernando E Figueroa
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile.
| | - Maroun Khoury
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile. .,Cells for Cells, Santiago, Chile.
| |
Collapse
|
25
|
Kılıç Ç, Uğur M, Ünlü BS, Yıldız Y, Artar İ, Karlı P, Turgut K. Comparison of serum granulocyte colony-stimulating factor levels between preterm and term births. J Turk Ger Gynecol Assoc 2015; 15:208-11. [PMID: 25584027 DOI: 10.5152/jtgga.2014.14092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 08/18/2014] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE Preterm birth (PTB) is the major obstetric problem in developed countries, accounting for the majority of neonatal mortality and morbidity. Granulocyte colony-stimulating factor (G-CSF) is a hematopoietic cytokine that mediates the increase in leukocytes in pregnancy and may play a role in placentation. We aimed to investigate the differences of serum G-CSF levels between subsequent spontaneous PTB and term-delivered healthy pregnant women. MATERIAL AND METHODS Serum samples, collected from total of 600 singleton otherwise healthy pregnants at 24-28 weeks of gestation during a routine antenatal visit, were used to assess G-CSF levels; 40 of the total pregnants who delivered their infants spontaneously after preterm labor before 37 weeks of gestation were selected as the study group. Also, 120 pregnants were selected as a control group using a 1/3 ratio. Student's t-test, chi-square test, Mann-Whitney U-tests, and ROC curve analysis for prediction of PTB were used for the comparison of groups. P<0.05 was accepted as statistically significant. RESULTS There was no significant difference in maternal serum G-CSF levels between the study and control groups (p=0.28) but maternal white blood cell (WBC) count was significantly different between them (p=0.00). In addition, G-CSF was insufficient in the prediction of PTB (AUC=0.419). In the preterm and term groups, no correlation was found between WBC and G-CSF (p=0.165 vs. p=0.703). CONCLUSION There were no differences in serum levels of G-CSF between term- and preterm-delivered pregnants. There was no predictive role for serum G-CSF in PTB.
Collapse
Affiliation(s)
- Çiğdem Kılıç
- Department of Obstetrics and Gynecology, Zekai Tahir Burak Women's Health Education and Research Hospital, Ankara, Turkey
| | - Mustafa Uğur
- Department of Obstetrics and Gynecology, Zekai Tahir Burak Women's Health Education and Research Hospital, Ankara, Turkey
| | - Bekir Serdar Ünlü
- Department of Obstetrics and Gynecology, Zekai Tahir Burak Women's Health Education and Research Hospital, Ankara, Turkey
| | - Yunus Yıldız
- Department of Obstetrics and Gynecology, Zekai Tahir Burak Women's Health Education and Research Hospital, Ankara, Turkey
| | - İshak Artar
- Department of Obstetrics and Gynecology, Zekai Tahir Burak Women's Health Education and Research Hospital, Ankara, Turkey
| | - Pervin Karlı
- Department of Obstetrics and Gynecology, Zekai Tahir Burak Women's Health Education and Research Hospital, Ankara, Turkey
| | - Kadriye Turgut
- Department of Obstetrics and Gynecology, Zekai Tahir Burak Women's Health Education and Research Hospital, Ankara, Turkey
| |
Collapse
|
26
|
Chatzistamatiou TK, Papassavas AC, Michalopoulos E, Gamaloutsos C, Mallis P, Gontika I, Panagouli E, Koussoulakos SL, Stavropoulos-Giokas C. Optimizing isolation culture and freezing methods to preserve Wharton's jelly's mesenchymal stem cell (MSC) properties: an MSC banking protocol validation for the Hellenic Cord Blood Bank. Transfusion 2014; 54:3108-3120. [PMID: 24894363 DOI: 10.1111/trf.12743] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 04/03/2014] [Accepted: 04/11/2014] [Indexed: 02/05/2023]
Abstract
BACKGROUND Mesenchymal stem or stromal cells (MSCs) are a heterogeneous population that can be isolated from many tissues including umbilical cord Wharton's jelly (UC-WJ). Although initially limited in studies such as a hematopoietic stem cell transplantation adjuvant, an increasing number of clinical trials consider MSCs as a potential anti-inflammatory or a regenerative medicine agent. It has been proposed that creating a repository of MSCs would increase their availability for clinical applications. The aim of this study was to assess the optimal isolation and cryopreservation procedures to facilitate WJ MSC banking. STUDY DESIGN AND METHODS Cells were isolated from UC-WJ using enzymatic digestion or plastic adhesion methods. Their isolation efficacy, growth kinetics, immunophenotype, and differentiation potential were studied, as well as the effects of freezing. Flow cytometry for common MSC markers was performed on all cases and differentiation was shown with histocytochemical staining. Finally, the isolation efficacy on cryopreserved WJ tissue fragments was tested. RESULTS MSC isolation was successful using both isolation methods on fresh UC-WJ tissue. However, UC-WJ MSC isolation from frozen tissue fragments was impossible. Flow cytometry analysis revealed that only MSC markers were expressed on the surface of the isolated cells while differentiation assays showed that they were capable of trilinear differentiation. All the above characteristics were also preserved in isolated UC-WJ MSCs over the cryopreservation study period. CONCLUSION These data showed that viable MSCs can only be isolated from fresh UC-WJ tissue, setting the foundation for clinical-grade banking.
Collapse
|
27
|
Jeon SY, Park JS, Yang HN, Lim HJ, Yi SW, Park H, Park KH. Co-delivery of Cbfa-1-targeting siRNA and SOX9 protein using PLGA nanoparticles to induce chondrogenesis of human mesenchymal stem cells. Biomaterials 2014; 35:8236-48. [PMID: 24965885 DOI: 10.1016/j.biomaterials.2014.05.092] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 05/29/2014] [Indexed: 01/16/2023]
Abstract
During stem cell differentiation, various cellular responses occur that are mediated by transcription factors and proteins. This study evaluated the abilities of SOX9, a crucial protein during the early stage of chondrogenesis, and siRNA targeting Cbfa-1, a transcription factor that promotes osteogenesis, to stimulate chondrogenesis. Non-toxic poly-(d,l-lactide-co-glycolide) (PLGA) nanoparticles (NPs) were coated with Cbfa-1-targeting siRNA and loaded with SOX9 protein. Coomassie blue staining and circular dichroism revealed that the loaded SOX9 protein maintained its stability and bioactivity. These NPs easily entered human mesenchymal stem cells (hMSCs) in vitro and caused them to differentiate into chondrocytes. Markers that are typically expressed in mature chondrocytes were examined. These markers were highly expressed at the mRNA and protein levels in hMSCs treated with PLGA NPs coated with Cbfa-1-targeting siRNA and loaded with SOX9 protein. By contrast, these cells did not express osteogenesis-related markers. hMSCs were injected into mice following internalization of PLGA NPs coated with Cbfa-1-targeting siRNA and loaded with SOX9 protein. When the injection site was excised, markers of chondrogenesis were found to be highly expressed at the mRNA and protein levels, similar to the in vitro results. When hMSCs internalized these NPs and were then cultured in vitro or injected into mice, chondrogenesis-related extracellular matrix components were highly expressed.
Collapse
Affiliation(s)
- Su Yeon Jeon
- Department of Biomedical Science, College of Life Science, CHA University, 3F, Yatap Ace-Core Building, 502 Yatap-Dong, Bundang-Gu, Seongnam-Si, Gyeonggi-Do 135-081, Republic of Korea
| | - Ji Sun Park
- Department of Biomedical Science, College of Life Science, CHA University, 3F, Yatap Ace-Core Building, 502 Yatap-Dong, Bundang-Gu, Seongnam-Si, Gyeonggi-Do 135-081, Republic of Korea
| | - Han Na Yang
- Department of Biomedical Science, College of Life Science, CHA University, 3F, Yatap Ace-Core Building, 502 Yatap-Dong, Bundang-Gu, Seongnam-Si, Gyeonggi-Do 135-081, Republic of Korea
| | - Hye Jin Lim
- Department of Biomedical Science, College of Life Science, CHA University, 3F, Yatap Ace-Core Building, 502 Yatap-Dong, Bundang-Gu, Seongnam-Si, Gyeonggi-Do 135-081, Republic of Korea
| | - Se Won Yi
- Department of Biomedical Science, College of Life Science, CHA University, 3F, Yatap Ace-Core Building, 502 Yatap-Dong, Bundang-Gu, Seongnam-Si, Gyeonggi-Do 135-081, Republic of Korea
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang Univeristy, 221 Heukseok-dong, Dongjak-gu, Seoul 156-756, Republic of Korea
| | - Keun-Hong Park
- Department of Biomedical Science, College of Life Science, CHA University, 3F, Yatap Ace-Core Building, 502 Yatap-Dong, Bundang-Gu, Seongnam-Si, Gyeonggi-Do 135-081, Republic of Korea.
| |
Collapse
|
28
|
IFN-γ-secreting-mesenchymal stem cells exert an antitumor effect in vivo via the TRAIL pathway. J Immunol Res 2014; 2014:318098. [PMID: 24971369 PMCID: PMC4058226 DOI: 10.1155/2014/318098] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 05/05/2014] [Accepted: 05/05/2014] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can exhibit either prooncogenic or antitumor properties depending on the context. Based on our previous study, we hypothesized that MSCs engineered to deliver IFN-γ would kill cancer cells through persistent activation of the TRAIL pathway. Human bone-marrow (BM-) derived MSCs were isolated, amplified, and transduced with a lentiviral vector encoding the IFN-γ gene under the control of the EF1α promoter. The IFN-γ-modified MSCs effectively secreted functional IFN-γ, which led to long-term expression of TRAIL. More importantly, the IFN-γ-modified MSCs selectively induced apoptosis in lung tumor cells through caspase-3 activation within the target cells. The percentage of activated-caspase-3-positive tumor cells in IFN-γ-modified MSCs cocultures was significantly higher than in control MSCs cocultures. Treatment with anti-TRAIL antibody dramatically suppressed the caspase-3 activation observed in H460 cells. After injection into nude mice, the IFN-γ-modified MSCs inhibited the growth and progression of lung carcinoma compared with control cells. Collectively, our results provide a new strategy for tumor therapy that utilizes IFN-γ-modified MSCs.
Collapse
|