51
|
Liu FD, Pishesha N, Poon Z, Kaushik T, Van Vliet KJ. Material Viscoelastic Properties Modulate the Mesenchymal Stem Cell Secretome for Applications in Hematopoietic Recovery. ACS Biomater Sci Eng 2017; 3:3292-3306. [DOI: 10.1021/acsbiomaterials.7b00644] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Frances D. Liu
- Department
of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- BioSystems
and Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore−MIT Alliance for Research and Technology, CREATE, Singapore 138602
| | - Novalia Pishesha
- Department
of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Zhiyong Poon
- BioSystems
and Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore−MIT Alliance for Research and Technology, CREATE, Singapore 138602
| | - Tanwi Kaushik
- BioSystems
and Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore−MIT Alliance for Research and Technology, CREATE, Singapore 138602
| | - Krystyn J. Van Vliet
- Department
of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- BioSystems
and Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore−MIT Alliance for Research and Technology, CREATE, Singapore 138602
- Department
of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
52
|
Co-transplantation of mesenchymal and neural stem cells and overexpressing stromal-derived factor-1 for treating spinal cord injury. Brain Res 2017; 1672:91-105. [DOI: 10.1016/j.brainres.2017.07.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/12/2017] [Accepted: 07/05/2017] [Indexed: 12/17/2022]
|
53
|
Garba A, Acar DD, Roukaerts IDM, Desmarets LMB, Devriendt B, Nauwynck HJ. Long-term culture and differentiation of porcine red bone marrow hematopoietic cells co-cultured with immortalized mesenchymal cells. Vet Immunol Immunopathol 2017; 191:44-50. [PMID: 28895865 DOI: 10.1016/j.vetimm.2017.08.002] [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: 02/27/2017] [Revised: 07/24/2017] [Accepted: 08/03/2017] [Indexed: 12/01/2022]
Abstract
Mesenchymal cells are multipotent stromal cells with self-renewal, differentiation and immunomodulatory capabilities. We aimed to develop a co-culture model for differentiating hematopoietic cells on top of immortalized mesenchymal cells for studying interactions between hematopoietic and mesenchymal cells, useful for adequately exploring the therapeutic potential of mesenchymal cells. In this study, we investigated the survival, proliferation and differentiation of porcine red bone marrow hematopoietic cells co-cultured with immortalized porcine bone marrow mesenchymal cells for a period of five weeks. Directly after collection, primary porcine bone marrow mesenchymal cells adhered firmly to the bottom of the culture plates and showed a fibroblast-like appearance, one week after isolation. Upon immortalization, porcine bone marrow mesenchymal cells were continuously proliferating. They were positive for simian virus 40 (SV40) large T antigen and the mesenchymal cell markers CD44 and CD55. Isolated red bone marrow cells were added to these immortalized mesenchymal cells. Five weeks post-seeding, 92±6% of the red bone marrow hematopoietic cells were still alive and their number increased 3-fold during five weekly subpassages on top of the immortalized mesenchymal cells. The red bone marrow hematopoietic cells were originally small and round; later, the cells increased in size. Some of them became elongated, while others remained round. Tiny dendrites appeared attaching hematopoietic cells to the underlying immortalized mesenchymal cells. Furthermore, weekly differential-quick staining of the cells indicated the presence of monoblasts, monocytes, macrophages and lymphocytes in the co-cultures. At three weeks of co-culture, flow cytometry analysis showed an increased surface expression of CD172a, CD14, CD163, CD169, CD4 and CD8 up to 37±0.8%, 40±8%, 41±4%, 23±3% and 19±5% of the hematopoietic cells, respectively. In conclusion, continuous mesenchymal cell cultures were successfully established and characterized and they supported the proliferation of red bone marrow hematopoietic cells, which finally differentiated into monocytic cells and CD4+ and CD8+ cells.
Collapse
Affiliation(s)
- Abubakar Garba
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - Delphine D Acar
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - Inge D M Roukaerts
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - Lowiese M B Desmarets
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - Bert Devriendt
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - Hans J Nauwynck
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium.
| |
Collapse
|
54
|
Romanov YA, Volgina NE, Balashova EE, Kabaeva NV, Dugina TN, Sukhikh GT. Human Umbilical Cord Mesenchymal Stromal Cells Support Viability of Umbilical Cord Blood Hematopoietic Stem Cells but not the “Stemness” of Their Progeny in Co-Culture. Bull Exp Biol Med 2017; 163:523-527. [DOI: 10.1007/s10517-017-3843-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Indexed: 12/18/2022]
|
55
|
Mesenchymal Stem Cells in Myeloid Malignancies: A Focus on Immune Escaping and Therapeutic Implications. Stem Cells Int 2017; 2017:6720594. [PMID: 28947904 PMCID: PMC5602646 DOI: 10.1155/2017/6720594] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/06/2017] [Accepted: 07/20/2017] [Indexed: 01/07/2023] Open
Abstract
The importance of the bone marrow microenvironment forming the so-called niche in physiologic hemopoiesis is largely known, and recent evidences support the presence of stromal alterations from the molecular to the cytoarchitectural level in hematologic malignancies. Various alterations in cell adhesion, metabolism, cytokine signaling, autophagy, and methylation patterns of tumor-derived mesenchymal stem cells have been demonstrated, contributing to the genesis of a leukemic permissive niche. This niche allows both the ineffective haematopoiesis typical of myelodysplastic syndromes and the differentiation arrest, proliferation advantage, and clone selection which is the hallmark of acute myeloid leukemia. Furthermore, the immune system, both adaptive and innate, encompassing mesenchymal-derived cells, has been shown to take part to the leukemic niche. Here, we critically review the state of art about mesenchymal stem cell role in myelodysplastic syndromes and acute myeloid leukemia, focusing on immune escaping mechanisms as a target for available and future anticancer therapies.
Collapse
|
56
|
Futrega K, Atkinson K, Lott WB, Doran MR. Spheroid Coculture of Hematopoietic Stem/Progenitor Cells and Monolayer Expanded Mesenchymal Stem/Stromal Cells in Polydimethylsiloxane Microwells Modestly Improves In Vitro Hematopoietic Stem/Progenitor Cell Expansion. Tissue Eng Part C Methods 2017; 23:200-218. [PMID: 28406754 PMCID: PMC5397247 DOI: 10.1089/ten.tec.2016.0329] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
While two-dimensional (2D) monolayers of mesenchymal stem/stromal cells (MSCs) have been shown to enhance hematopoietic stem/progenitor cell (HSPC) expansion in vitro, expanded cells do not engraft long term in human recipients. This outcome is attributed to the failure of 2D culture to recapitulate the bone marrow (BM) niche signal milieu. Herein, we evaluated the capacity of a novel three-dimensional (3D) coculture system to support HSPC expansion in vitro. A high-throughput polydimethylsiloxane (PDMS) microwell platform was used to manufacture thousands of uniform 3D multicellular coculture spheroids. Relative gene expression in 3D spheroid versus 2D adherent BM-derived MSC cultures was characterized and compared with literature reports. We evaluated coculture spheroids, each containing 25-400 MSCs and 10 umbilical cord blood (CB)-derived CD34+ progenitor cells. At low exogenous cytokine concentrations, 2D and 3D MSC coculture modestly improved overall hematopoietic cell and CD34+ cell expansion outcomes. By contrast, a substantial increase in CD34+CD38- cell yield was observed in PDMS microwell cultures, regardless of the presence or absence of MSCs. This outcome indicated that CD34+CD38- cell culture yield could be increased using the microwell platform alone, even without MSC coculture support. We found that the increase in CD34+CD38- cell yield observed in PDMS microwell cultures did not translate to enhanced engraftment in NOD/SCID gamma (NSG) mice or a modification in the relative human hematopoietic lineages established in engrafted mice. In summary, there was no statistical difference in CD34+ cell yield from 2D or 3D cocultures, and MSC coculture support provided only modest benefit in either geometry. While the high-throughput 3D microwell platform may provide a useful model system for studying cells in coculture, further optimization will be required to generate HSPC yields suitable for use in clinical applications.
Collapse
Affiliation(s)
- Kathryn Futrega
- 1 Stem Cell Therapies Laboratory, Translational Research Institute, Queensland University of Technology , Brisbane, Australia
| | - Kerry Atkinson
- 1 Stem Cell Therapies Laboratory, Translational Research Institute, Queensland University of Technology , Brisbane, Australia
| | - William B Lott
- 1 Stem Cell Therapies Laboratory, Translational Research Institute, Queensland University of Technology , Brisbane, Australia
| | - Michael R Doran
- 1 Stem Cell Therapies Laboratory, Translational Research Institute, Queensland University of Technology , Brisbane, Australia .,2 Mater Research Institute - University of Queensland, Translational Research Institute , Brisbane, Australia
| |
Collapse
|
57
|
Pan X, Sun Q, Zhang Y, Cai H, Gao Y, Shen Y, Zhang W. Biomimetic Macroporous PCL Scaffolds for Ex Vivo Expansion of Cord Blood-Derived CD34 + Cells with Feeder Cells Support. Macromol Biosci 2017; 17. [PMID: 28544462 DOI: 10.1002/mabi.201700054] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/24/2017] [Indexed: 01/10/2023]
Abstract
Ex vivo expansion of hematopoietic stem cells (HSCs) with most current methods can hardly satisfy clinical application requirement. While in vivo, HSCs efficiently self-renew in niche where they interact with 3D extracellular matrix and stromal cells. Therefore, co-cultures of CD34+ cells and mesenchyme stem cells derived from human amniotic membrane (hAMSCs) on the basis of biomimetic macroporous three-dimensional (3D) poly(ε-caprolactone) (PCL) scaffolds are developed, where scaffolds and hAMSCs are applied to mimic structural and cellular microenvironment of HSCs. The influence of scaffolds, feeder cells, and contact manners on expansion and stemness maintenance of CD34+ cells is investigated in this protocol. Biomimetic scaffolds-dependent co-cultures of CD34+ cells and hAMSCs can effectively promote the expansion of CD34+ cells; meanwhile, indirect contact is superior to direct contact. The combination of biomimetic scaffolds and hAMSCs represents a new strategy for achieving clinical-scale ex vivo expansion of CD34+ cells.
Collapse
Affiliation(s)
- Xiuwei Pan
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Qiong Sun
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Yuanhao Zhang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Haibo Cai
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Yun Gao
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Yongjia Shen
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Weian Zhang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| |
Collapse
|
58
|
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]
|
59
|
Houshmand M, Soleimani M, Atashi A, Saglio G, Abdollahi M, Nikougoftar Zarif M. Mimicking the Acute Myeloid Leukemia Niche for Molecular Study and Drug Screening. Tissue Eng Part C Methods 2017; 23:72-85. [PMID: 28007011 DOI: 10.1089/ten.tec.2016.0404] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Bone marrow niche is a major contributing factor in leukemia development and drug resistance in acute myeloid leukemia (AML) patients. Although mimicking leukemic bone marrow niche relies on two-dimensional (2D) culture conditions, it cannot recapitulate complex bone marrow structure that causes introduction of different three-dimensional (3D) scaffolds. Simultaneously, microfluidic platform by perfusing medium culture mimic interstitial fluid flow, along with 3D scaffold would help for mimicking bone marrow microenvironment. In this study TF-1 cells were cocultured with bone marrow mesenchymal stem cells (BM-MSCs) in 2D and 3D microfluidic devices. Phenotype maintenance during cell culture and proliferation rate was assayed and confirmed by cell cycle analysis. Morphology of cells in 2D and 3D culture conditions was demonstrated by scanning electron microscopy. After these experiments, drug screening was performed by applying azacitidine and cytarabine and cytotoxicity assay and quantitative reverse transcription-polymerase chain reaction (qRT-PCR) for B cell lymphoma 2 (BCL2) were done to compare drug resistance in 2D and 3D culture conditions. Our result shows leukemic cells in 3D microfluidic device retaining their phenotype and proliferation rate was significantly higher in 3D culture condition in comparison to 2D culture condition (p < 0.05), which was confirmed by cell cycle analysis. Cytotoxicity assay also illustrated drug resistance in 3D culture condition and qRT-PCR demonstrated higher BCL2 expression in 3D microfluidic device in contrast to 2D microfluidic device (p < 0.05). On balance, mimicking bone marrow niche would help the target therapy and specify the role of niche in development of leukemia in AML patients.
Collapse
Affiliation(s)
- Mohammad Houshmand
- 1 Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine , Tehran, Iran
| | - Masoud Soleimani
- 2 Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University , Tehran, Iran
| | - Amir Atashi
- 3 Stem cell and Tissue Engineering Research Center, Shahroud University of Medical Sciences , Shahroud, Iran
| | - Giuseppe Saglio
- 4 Department of Clinical and Biological Sciences, "S. Luigi Gonzaga" Hospital, University of Turin , Orbassano, Italy
| | - Mohammad Abdollahi
- 2 Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University , Tehran, Iran
| | - Mahin Nikougoftar Zarif
- 1 Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine , Tehran, Iran
| |
Collapse
|
60
|
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]
|
61
|
Kovtonyuk LV, Fritsch K, Feng X, Manz MG, Takizawa H. Inflamm-Aging of Hematopoiesis, Hematopoietic Stem Cells, and the Bone Marrow Microenvironment. Front Immunol 2016; 7:502. [PMID: 27895645 PMCID: PMC5107568 DOI: 10.3389/fimmu.2016.00502] [Citation(s) in RCA: 257] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 10/27/2016] [Indexed: 12/19/2022] Open
Abstract
All hematopoietic and immune cells are continuously generated by hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) through highly organized process of stepwise lineage commitment. In the steady state, HSCs are mostly quiescent, while HPCs are actively proliferating and contributing to daily hematopoiesis. In response to hematopoietic challenges, e.g., life-threatening blood loss, infection, and inflammation, HSCs can be activated to proliferate and engage in blood formation. The HSC activation induced by hematopoietic demand is mediated by direct or indirect sensing mechanisms involving pattern recognition receptors or cytokine/chemokine receptors. In contrast to the hematopoietic challenges with obvious clinical symptoms, how the aging process, which involves low-grade chronic inflammation, impacts hematopoiesis remains undefined. Herein, we summarize recent findings pertaining to functional alternations of hematopoiesis, HSCs, and the bone marrow (BM) microenvironment during the processes of aging and inflammation and highlight some common cellular and molecular changes during the processes that influence hematopoiesis and its cells of origin, HSCs and HPCs, as well as the BM microenvironment. We also discuss how age-dependent alterations of the immune system lead to subclinical inflammatory states and how inflammatory signaling might be involved in hematopoietic aging. Our aim is to present evidence supporting the concept of “Inflamm-Aging,” or inflammation-associated aging of hematopoiesis.
Collapse
Affiliation(s)
- Larisa V Kovtonyuk
- Division of Hematology, University Hospital Zurich, University of Zurich , Zurich , Switzerland
| | - Kristin Fritsch
- Division of Hematology, University Hospital Zurich, University of Zurich , Zurich , Switzerland
| | - Xiaomin Feng
- International Research Center for Medical Sciences , Kumamoto , Japan
| | - Markus G Manz
- Division of Hematology, University Hospital Zurich, University of Zurich , Zurich , Switzerland
| | - Hitoshi Takizawa
- International Research Center for Medical Sciences , Kumamoto , Japan
| |
Collapse
|
62
|
Qu Q, Liu L, Chen G, Xu Y, Wu X, Wu D. Endothelial progenitor cells promote efficient ex vivo expansion of cord blood-derived hematopoietic stem/progenitor cells. Cytotherapy 2016; 18:452-64. [PMID: 26857234 DOI: 10.1016/j.jcyt.2015.12.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 12/27/2015] [Accepted: 12/30/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND AIMS Cord blood (CB) hematopoietic stem cell transplantation has often been limited by the scarcity of stem cells. Therefore, the number of CB hematopoietic stem/progenitor cells (HSPCs) should be increased while maintaining the stem cell characteristics. METHODS We designed an ex vivo culture system using endothelial progenitor cells (EPCs) as stroma to determine the capacity of expanding CB-HSPCs in a defined medium, the effect on engraftment of the expanded cells in a mouse model and the underlying mechanism. RESULTS After 7 days of culture, compared with those cultured with cytokines alone (3.25 ± 0.59), CD34+ cells under contact and non-contact co-culture with EPCs were expanded by 5.38 ± 0.61 (P = 0.003) and 4.06 ± 0.43 (P = 0.025)-fold, respectively. Direct cell-to-cell contact co-culture with EPCs resulted in more primitive CD34+ CD38- cells than stroma-free culture (156.17 ± 21.32 versus 79.12 ± 19.77-fold; P = 0.010). Comparable engraftment of day 7 co-cultured HSPCs with respect to HSPCs at day 0 in nonobese diabetic-severe combined immunodeficiency disease (NOD/SCID) mice was measured as a percentage of chimerism (13.3% ± 11.0% versus 16.0% ± 14.3%; P = 0.750). EPCs highly expressed interleukin 6 (IL6) and angiopoietin 1 (ANGPT1), the hematopoietic- related cytokines. A higher transcriptional level of WNT5A genes in EPCs and co-cultured HSPCs suggests that the activation of Wnt signaling pathway may play a role in HSPCs' expansion ex vivo. DISCUSSION These data demonstrated that EPCs improve the CD34+ population but do not compromise the repopulating efficacy of the amplified HSPCs, possibly via cytokine secretion and Wnt signaling pathway activation.
Collapse
Affiliation(s)
- Qi Qu
- Jiangsu Institute of Hematology, Suzhou Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Limin Liu
- Jiangsu Institute of Hematology, Suzhou Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Guanghua Chen
- Jiangsu Institute of Hematology, Suzhou Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Xu
- Jiangsu Institute of Hematology, Suzhou Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaojin Wu
- Jiangsu Institute of Hematology, Suzhou Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Depei Wu
- Jiangsu Institute of Hematology, Suzhou Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
63
|
Nanoparticle-mediated siRNA delivery assessed in a 3D co-culture model simulating prostate cancer bone metastasis. Int J Pharm 2016; 511:1058-69. [PMID: 27492023 DOI: 10.1016/j.ijpharm.2016.07.079] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 07/30/2016] [Indexed: 01/27/2023]
Abstract
siRNA has emerged as a potential therapeutic for the treatment of prostate cancer but effective delivery remains a major barrier to its clinical application. This study aimed to develop and characterise a 3D in vitro co-culture model to simulate prostate cancer bone metastasis and to assess the ability of the model to investigate nanoparticle-mediated siRNA delivery and gene knockdown. PC3 or LNCaP prostate cancer cells were co-cultured with hFOB 1.19 osteoblast cells in 2D on plastic tissue culture plates and in 3D on collagen scaffolds mimicking the bone microenvironment. To characterise the co-culture model, cell proliferation, enzyme secretion and the utility of two different gene delivery vectors to mediate siRNA uptake and gene knockdown were assessed. Cell proliferation was reduced by∼50% by day 7 in the co-culture system relative to monoculture (PC3 and LNCaP co-cultures, in 2D and 3D) and an enhanced level of MMP9 (a marker of bone metastasis) was secreted into the media (1.2-4-fold increase depending on the co-culture system). A cationic cyclodextrin gene delivery vector proved significantly less toxic in the co-culture system relative to the commercially available vector Lipofectamine 2000(®). In addition, knockdown of both the GAPDH gene (minimum 15%) and RelA subunit of the NF-κB transcription factor (minimum 20%) was achieved in 2D and 3D cell co-cultures. Results indicate that the prostate cancer-osteoblast in vitro co-culture model was more physiologically relevant vs the monoculture. This model has the potential to help improve the design and efficacy of gene delivery formulations, to more accurately predict in vivo performance and, therefore, to reduce the risk of product failure in late-stage clinical development.
Collapse
|
64
|
Pleyer L, Valent P, Greil R. Mesenchymal Stem and Progenitor Cells in Normal and Dysplastic Hematopoiesis-Masters of Survival and Clonality? Int J Mol Sci 2016; 17:ijms17071009. [PMID: 27355944 PMCID: PMC4964385 DOI: 10.3390/ijms17071009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 05/20/2016] [Accepted: 06/08/2016] [Indexed: 02/07/2023] Open
Abstract
Myelodysplastic syndromes (MDS) are malignant hematopoietic stem cell disorders that have the capacity to progress to acute myeloid leukemia (AML). Accumulating evidence suggests that the altered bone marrow (BM) microenvironment in general, and in particular the components of the stem cell niche, including mesenchymal stem cells (MSCs) and their progeny, play a pivotal role in the evolution and propagation of MDS. We here present an overview of the role of MSCs in the pathogenesis of MDS, with emphasis on cellular interactions in the BM microenvironment and related stem cell niche concepts. MSCs have potent immunomodulatory capacities and communicate with diverse immune cells, but also interact with various other cellular components of the microenvironment as well as with normal and leukemic stem and progenitor cells. Moreover, compared to normal MSCs, MSCs in MDS and AML often exhibit altered gene expression profiles, an aberrant phenotype, and abnormal functional properties. These alterations supposedly contribute to the “reprogramming” of the stem cell niche into a disease-permissive microenvironment where an altered immune system, abnormal stem cell niche interactions, and an impaired growth control lead to disease progression. The current article also reviews molecular targets that play a role in such cellular interactions and possibilities to interfere with abnormal stem cell niche interactions by using specific targeted drugs.
Collapse
Affiliation(s)
- Lisa Pleyer
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria.
- Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute, 5020 Salzburg, Austria.
- 3rd Medical Department, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology & Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Richard Greil
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria.
- Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute, 5020 Salzburg, Austria.
- 3rd Medical Department, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| |
Collapse
|
65
|
Vasko T, Frobel J, Lubberich R, Goecke TW, Wagner W. iPSC-derived mesenchymal stromal cells are less supportive than primary MSCs for co-culture of hematopoietic progenitor cells. J Hematol Oncol 2016; 9:43. [PMID: 27098268 PMCID: PMC4839158 DOI: 10.1186/s13045-016-0273-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 04/13/2016] [Indexed: 02/11/2023] Open
Abstract
In vitro culture of hematopoietic stem and progenitor cells (HPCs) is supported by a suitable cellular microenvironment, such as mesenchymal stromal cells (MSCs)-but MSCs are heterogeneous and poorly defined. In this study, we analyzed whether MSCs derived from induced pluripotent stem cells (iPS-MSCs) provide a suitable cellular feeder layer too. iPS-MSCs clearly supported proliferation of HPCs, maintenance of a primitive immunophenotype (CD34(+), CD133(+), CD38(-)), and colony-forming unit (CFU) potential of CD34(+) HPCs. However, particularly long-term culture-initiating cell (LTC-IC) frequency was lower with iPS-MSCs as compared to primary MSCs. Relevant genes for cell-cell interaction were overall expressed at similar level in MSCs and iPS-MSCs, whereas VCAM1 was less expressed in the latter. In conclusion, our iPS-MSCs support in vitro culture of HPCs; however, under the current differentiation and culture conditions, they are less suitable than primary MSCs from bone marrow.
Collapse
Affiliation(s)
- Theresa Vasko
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Pauwelsstrasse 20, 52074, Aachen, Germany.,Institute for Biomedical Engineering-Cell Biology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Joana Frobel
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Pauwelsstrasse 20, 52074, Aachen, Germany.,Institute for Biomedical Engineering-Cell Biology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Richard Lubberich
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Pauwelsstrasse 20, 52074, Aachen, Germany.,Institute for Biomedical Engineering-Cell Biology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Tamme W Goecke
- Department of Obstetrics and Gynecology, RWTH Aachen University Hospital, Aachen, Germany
| | - Wolfgang Wagner
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Pauwelsstrasse 20, 52074, Aachen, Germany. .,Institute for Biomedical Engineering-Cell Biology, University Hospital of RWTH Aachen, Aachen, Germany.
| |
Collapse
|
66
|
A Three-Dimensional Scaffold-Based System for Modeling the Bone Marrow Tissue. Stem Cells Dev 2016; 25:492-8. [DOI: 10.1089/scd.2015.0182] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
67
|
Wuchter P, Vetter M, Saffrich R, Diehlmann A, Bieback K, Ho AD, Horn P. Evaluation of GMP-compliant culture media for in vitro expansion of human bone marrow mesenchymal stromal cells. Exp Hematol 2016; 44:508-18. [PMID: 26911671 DOI: 10.1016/j.exphem.2016.02.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 01/31/2016] [Accepted: 02/10/2016] [Indexed: 12/31/2022]
Abstract
Mesenchymal stromal cells (MSCs) from human bone marrow serve as a resource for cell-based therapies in regenerative medicine. Clinical applications require standardized protocols according to good manufacturing practice (GMP) guidelines. Donor variability as well as the intrinsic heterogeneity of MSC populations must be taken into consideration. The composition of the culture medium is a key factor in successful MSC expansion. The aim of this study was to comparatively assess the efficiency of xeno-free human platelet lysate (HPL)-based cell expansion with two commercially available media-StemPro MSC SFM CTS (for human ex vivo tissue and cell culture processing applications) and MSCGM (non-GMP-compliant, for research only)-in an academic setting as the first optimization step toward GMP-compliant manufacturing. We report the feasibility of MSC expansion up to the yielded cell number with all three media. MSCs exhibited the typical fibroblastoid morphology, with distinct differences in cell size depending on the medium. The differentiation capacity and characteristic immunophenotype were confirmed for all MSC populations. Proliferation was highest using StemPro MSC SFM CTS, whereas HPL medium was more cost-effective and its composition could be adjusted individually according to the respective needs. In summary, we present a comprehensive evaluation of GMP-compatible culture media for MSC expansion. Both StemPro and HPL medium proved to be suitable for clinical application and allowed sufficient cell proliferation. Specific differences were observed and should be considered according to the intended use. This study provides a detailed cost analysis and tools that may be helpful for the establishment of GMP-compliant MSC expansion.
Collapse
Affiliation(s)
- Patrick Wuchter
- Department of Medicine V, Heidelberg University, Heidelberg, Germany.
| | - Marcel Vetter
- Department of Medicine V, Heidelberg University, Heidelberg, Germany
| | - Rainer Saffrich
- Department of Medicine V, Heidelberg University, Heidelberg, Germany
| | - Anke Diehlmann
- Department of Medicine V, Heidelberg University, Heidelberg, Germany
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Anthony D Ho
- Department of Medicine V, Heidelberg University, Heidelberg, Germany
| | - Patrick Horn
- Department of Medicine V, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
68
|
Wuchter P, Saffrich R, Giselbrecht S, Nies C, Lorig H, Kolb S, Ho AD, Gottwald E. Microcavity arrays as an in vitro model system of the bone marrow niche for hematopoietic stem cells. Cell Tissue Res 2016; 364:573-584. [PMID: 26829941 DOI: 10.1007/s00441-015-2348-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 12/10/2015] [Indexed: 12/28/2022]
Abstract
In previous studies human mesenchymal stromal cells (MSCs) maintained the "stemness" of human hematopoietic progenitor cells (HPCs) through direct cell-cell contact in two-dimensional co-culture systems. We establish a three-dimensional (3D) co-culture system based on a custom-made chip, the 3(D)-KITChip, as an in vitro model system of the human hematopoietic stem cell niche. This array of up to 625 microcavities, with 300 μm size in each orientation, was inserted into a microfluidic bioreactor. The microcavities of the 3(D)-KITChip were inoculated with human bone marrow MSCs together with umbilical cord blood HPCs. MSCs used the microcavities as a scaffold to build a complex 3D mesh. HPCs were distributed three-dimensionally inside this MSC network and formed ß-catenin- and N-cadherin-based intercellular junctions to the surrounding MSCs. Using RT(2)-PCR and western blots, we demonstrate that a proportion of HPCs maintained the expression of CD34 throughout a culture period of 14 days. In colony-forming unit assays, the hematopoietic stem cell plasticity remained similar after 14 days of bioreactor co-culture, whereas monolayer co-cultures showed increasing signs of HPC differentiation and loss of stemness. These data support the notion that the 3D microenvironment created within the microcavity array preserves vital stem cell functions of HPCs more efficiently than conventional co-culture systems.
Collapse
Affiliation(s)
- Patrick Wuchter
- Department of Medicine V, Heidelberg University, 69120, Heidelberg, Germany. .,HEiKA - Heidelberg Karlsruhe Research Partnership, Heidelberg University and Karlsruhe Institute of Technology, Heidelberg and Karlsruhe, Germany.
| | - Rainer Saffrich
- Department of Medicine V, Heidelberg University, 69120, Heidelberg, Germany.,HEiKA - Heidelberg Karlsruhe Research Partnership, Heidelberg University and Karlsruhe Institute of Technology, Heidelberg and Karlsruhe, Germany
| | - Stefan Giselbrecht
- HEiKA - Heidelberg Karlsruhe Research Partnership, Heidelberg University and Karlsruhe Institute of Technology, Heidelberg and Karlsruhe, Germany.,Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Cordula Nies
- Institute for Biological Interfaces-5, Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Karlsruhe, Germany.,HEiKA - Heidelberg Karlsruhe Research Partnership, Heidelberg University and Karlsruhe Institute of Technology, Heidelberg and Karlsruhe, Germany
| | - Hanna Lorig
- Institute for Biological Interfaces-5, Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Karlsruhe, Germany.,HEiKA - Heidelberg Karlsruhe Research Partnership, Heidelberg University and Karlsruhe Institute of Technology, Heidelberg and Karlsruhe, Germany
| | - Stephanie Kolb
- Institute for Biological Interfaces-5, Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Karlsruhe, Germany.,HEiKA - Heidelberg Karlsruhe Research Partnership, Heidelberg University and Karlsruhe Institute of Technology, Heidelberg and Karlsruhe, Germany
| | - Anthony D Ho
- Department of Medicine V, Heidelberg University, 69120, Heidelberg, Germany.,HEiKA - Heidelberg Karlsruhe Research Partnership, Heidelberg University and Karlsruhe Institute of Technology, Heidelberg and Karlsruhe, Germany
| | - Eric Gottwald
- Institute for Biological Interfaces-5, Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Karlsruhe, Germany. .,HEiKA - Heidelberg Karlsruhe Research Partnership, Heidelberg University and Karlsruhe Institute of Technology, Heidelberg and Karlsruhe, Germany.
| |
Collapse
|
69
|
Pan X, Sun Q, Cai H, Gao Y, Tan W, Zhang W. Encapsulated feeder cells within alginate beads for ex vivo expansion of cord blood-derived CD34+ cells. Biomater Sci 2016; 4:1441-53. [DOI: 10.1039/c6bm00191b] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A co-culture system based on encapsulated feeder cells within alginate beads was developed through optimizing the detailed aspects of the cell culture system to expand CD34-positive (CD34+) cells ex vivo.
Collapse
Affiliation(s)
- Xiuwei Pan
- State Key Laboratory of Bioreactor Engineering
- Shanghai Key Laboratory of Functional Materials Chemistry
- School of Materials Science and Engineering
- East China University of Science and Technology
- Shanghai 200237
| | - Qiong Sun
- State Key Laboratory of Bioreactor Engineering
- Shanghai Key Laboratory of Functional Materials Chemistry
- School of Materials Science and Engineering
- East China University of Science and Technology
- Shanghai 200237
| | - Haibo Cai
- State Key Laboratory of Bioreactor Engineering
- Shanghai Key Laboratory of Functional Materials Chemistry
- School of Materials Science and Engineering
- East China University of Science and Technology
- Shanghai 200237
| | - Yun Gao
- State Key Laboratory of Bioreactor Engineering
- Shanghai Key Laboratory of Functional Materials Chemistry
- School of Materials Science and Engineering
- East China University of Science and Technology
- Shanghai 200237
| | - Wensong Tan
- State Key Laboratory of Bioreactor Engineering
- Shanghai Key Laboratory of Functional Materials Chemistry
- School of Materials Science and Engineering
- East China University of Science and Technology
- Shanghai 200237
| | - Weian Zhang
- State Key Laboratory of Bioreactor Engineering
- Shanghai Key Laboratory of Functional Materials Chemistry
- School of Materials Science and Engineering
- East China University of Science and Technology
- Shanghai 200237
| |
Collapse
|
70
|
Hettiaratchi MH, Guldberg RE, McDevitt TC. Biomaterial strategies for controlling stem cell fate via morphogen sequestration. J Mater Chem B 2016; 4:3464-3481. [DOI: 10.1039/c5tb02575c] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
This review explores the role of protein sequestration in the stem cell niche and how it has inspired the design of biomaterials that exploit natural protein sequestration to influence stem cell fate.
Collapse
Affiliation(s)
- M. H. Hettiaratchi
- The Parker H. Petit Institute for Bioengineering and Bioscience
- Georgia Institute of Technology
- Atlanta
- USA
- The Wallace H. Coulter Department of Biomedical Engineering
| | - R. E. Guldberg
- The Parker H. Petit Institute for Bioengineering and Bioscience
- Georgia Institute of Technology
- Atlanta
- USA
- The George W. Woodruff School of Mechanical Engineering
| | - T. C. McDevitt
- The Gladstone Institute of Cardiovascular Disease
- San Francisco
- USA
- The Department of Bioengineering and Therapeutic Sciences
- University of California San Francisco
| |
Collapse
|
71
|
Ziegler P, Boettcher S, Takizawa H, Manz MG, Brümmendorf TH. LPS-stimulated human bone marrow stroma cells support myeloid cell development and progenitor cell maintenance. Ann Hematol 2015; 95:173-8. [PMID: 26555286 DOI: 10.1007/s00277-015-2550-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 11/04/2015] [Indexed: 01/22/2023]
Abstract
The nonhematopoietic bone marrow (BM) microenvironment provides a functional niche for hematopoietic cell maintenance, recruitment, and differentiation. It consists of multiple cell types including vasculature, bone, adipose tissue, and fibroblast-like bone marrow stromal cells (BMSC), which can be summarized under the generic term niche cells. BMSC express Toll-like receptors (TLRs) and are capable to respond to TLR-agonists by changing their cytokine expression pattern in order to more efficiently support hematopoiesis. Here, we show that in addition to enhanced myeloid colony formation from human CD34+ cells, lipopolysaccharide (LPS) stimulation retains overall higher numbers of CD34+ cells in co-culture assays using BMSC, with eightfold more CD34+ cells that underwent up to three divisions as compared to non-stimulated assays. When subjected to cytokine-supplemented myeloid colony-forming unit (CFU) assays or transplanted into newborn RAG2(-/-) γc (-/-) mice, CD34(+) cells from LPS-stimulated BMSC cultures give rise to the full spectrum of myeloid colonies and T and B cells, respectively, thus supporting maintenance of myeloid and lymphoid primed hematopoietic progenitor cells (HPCs) under inflammatory conditions. Collectively, we suggest that BMSC enhance hematopoiesis during inflammatory conditions to support the replenishment of innate immune effector cells and to prevent the exhaustion of the hematopoietic stem and progenitor cell (HSPC) pool.
Collapse
Affiliation(s)
- Patrick Ziegler
- Institute for Occupational and Social Medicine, RWTH Aachen University, Aachen, Germany.
- Institute for Research in Biomedicine (IRB), Via Vincenzo Vela 6, 6500, Bellinzona, Switzerland.
| | - Steffen Boettcher
- Institute for Research in Biomedicine (IRB), Via Vincenzo Vela 6, 6500, Bellinzona, Switzerland
- Division of Hematology, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
| | - Hitoshi Takizawa
- Institute for Research in Biomedicine (IRB), Via Vincenzo Vela 6, 6500, Bellinzona, Switzerland
- Division of Hematology, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
| | - Markus G Manz
- Institute for Research in Biomedicine (IRB), Via Vincenzo Vela 6, 6500, Bellinzona, Switzerland
- Division of Hematology, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
| | - Tim H Brümmendorf
- Department of Oncology, Hematology, Hemostaseology and Stem cell transplantation, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| |
Collapse
|
72
|
Reichert D, Friedrichs J, Ritter S, Käubler T, Werner C, Bornhäuser M, Corbeil D. Phenotypic, Morphological and Adhesive Differences of Human Hematopoietic Progenitor Cells Cultured on Murine versus Human Mesenchymal Stromal Cells. Sci Rep 2015; 5:15680. [PMID: 26498381 PMCID: PMC4620509 DOI: 10.1038/srep15680] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 10/01/2015] [Indexed: 02/07/2023] Open
Abstract
Xenogenic transplantation models have been developed to study human hematopoiesis in immunocompromised murine recipients. They still have limitations and therefore it is important to delineate all players within the bone marrow that could account for species-specific differences. Here, we evaluated the proliferative capacity, morphological and physical characteristics of human CD34+ hematopoietic stem and progenitor cells (HSPCs) after co-culture on murine or human bone marrow-derived mesenchymal stromal cells (MSCs). After seven days, human CD34+CD133– HSPCs expanded to similar extents on both feeder layers while cellular subsets comprising primitive CD34+CD133+ and CD133+CD34– phenotypes are reduced fivefold on murine MSCs. The number of migrating HSPCs was also reduced on murine cells suggesting that MSC adhesion influences cellular polarization of HSPC. We used atomic force microscopy-based single-cell force spectroscopy to quantify their adhesive interactions. We found threefold higher detachment forces of human HSPCs from murine MSCs compared to human ones. This difference is related to the N-cadherin expression level on murine MSCs since its knockdown abolished their differential adhesion properties with human HSPCs. Our observations highlight phenotypic, morphological and adhesive differences of human HSPCs when cultured on murine or human MSCs, which raise some caution in data interpretation when xenogenic transplantation models are used.
Collapse
Affiliation(s)
- Doreen Reichert
- Tissue Engineering Laboratories (BIOTEC), Technische Universität Dresden, 01307 Dresden, Germany
| | - Jens Friedrichs
- Institute for Biofunctional Polymer Materials, Leibniz Institute of Polymer Research Dresden, 01069 Dresden, Germany
| | - Steffi Ritter
- Tissue Engineering Laboratories (BIOTEC), Technische Universität Dresden, 01307 Dresden, Germany
| | - Theresa Käubler
- Tissue Engineering Laboratories (BIOTEC), Technische Universität Dresden, 01307 Dresden, Germany
| | - Carsten Werner
- Institute for Biofunctional Polymer Materials, Leibniz Institute of Polymer Research Dresden, 01069 Dresden, Germany.,DFG Research Center and Cluster of Excellence for Regenerative Therapies Dresden 01307 Dresden, Germany
| | - Martin Bornhäuser
- Medical Clinic and Polyclinic I, University Hospital Carl Gustav Carus, 01307 Dresden, Germany.,DFG Research Center and Cluster of Excellence for Regenerative Therapies Dresden 01307 Dresden, Germany
| | - Denis Corbeil
- Tissue Engineering Laboratories (BIOTEC), Technische Universität Dresden, 01307 Dresden, Germany.,DFG Research Center and Cluster of Excellence for Regenerative Therapies Dresden 01307 Dresden, Germany
| |
Collapse
|
73
|
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
|
74
|
Choi JS, Mahadik BP, Harley BAC. Engineering the hematopoietic stem cell niche: Frontiers in biomaterial science. Biotechnol J 2015; 10:1529-45. [PMID: 26356030 PMCID: PMC4724421 DOI: 10.1002/biot.201400758] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 06/15/2015] [Accepted: 07/16/2015] [Indexed: 12/13/2022]
Abstract
Hematopoietic stem cells (HSCs) play a crucial role in the generation of the body's blood and immune cells. This process takes place primarily in the bone marrow in specialized 'niche' microenvironments, which provide signals responsible for maintaining a balance between HSC quiescence, self-renewal, and lineage specification required for life-long hematopoiesis. While our understanding of these signaling mechanisms continues to improve, our ability to engineer them in vitro for the expansion of clinically relevant HSC populations is still lacking. In this review, we focus on development of biomaterials-based culture platforms for in vitro study of interactions between HSCs and their local microenvironment. The tools and techniques used for both examining HSC-niche interactions as well as applying these findings towards controlled HSC expansion or directed differentiation in 2D and 3D platforms are discussed. These novel techniques hold the potential to push the existing boundaries of HSC cultures towards high-throughput, real-time, and single-cell level biomimetic approaches that enable a more nuanced understanding of HSC regulation and function. Their application in conjunction with innovative biomaterial platforms can pave the way for engineering artificial bone marrow niches for clinical applications as well as elucidating the pathology of blood-related cancers and disorders.
Collapse
Affiliation(s)
- Ji Sun Choi
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Bhushan P Mahadik
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Brendan A C Harley
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
75
|
Saleh M, Shamsasanjan K, Movassaghpourakbari A, Akbarzadehlaleh P, Molaeipour Z. The Impact of Mesenchymal Stem Cells on Differentiation of Hematopoietic Stem Cells. Adv Pharm Bull 2015; 5:299-304. [PMID: 26504750 DOI: 10.15171/apb.2015.042] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 04/15/2015] [Accepted: 04/18/2015] [Indexed: 12/30/2022] Open
Abstract
Bone marrow microenvironment contains cellular and acellular compartments. The cellular compartment includes hematopoietic stem cells, mesenchymal stem cells and some other stromal cell types, while the acellular compartment is composed of scaffold proteins known as the extra cellular matrix. Direct cell-cell contact as well as cytokines secreted by mesenchymal stem cells during coculture of hematopoietic stem cells and mesenchymal stem cells play a critical role in hematopoiesis, and determines the fate of hematopoietic stem cells. Several studies have demonstrated the impact of mesenchymal stem cells on self-renewal, expansion, proliferation and differentiation of hematopoietic stem cells in vitro, which have shown different and contradictory results. In this paper, we will investigate the effect of mesenchymal stem cells on differentiation of hematopoietic stem cells in vitro.
Collapse
Affiliation(s)
- Mahshid Saleh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasanjan
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. ; 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
| | - Zahra Molaeipour
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
76
|
Cheng HC, Liu SW, Li W, Zhao XF, Zhao X, Cheng M, Qiu L, Ma J. Arsenic trioxide regulates adipogenic and osteogenic differentiation in bone marrow MSCs of aplastic anemia patients through BMP4 gene. Acta Biochim Biophys Sin (Shanghai) 2015. [PMID: 26215597 DOI: 10.1093/abbs/gmv065] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The typical pathological feature of aplastic anemia (AA) is the rise in the number of fat cells and the reduction of osteoblasts in bone marrow. However, both fat cells and osteobalsts in bone marrow are derived from the mesenchymal stem cells (MSCs). Generally, the adipogenic and osteogenic differentiation is a dynamic and balanceable process. The imbalance of the adipogenic and osteogenic differentiation may participate in the occurrence and progress of many diseases. Arsenic trioxide (ATO) could induce differentiation and apoptosis in tumor cells. In this study, Oil Red-O and Alizarin red were used to detect the adipogenic and osteogenic differentiation. The ability of adipogenic differentiation is much higher, whereas the osteogenic differentiation is much lower in the MSCs of AA patients compared with healthy controls. ATO inhibits adipogenic differentiation and promotes osteogenic differentiation in the MSC of AA patients. The expression of BMP4 is increased with ATO treatment. The ability of adipogenic differentiation is decreased, whereas the osteogenic differentiation is increased after transfection of BMP4 gene into the MSCs of AA patients. This study shows that ATO regulates the adipogenic and osteogenic differentiation balance of MSCs in AA, which provides a theoretical basis for the adjunctive therapy of ATO on AA. The BMP4 gene is involved in the ATO regulation of adipogenic and osteogenic differentiation balance, which provides a new target for the treatment of AA.
Collapse
Affiliation(s)
- Huan Chen Cheng
- Institute of Hematology and Oncology, the First Hospital of Harbin, Harbin 150010, China
| | - Sheng Wei Liu
- Institute of Hematology and Oncology, the First Hospital of Harbin, Harbin 150010, China
| | - Wei Li
- Institute of Hematology and Oncology, the First Hospital of Harbin, Harbin 150010, China
| | - Xue Fei Zhao
- Institute of Hematology and Oncology, the First Hospital of Harbin, Harbin 150010, China
| | - Xu Zhao
- Institute of Hematology and Oncology, the First Hospital of Harbin, Harbin 150010, China
| | - Mei Cheng
- Institute of Hematology and Oncology, the First Hospital of Harbin, Harbin 150010, China
| | - Lin Qiu
- Institute of Hematology and Oncology, the First Hospital of Harbin, Harbin 150010, China
| | - Jun Ma
- Institute of Hematology and Oncology, the First Hospital of Harbin, Harbin 150010, China
| |
Collapse
|
77
|
Walenda T, Diener Y, Jost E, Morin-Kensicki E, Goecke TW, Bosio A, Rath B, Brümmendorf TH, Bissels U, Wagner W. MicroRNAs and Metabolites in Serum Change after Chemotherapy: Impact on Hematopoietic Stem and Progenitor Cells. PLoS One 2015; 10:e0128231. [PMID: 26024523 PMCID: PMC4449031 DOI: 10.1371/journal.pone.0128231] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 04/24/2015] [Indexed: 11/21/2022] Open
Abstract
Hematopoietic regeneration after high dose chemotherapy necessitates activation of the stem cell pool. There is evidence that serum taken after chemotherapy comprises factors stimulating proliferation and self-renewal of CD34+ hematopoietic stem and progenitor cells (HSPCs) – however, the nature of these feedback signals is yet unclear. Here, we addressed the question if specific microRNAs (miRNAs) or metabolites are affected after high dose chemotherapy. Serum taken from the same patients before and after chemotherapy was supplemented for in vitro cultivation of HSPCs. Serum taken after chemotherapy significantly enhanced HSPC proliferation, better maintained a CD34+ immunophenotype, and stimulated colony forming units. Microarray analysis revealed that 23 miRNAs changed in serum after chemotherapy – particularly, miRNA-320c and miRNA-1275 were down-regulated whereas miRNA-3663-3p was up-regulated. miRNA-320c was exemplarily inhibited by an antagomiR, which seemed to increase proliferation. Metabolomic profiling demonstrated that 44 metabolites were less abundant, whereas three (including 2-hydroxybutyrate and taurocholenate sulphate) increased in serum upon chemotherapy. Nine of these metabolites were subsequently tested for effects on HSPCs in vitro, but none of them exerted a clear concentration dependent effect on proliferation, immunophenotype and colony forming unit formation. Taken together, serum profiles of miRNAs and metabolites changed after chemotherapy. Rather than individually, these factors may act in concert to recruit HSPCs into action for hematopoietic regeneration.
Collapse
Affiliation(s)
- Thomas Walenda
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University Medical School, Aachen, Germany
| | | | - Edgar Jost
- Department for Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, RWTH Aachen University Medical School, Aachen, Germany
| | | | - Tamme W. Goecke
- Department of Obstetrics and Gynecology, RWTH Aachen University Medical School, Aachen, German
| | | | - Björn Rath
- Department for Orthopedics, RWTH Aachen University Medical School, Aachen, Germany
| | - Tim H. Brümmendorf
- Department for Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, RWTH Aachen University Medical School, Aachen, Germany
| | - Ute Bissels
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | - Wolfgang Wagner
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University Medical School, Aachen, Germany
- * E-mail:
| |
Collapse
|
78
|
Yong KSM, Keng CT, Tan SQ, Loh E, Chang KT, Tan TC, Hong W, Chen Q. Human CD34(lo)CD133(lo) fetal liver cells support the expansion of human CD34(hi)CD133(hi) hematopoietic stem cells. Cell Mol Immunol 2015; 13:605-14. [PMID: 27593483 DOI: 10.1038/cmi.2015.40] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 03/30/2015] [Accepted: 04/18/2015] [Indexed: 12/18/2022] Open
Abstract
We have recently discovered a unique CD34(lo)CD133(lo) cell population in the human fetal liver (FL) that gives rise to cells in the hepatic lineage. In this study, we further characterized the biological functions of FL CD34(lo)CD133(lo) cells. Our findings show that these CD34(lo)CD133(lo) cells express markers of both endodermal and mesodermal lineages and have the capability to differentiate into hepatocyte and mesenchymal lineage cells by ex vivo differentiation assays. Furthermore, we show that CD34(lo)CD133(lo) cells express growth factors that are important for human hematopoietic stem cell (HSC) expansion: stem cell factor (SCF), insulin-like growth factor 2 (IGF2), C-X-C motif chemokine 12 (CXCL12), and factors in the angiopoietin-like protein family. Co-culture of autologous FL HSCs and allogenic HSCs derived from cord blood with CD34(lo)CD133(lo) cells supports and expands both types of HSCs.These findings are not only essential for extending our understanding of the HSC niche during the development of embryonic and fetal hematopoiesis but will also potentially benefit adult stem cell transplantations in clinics because expanded HSCs demonstrate the same capacity as primary cells to reconstitute the human immune system and mediate long-term hematopoiesis in vivo. Together, CD34(lo)CD133(lo) cells not only serve as stem/progenitor cells for liver development but are also an essential component of the HSC niche in the human FL.
Collapse
Affiliation(s)
- Kylie Su Mei Yong
- Humanized Mouse Unit, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore
| | - Choong Tat Keng
- Humanized Mouse Unit, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore
| | - Shu Qi Tan
- Department of Obstetrics & Gynaecology, KK Women's and Children's Hospital, Singapore 229899, Singapore
| | - Eva Loh
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore
| | - Kenneth Te Chang
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore.,Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Thiam Chye Tan
- Department of Obstetrics & Gynaecology, KK Women's and Children's Hospital, Singapore 229899, Singapore.,Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Wanjin Hong
- Humanized Mouse Unit, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore
| | - Qingfeng Chen
- Humanized Mouse Unit, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore.,Interdisciplinary Research Group in Infectious Diseases, Singapore-Massachusetts Institute of Technology Alliance for Research and Technology, Singapore 138602, Singapore.,Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| |
Collapse
|
79
|
Vertès AA. The potential of cytotherapeutics in hematologic reconstitution and in the treatment and prophylaxis of graft-versus-host disease. Chapter II: emerging transformational cytotherapies. Regen Med 2015; 10:345-73. [DOI: 10.2217/rme.15.13] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is a life-saving treatment for inherited anemias, immunodeficiencies or hematologic malignancies. A major complication of allo-HSCT associated with high transplant-related mortality rates is graft-versus-host disease (GvHD). Current and future clinical benefits in HSCT enabled by advances in hematopoietic stem cells, mesenchymal stem cells, Tregs and natural killer cells technologies are reviewed here and discussed. Among these evolutions, based on the need for mesenchymal stem cells to be recruited by an inflammatory environment, the development and use of novel GvHD biomarkers could be explored further to deliver the right pharmaceutical to the right patient at the right time. The successful commercialization of cytotherapeutics to efficiently manage GvHD will create a virtuous ‘halo’ effect for regenerative medicine.
Collapse
Affiliation(s)
- Alain A Vertès
- Sloan Fellow, London Business School, London, UK
- NxR Biotechnologies GmbH, Basel, Switzerland
| |
Collapse
|
80
|
Ren J, Wang H, Tran K, Civini S, Jin P, Castiello L, Feng J, Kuznetsov SA, Robey PG, Sabatino M, Stroncek DF. Human bone marrow stromal cell confluence: effects on cell characteristics and methods of assessment. Cytotherapy 2015; 17:897-911. [PMID: 25882666 DOI: 10.1016/j.jcyt.2015.03.607] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 03/09/2015] [Indexed: 01/05/2023]
Abstract
BACKGROUND AIMS Ex vivo expansion and serial passage of human bone marrow stromal cells (BMSCs, also known as bone marrow-derived mesenchymal stem cells) is required to obtain sufficient quantities for clinical therapy. The BMSC confluence criteria used to determine passage and harvest timing vary widely, and the impact of confluence on BMSC properties remains controversial. The effects of confluence on BMSC properties were studied and confluence-associated markers were identified. METHODS BMSC characteristics were analyzed as they grew from 50% to 100% confluence, including viability, population doubling time, apoptosis, colony formation, immunosuppression, surface marker expression, global gene expression and microRNA expression. In addition, culture supernatant protein, glucose, lactate and pH levels were analyzed. RESULTS Confluence-dependent changes were detected in the expression of several cell surface markers: 39 culture supernatant proteins, 26 microRNAs and 2078 genes. Many of these surface markers, proteins, microRNAs and genes have been reported to be important in BMSC function. The pigment epithelium-derived factor/vascular endothelial growth factor ratio increased with confluence, but 80% and 100% confluent BMSCs demonstrated a similar level of immunosuppression of mixed lymphocyte reactions. In addition, changes in lactate and glucose levels correlated with BMSC density. CONCLUSIONS BMSC characteristics change as confluence increases. 100% confluent BMSCs may have compromised pro-angiogenesis properties but may retain their immunomodulatory properties. Supernatant lactate and glucose levels can be used to estimate confluence and ensure consistency in passage and harvest timing. Flow cytometry or microRNA expression can be used to confirm that the BMSCs have been harvested at the appropriate confluence.
Collapse
Affiliation(s)
- Jiaqiang Ren
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Huan Wang
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Katherine Tran
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Sara Civini
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Ping Jin
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Luciano Castiello
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Ji Feng
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Sergei A Kuznetsov
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Pamela G Robey
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Marianna Sabatino
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - David F Stroncek
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
81
|
Burk AS, Monzel C, Yoshikawa HY, Wuchter P, Saffrich R, Eckstein V, Tanaka M, Ho AD. Quantifying adhesion mechanisms and dynamics of human hematopoietic stem and progenitor cells. Sci Rep 2015; 5:9370. [PMID: 25824493 PMCID: PMC5380331 DOI: 10.1038/srep09370] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 03/02/2015] [Indexed: 11/22/2022] Open
Abstract
Using planar lipid membranes with precisely defined concentrations of specific ligands, we have determined the binding strength between human hematopoietic stem cells (HSC) and the bone marrow niche. The relative significance of HSC adhesion to the surrogate niche models via SDF1α-CXCR4 or N-cadherin axes was quantified by (a) the fraction of adherent cells, (b) the area of tight adhesion, and (c) the critical pressure for cell detachment. We have demonstrated that the binding of HSC to the niche model is a cooperative process, and the adhesion mediated by the CXCR4- SDF1α axis is stronger than that by homophilic N-cadherin binding. The statistical image analysis of stochastic morphological dynamics unraveled that HSC dissipated energy by undergoing oscillatory deformation. The combination of an in vitro niche model and novel physical tools has enabled us to quantitatively determine the relative significance of binding mechanisms between normal HSC versus leukemia blasts to the bone marrow niche.
Collapse
Affiliation(s)
- Alexandra S Burk
- 1] Physical Chemistry of Biosystems, Institute of Physical Chemistry, University of Heidelberg, 69120 Heidelberg, Germany [2] Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, 76021 Karlsruhe, Germany
| | - Cornelia Monzel
- Physical Chemistry of Biosystems, Institute of Physical Chemistry, University of Heidelberg, 69120 Heidelberg, Germany
| | - Hiroshi Y Yoshikawa
- 1] Physical Chemistry of Biosystems, Institute of Physical Chemistry, University of Heidelberg, 69120 Heidelberg, Germany [2] Department of Chemistry, Faculty of Science, Saitama University, Saitama, 338-8570, Japan
| | - Patrick Wuchter
- Department of Medicine V (Hematology, Oncology &Rheumatology), University of Heidelberg, 69120 Heidelberg, Germany
| | - Rainer Saffrich
- Department of Medicine V (Hematology, Oncology &Rheumatology), University of Heidelberg, 69120 Heidelberg, Germany
| | - Volker Eckstein
- Department of Medicine V (Hematology, Oncology &Rheumatology), University of Heidelberg, 69120 Heidelberg, Germany
| | - Motomu Tanaka
- 1] Physical Chemistry of Biosystems, Institute of Physical Chemistry, University of Heidelberg, 69120 Heidelberg, Germany [2] Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, 76021 Karlsruhe, Germany [3] Institute for Integrated Cell-Material Sciences (WPI iCeMS), Kyoto University, 606-8501, Kyoto, Japan
| | - Anthony D Ho
- Department of Medicine V (Hematology, Oncology &Rheumatology), University of Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
82
|
Batnyam O, Shimizu H, Saito K, Ishida T, Suye SI, Fujita S. Biohybrid hematopoietic niche for expansion of hematopoietic stem/progenitor cells by using geometrically controlled fibrous layers. RSC Adv 2015. [DOI: 10.1039/c5ra13332g] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Biohybrid hematopoietic niche for expansion of hematopoietic stem/progenitor cells.
Collapse
Affiliation(s)
- Onon Batnyam
- Department of Frontier Fiber Technology and Science
- Graduate School of Engineering
- University of Fukui
- Fukui
- Japan
| | - Harue Shimizu
- Department of Frontier Fiber Technology and Science
- Graduate School of Engineering
- University of Fukui
- Fukui
- Japan
| | - Koichi Saito
- Research Center for Regenerative Medicine
- EIL Inc
- Tokyo 174-0051
- Japan
| | - Tomohiko Ishida
- Department of Obstetrics and Gynaecology
- Itabashi Chuo Medical Center
- Tokyo 174-0051
- Japan
| | - Shin-ichiro Suye
- Department of Frontier Fiber Technology and Science
- Graduate School of Engineering
- University of Fukui
- Fukui
- Japan
| | - Satoshi Fujita
- Department of Frontier Fiber Technology and Science
- Graduate School of Engineering
- University of Fukui
- Fukui
- Japan
| |
Collapse
|
83
|
Janeczek AA, Scarpa E, A. Newman T, Oreffo ROC, S. Tare R, Evans ND. Skeletal Stem Cell Niche of the Bone Marrow. TISSUE-SPECIFIC STEM CELL NICHE 2015. [DOI: 10.1007/978-3-319-21705-5_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
84
|
Zhao Y, Wu D, Fei C, Guo J, Gu S, Zhu Y, Xu F, Zhang Z, Wu L, Li X, Chang C. Down-regulation of Dicer1 promotes cellular senescence and decreases the differentiation and stem cell-supporting capacities of mesenchymal stromal cells in patients with myelodysplastic syndrome. Haematologica 2014; 100:194-204. [PMID: 25361944 DOI: 10.3324/haematol.2014.109769] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Although it has been reported that mesenchymal stromal cells are unable to provide sufficient hematopoietic support in myelodysplastic syndrome, the underlying mechanisms remain elusive. In this study, we found that mesenchymal stromal cells from patients with myelodysplastic syndrome displayed a significant increase in senescence, as evidenced by their decreased proliferative capacity, flattened morphology and increased expression of SA-β-gal and p21. Senescent mesenchymal stromal cells from patients had decreased differentiation potential and decreased stem cell support capacity. Gene knockdown of Dicer1, which was down-regulated in mesenchymal stromal cells from patients, induced senescence. The differentiation and stem cell-supporting capacities were significantly inhibited by Dicer1 knockdown. Overexpression of Dicer1 in mesenchymal stromal cells from patients reversed cellular senescence and enhanced stem cell properties. Furthermore, we identified reduced expression in the microRNA-17 family (miR-17-5p, miR-20a/b, miR-106a/b and miR-93) as a potential factor responsible for increased p21 expression, a key senescence mediator, in Dicer1 knockdown cells. Moreover, we found that miR-93 and miR-20a expression levels were significantly reduced in mesenchymal stromal cells from patients and miR-93/miR-20a gain of function resulted in a decrease of cellular senescence. Collectively, the results of our study show that mesenchymal stromal cells from patients with myelodysplastic syndrome are prone to senescence and that Dicer1 down-regulation promotes cellular senescence and decreases the differentiation and stem cell-supporting capacities of mesenchymal stromal cells. Dicer1 down-regulation seems to contribute to the insufficient hematopoietic support capacities of mesenchymal stromal cells from patients with myelodysplastic syndrome.
Collapse
Affiliation(s)
- Youshan Zhao
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Dong Wu
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chengming Fei
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Juan Guo
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Shuncheng Gu
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yang Zhu
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Feng Xu
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zheng Zhang
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Lingyun Wu
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiao Li
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chunkang Chang
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
85
|
Walenda T, Stiehl T, Braun H, Fröbel J, Ho AD, Schroeder T, Goecke TW, Rath B, Germing U, Marciniak-Czochra A, Wagner W. Feedback signals in myelodysplastic syndromes: increased self-renewal of the malignant clone suppresses normal hematopoiesis. PLoS Comput Biol 2014; 10:e1003599. [PMID: 24763223 PMCID: PMC3998886 DOI: 10.1371/journal.pcbi.1003599] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 03/18/2014] [Indexed: 12/20/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are triggered by an aberrant hematopoietic stem cell (HSC). It is, however, unclear how this clone interferes with physiologic blood formation. In this study, we followed the hypothesis that the MDS clone impinges on feedback signals for self-renewal and differentiation and thereby suppresses normal hematopoiesis. Based on the theory that the MDS clone affects feedback signals for self-renewal and differentiation and hence suppresses normal hematopoiesis, we have developed a mathematical model to simulate different modifications in MDS-initiating cells and systemic feedback signals during disease development. These simulations revealed that the disease initiating cells must have higher self-renewal rates than normal HSCs to outcompete normal hematopoiesis. We assumed that self-renewal is the default pathway of stem and progenitor cells which is down-regulated by an increasing number of primitive cells in the bone marrow niche – including the premature MDS cells. Furthermore, the proliferative signal is up-regulated by cytopenia. Overall, our model is compatible with clinically observed MDS development, even though a single mutation scenario is unlikely for real disease progression which is usually associated with complex clonal hierarchy. For experimental validation of systemic feedback signals, we analyzed the impact of MDS patient derived serum on hematopoietic progenitor cells in vitro: in fact, MDS serum slightly increased proliferation, whereas maintenance of primitive phenotype was reduced. However, MDS serum did not significantly affect colony forming unit (CFU) frequencies indicating that regulation of self-renewal may involve local signals from the niche. Taken together, we suggest that initial mutations in MDS particularly favor aberrant high self-renewal rates. Accumulation of primitive MDS cells in the bone marrow then interferes with feedback signals for normal hematopoiesis – which then results in cytopenia. Myelodysplastic syndromes are diseases which are characterized by ineffective blood formation. There is accumulating evidence that they are caused by an aberrant hematopoietic stem cell. However, it is yet unclear how this malignant clone suppresses normal hematopoiesis. To this end, we generated mathematical models under the assumption that feedback signals regulate self-renewal and proliferation of normal and diseased stem cells. The simulations demonstrate that the malignant cells must have particularly higher self-renewal rates than normal stem cells – rather than higher proliferation rates. On the other hand, down-regulation of self-renewal by the increasing number of malignant cells in the bone marrow niche can explain impairment of normal blood formation. In fact, we show that serum of patients with myelodysplastic syndrome, as compared to serum of healthy donors, stimulates proliferation and moderately impacts on maintenance of hematopoietic stem and progenitor cells in vitro. Thus, aberrant high self-renewal rates of the malignant clone seem to initiate disease development; suppression of normal blood formation is then caused by a rebound effect of feedback signals which down-regulate self-renewal of normal stem and progenitor cells as well.
Collapse
Affiliation(s)
- Thomas Walenda
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University Medical School, Aachen, Germany
| | - Thomas Stiehl
- Interdisciplinary Center of Scientific Computing (IWR), Institute of Applied Mathematics, University of Heidelberg, Heidelberg, Germany
| | - Hanna Braun
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University Medical School, Aachen, Germany
| | - Julia Fröbel
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Anthony D. Ho
- Department of Medicine V, Medical Center, University of Heidelberg, Heidelberg, Germany
| | - Thomas Schroeder
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Tamme W. Goecke
- Department of Obstetrics and Gynecology, RWTH Aachen University Medical School, Aachen, Germany
| | - Björn Rath
- Department for Orthopedics, RWTH Aachen University Medical School, Aachen, Germany
| | - Ulrich Germing
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Anna Marciniak-Czochra
- Interdisciplinary Center of Scientific Computing (IWR), Institute of Applied Mathematics, University of Heidelberg, Heidelberg, Germany
| | - Wolfgang Wagner
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University Medical School, Aachen, Germany
- * E-mail:
| |
Collapse
|
86
|
van Veen T, Hunt JA. Tissue engineering red blood cells: a therapeutic. J Tissue Eng Regen Med 2014; 9:760-70. [DOI: 10.1002/term.1885] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 01/14/2014] [Accepted: 02/18/2014] [Indexed: 01/10/2023]
Affiliation(s)
- Theun van Veen
- Clinical Engineering, Institute of Ageing and Chronic Disease; University of Liverpool; UK
| | - John A. Hunt
- Clinical Engineering, Institute of Ageing and Chronic Disease; University of Liverpool; UK
| |
Collapse
|
87
|
Erb U, Megaptche AP, Gu X, Büchler MW, Zöller M. CD44 standard and CD44v10 isoform expression on leukemia cells distinctly influences niche embedding of hematopoietic stem cells. J Hematol Oncol 2014; 7:29. [PMID: 24684724 PMCID: PMC4022365 DOI: 10.1186/1756-8722-7-29] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 03/25/2014] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND A blockade of CD44 is considered a therapeutic option for the elimination of leukemia initiating cells. However, anti-panCD44 can interfere with hematopoiesis. Therefore we explored, whether a CD44 variant isoform (CD44v)-specific antibody can inhibit leukemia growth without attacking hematopoiesis. As a model we used CD44v10 transfected EL4 thymoma cells (EL4-v10). METHODS The therapeutic efficacy of anti-panCD44 and anti-CD44v10 was evaluated after intravenous application of EL4/EL4-v10. Ex vivo and in vitro studies evaluated the impact of anti-panCD44 and anti-CD44v10 as well as of EL4 and EL4-v10 on hematopoietic stem cells (HSC) in cocultures with bone marrow stroma cells with a focus on adhesion, migration, cell cycle progression and apoptosis resistance. RESULTS Intravenously injected EL4-v10 grow in bone marrow and spleen. Anti-panCD44 and, more pronounced anti-CD44v10 prolong the survival time. The higher efficacy of anti-CD44v10 compared to anti-panCD44 does not rely on stronger antibody-dependent cellular cytotoxicity or on promoting EL4-v10 apoptosis. Instead, EL4 compete with HSC niche embedding. This has consequences on quiescence and apoptosis-protecting signals provided by the stroma. Anti-panCD44, too, more efficiently affected embedding of HSC than of EL4 in the bone marrow stroma. EL4-v10, by catching osteopontin, migrated on bone marrow stroma and did not or weakly interfere with HSC adhesion. Anti-CD44v10, too, did not affect the HSC--bone marrow stroma crosstalk. CONCLUSION The therapeutic effect of anti-panCD44 and anti-CD44v10 is based on stimulation of antibody-dependent cellular cytotoxicity. The superiority of anti-CD44v10 is partly due to blocking CD44v10-stimulated osteopontin expression that could drive HSC out of the niche. However, the main reason for the superiority of anti-CD44v10 relies on neither EL4-v10 nor anti-CD44v10 severely interfering with HSC--stroma cell interactions that, on the other hand, are affected by EL4 and anti-panCD44. Anti-panCD44 disturbing HSC embedding in the osteogenic niche weakens its therapeutic effect towards EL4. Thus, as far as leukemic cells express CD44v isoforms, the therapeutic use of anti-panCD44 should be avoided in favor of CD44v-specific antibodies.
Collapse
Affiliation(s)
- Ulrike Erb
- Department of Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
| | | | - Xiaoyu Gu
- Department of Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
| | | | - Margot Zöller
- Department of Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
| |
Collapse
|
88
|
Boyette LB, Tuan RS. Adult Stem Cells and Diseases of Aging. J Clin Med 2014; 3:88-134. [PMID: 24757526 PMCID: PMC3992297 DOI: 10.3390/jcm3010088] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 12/15/2013] [Accepted: 12/17/2013] [Indexed: 02/06/2023] Open
Abstract
Preservation of adult stem cells pools is critical for maintaining tissue homeostasis into old age. Exhaustion of adult stem cell pools as a result of deranged metabolic signaling, premature senescence as a response to oncogenic insults to the somatic genome, and other causes contribute to tissue degeneration with age. Both progeria, an extreme example of early-onset aging, and heritable longevity have provided avenues to study regulation of the aging program and its impact on adult stem cell compartments. In this review, we discuss recent findings concerning the effects of aging on stem cells, contributions of stem cells to age-related pathologies, examples of signaling pathways at work in these processes, and lessons about cellular aging gleaned from the development and refinement of cellular reprogramming technologies. We highlight emerging therapeutic approaches to manipulation of key signaling pathways corrupting or exhausting adult stem cells, as well as other approaches targeted at maintaining robust stem cell pools to extend not only lifespan but healthspan.
Collapse
Affiliation(s)
- Lisa B Boyette
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; ; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; ; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA ; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
89
|
Ludwig A, Saffrich R, Eckstein V, Bruckner T, Wagner W, Ho AD, Wuchter P. Functional potentials of human hematopoietic progenitor cells are maintained by mesenchymal stromal cells and not impaired by plerixafor. Cytotherapy 2014; 16:111-21. [DOI: 10.1016/j.jcyt.2013.07.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 07/22/2013] [Accepted: 07/27/2013] [Indexed: 11/15/2022]
|
90
|
Wuchter P, Leinweber C, Saffrich R, Hanke M, Eckstein V, Ho AD, Grunze M, Rosenhahn A. Plerixafor induces the rapid and transient release of stromal cell-derived factor-1 alpha from human mesenchymal stromal cells and influences the migration behavior of human hematopoietic progenitor cells. Cell Tissue Res 2013; 355:315-26. [DOI: 10.1007/s00441-013-1759-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 10/29/2013] [Indexed: 12/17/2022]
|
91
|
Hematopoietic stem and progenitor cells acquire distinct DNA-hypermethylation during in vitro culture. Sci Rep 2013; 3:3372. [PMID: 24284763 PMCID: PMC3842544 DOI: 10.1038/srep03372] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 11/11/2013] [Indexed: 01/08/2023] Open
Abstract
Hematopoietic stem and progenitor cells (HPCs) can be maintained invitro, but the vast majority of their progeny loses stemness during culture. In this study, we compared DNA-methylation (DNAm) profiles of freshly isolated and culture-expanded HPCs. Culture conditions of CD34+ cells - either with or without mesenchymal stromal cells (MSCs) - had relatively little impact on DNAm, although proliferation is greatly increased by stromal support. However, all cultured HPCs - even those which remained CD34+ - acquired significant DNA-hypermethylation. DNA-hypermethylation occurred particularly in up-stream promoter regions, shore-regions of CpG islands, binding sites for PU.1, HOXA5 and RUNX1, and it was reflected in differential gene expression and variant transcripts of DNMT3A. Low concentrations of DNAm inhibitors slightly increased the frequency of colony-forming unit initiating cells. Our results demonstrate that HPCs acquire DNA-hypermethylation at specific sites in the genome which is relevant for the rapid loss of stemness during in vitro manipulation.
Collapse
|
92
|
Raynaud CM, Butler JM, Halabi NM, Ahmad FS, Ahmed B, Rafii S, Rafii A. Endothelial cells provide a niche for placental hematopoietic stem/progenitor cell expansion through broad transcriptomic modification. Stem Cell Res 2013; 11:1074-90. [PMID: 23978474 DOI: 10.1016/j.scr.2013.07.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 07/17/2013] [Accepted: 07/31/2013] [Indexed: 11/26/2022] Open
Abstract
Umbilical cord blood (UCB) is an attractive source of hematopoietic stem cells (HSCs). However, the number of HSCs in UCB is limited, and attempts to amplify them in vitro remain inefficient. Several publications have documented amplification of hematopoietic stem/progenitor cells (HSPCs) on endothelial or mesenchymal cells, but the lack of homogeneity in culture conditions and HSC definition impairs direct comparison of these results. We investigated the ability of different feeder layers, mesenchymal progenitors (MPs) and endothelial cells (ECs), to amplify hematopoietic stem/progenitor cells. Placental derived HSPCs (defined as Lin(-)CD45(-/dim)CD34(+)CD38(-)CD90(+)) were maintained on confluent feeder layers and the number of cells and their marker expression were monitored over 21 days. Although both types of feeder layers supported hematopoietic expansion, only endothelial cells triggered amplification of Lin(-)CD45(-/dim)CD34(+)CD38(-)CD90(+) cells, which peaked at 14 days. The amplified cells differentiated into all cell lineages, as attested by in vitro colony-forming assays, and were capable of engraftment and multi-lineage differentiation in sub-lethally irradiated mice. Mesenchymal progenitors promoted amplification of CD38(+) cells, previously defined as precursors with more limited differentiation potential. A competitive assay demonstrated that hematopoietic stem/progenitor cells had a preference for interacting with endothelial cells in vitro. Cytokine and transcriptomic analysis of both feeder cell types identified differences in gene expression that correlated with propensity of ECs and MPs to support hematopoietic cell amplification and differentiation respectively. Finally, we used RNA sequencing of endothelial cells and HSPCs to uncover relevant networks illustrating the complex interaction between endothelial cells and HSPCs leading to stem/progenitor cell expansion.
Collapse
Affiliation(s)
- Christophe M Raynaud
- Qatar Cardiovascular Research Center, Qatar Foundation, Qatar Science and Technology Park, Doha, Qatar
| | | | | | | | | | | | | |
Collapse
|
93
|
Mesenchymal stem cells in the treatment of pediatric diseases. World J Pediatr 2013; 9:197-211. [PMID: 23929252 DOI: 10.1007/s12519-013-0425-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Accepted: 06/04/2013] [Indexed: 02/06/2023]
Abstract
BACKGROUND In recent years, the incredible interests in mesenchymal stem cells have boosted the expectations of both patients and physicians. Unlike embryonic stem cells, neither their procurement nor their use is deemed controversial. Moreover, their immunomodulatory capacity coupled with low immunogenicity has opened up their allogenic use, consequently broadening the possibilities for their application. In May 2012, Canadian health regulators approved Prochymal, the first mesenchymal stem cells-based drug, for acute graft-versus-host diseases in children who have failed to respond to steroid treatment. The aim of this article is to review the recent advances in mesenchymal stem cells for pediatric diseases. DATA SOURCES A literature review was performed on PubMed from 1966 to 2013 using the MeSH terms "mesenchymal stem cells", "clinical trials" and "children". Additional articles were identified by a hand search of the references list in the initial search. RESULTS The following categories are described: general properties, mechanisms of action, graft-versus-host diseases, cardiovascular diseases, liver diseases, inflammatory bowel diseases, osteoarticular diseases, autoimmune diseases, type 1 diabetes, and lung diseases. CONCLUSIONS Mesenchymal stem cells, owing to their availability, immunomodulatory properties, low immunogenicity, and therapeutic potential, have become one of the most attractive options for the treatment of a wide range of diseases. It is expected to see more and more clinical trials and applications of mesenchymal stem cells for pediatric diseases in the near future.
Collapse
|
94
|
Soltanpour MS, Amirizadeh N, Zaker F, Oodi A, Nikougoftar M, Kazemi A. mRNA expression and promoter DNA methylation status of CDKi p21 and p57 genes inex vivoexpanded CD34+cells following co-culture with mesenchymal stromal cells and growth factors. Hematology 2013; 18:30-8. [DOI: 10.1179/1607845412y.0000000030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Affiliation(s)
- Mohammad Soleiman Soltanpour
- Department of Hematology and Blood BankingSchool of Allied Medical Sciences, Tehran University of Medical Science, Tehran, Iran
| | - Naser Amirizadeh
- High Institute for Research and Education in Transfusion Medicine, Blood Transfusion Research Center, Tehran, Iran
| | - Farhad Zaker
- Department of Hematology and Blood BankingSchool of Allied Medical Sciences, Tehran University of Medical Science, Tehran, Iran
| | - Arezoo Oodi
- High Institute for Research and Education in Transfusion Medicine, Blood Transfusion Research Center, Tehran, Iran
| | - Mahin Nikougoftar
- High Institute for Research and Education in Transfusion Medicine, Blood Transfusion Research Center, Tehran, Iran
| | - Ahmad Kazemi
- Department of Hematology and Blood BankingSchool of Allied Medical Sciences, Tehran University of Medical Science, Tehran, Iran
| |
Collapse
|
95
|
Ferrer RA, Wobus M, List C, Wehner R, Schönefeldt C, Brocard B, Mohr B, Rauner M, Schmitz M, Stiehler M, Ehninger G, Hofbauer LC, Bornhäuser M, Platzbecker U. Mesenchymal stromal cells from patients with myelodyplastic syndrome display distinct functional alterations that are modulated by lenalidomide. Haematologica 2013; 98:1677-85. [PMID: 23716561 DOI: 10.3324/haematol.2013.083972] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The contribution of the bone marrow microenvironment in myelodysplastic syndrome is controversial. We therefore analyzed the functional properties of primary mesenchymal stromal cells from patients with myelodysplastic syndrome in the presence or absence of lenalidomide. Compared to healthy controls, clonality and growth were reduced across all disease stages. Furthermore, differentiation defects and particular expression of adhesion and cell surface molecules (e.g. CD166, CD29, CD146) were detected. Interestingly, the levels of stromal derived factor 1-alpha in patients' cells culture supernatants were almost 2-fold lower (P<0.01) than those in controls and this was paralleled by a reduced induction of migration of CD34(+) hematopoietic cells. Co-cultures of mesenchymal stromal cells from patients with CD34(+) cells from healthy donors resulted in reduced numbers of cobblestone area-forming cells and fewer colony-forming units. Exposure of stromal cells from patients and controls to lenalidomide led to a further reduction of stromal derived factor 1-alpha secretion and cobblestone area formation, respectively. Moreover, lenalidomide pretreatment of mesenchymal stromal cells from patients with low but not high-risk myelodysplastic syndrome was able to rescue impaired erythroid and myeloid colony formation of early hematopoietic progenitors. In conclusion, our analyses support the notion that the stromal microenvironment is involved in the pathophysiology of myelodysplastic syndrome thus representing a potential target for therapeutic interventions.
Collapse
|
96
|
Kapetanaki MG, Mora AL, Rojas M. Influence of age on wound healing and fibrosis. J Pathol 2013; 229:310-22. [PMID: 23124998 DOI: 10.1002/path.4122] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 09/30/2012] [Accepted: 10/02/2012] [Indexed: 12/18/2022]
Abstract
The incidence and severity of fibrotic lung diseases increase with age, but very little is known about how age-related changes affect the mechanisms that underlie disease emergence and progression. Normal ageing includes accumulation of DNA mutations, oxidative and cell stresses, mitochondria dysfunction, increased susceptibility to apoptosis, telomere length dysfunction and differential gene expression as a consequence of epigenetic changes and miR regulation. These inevitable ageing-related phenomena may cause dysfunction and impaired repair capacity of lung epithelial cells, fibroblasts and MSCs. As a consequence, the composition of the extracellular matrix changes and the dynamic interaction between cells and their environment is damaged, resulting ultimately in predisposition for several diseases. This review summarizes what is known about age-related molecular changes that are implicated in the pathobiology of lung fibrosis in lung tissue.
Collapse
Affiliation(s)
- Maria G Kapetanaki
- Dorothy P and Richard P Simmons Center for Interstitial Lung Disease, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | |
Collapse
|
97
|
Stromal cell-mediated inhibition of erythropoiesis can be attenuated by Sotatercept (ACE-011), an activin receptor type II ligand trap. Exp Hematol 2013; 41:155-166.e17. [DOI: 10.1016/j.exphem.2012.12.002] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 11/27/2012] [Accepted: 12/06/2012] [Indexed: 12/24/2022]
|
98
|
Schellenberg A, Hemeda H, Wagner W. Tracking of replicative senescence in mesenchymal stem cells by colony-forming unit frequency. Methods Mol Biol 2013; 976:143-154. [PMID: 23400440 DOI: 10.1007/978-1-62703-317-6_11] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Long-term culture of mesenchymal stem cells (MSC) has major impact on cellular characteristics and differentiation potential. Numerous clinical trials raise high hopes in regenerative medicine and this necessitates reliable quality control of the cellular products-also with regard to replicative senescence. The maximum number of population doublings before entering the senescent state depends on the cell type, tissue of origin, culture medium as well as cell culture methods. Therefore, it would be valuable to predict the remaining proliferative potential in the course of culture expansion. Here, we describe a refined fibroblastic colony forming unit (CFU-f) assay which can be performed at any passage during culture expansion with simple cell culture techniques. This method is based on limiting dilutions in the 96-well format to determine the proportion of highly proliferative and clonogenic cells. The number of CFU-f declines rapidly during culture expansion. Especially at higher passages the CFU-f frequency correlates very well with the remaining cumulative population doublings. This approach can be used as quality measure to estimate the remaining proliferative potential of MSC in culture.
Collapse
Affiliation(s)
- Anne Schellenberg
- Stem Cell Biology and Cellular Engineering, Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University Medical School, Aachen, Germany
| | | | | |
Collapse
|
99
|
Zhou Y, Chakravorty N, Xiao Y, Gu W. Mesenchymal stem cells and nano-structured surfaces. Methods Mol Biol 2013; 1058:133-48. [PMID: 23943531 DOI: 10.1007/7651_2013_30] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSCs) represent multipotent stromal cells that can differentiate into a variety of cell types, including osteoblasts (bone cells), chondrocytes (cartilage cells), and adipocytes (fat cells). Their multi-potency provides a great promise as a cell source for tissue engineering and cell-based therapy for many diseases, particularly bone diseases and bone formation. To be able to direct and modulate the differentiation of MSCs into the desired cell types in situ in the tissue, nanotechnology is introduced and used to facilitate or promote cell growth and differentiation. These nano-materials can provide a fine structure and tuneable surface in nanoscales to help the cell adhesion and promote the cell growth and differentiation of MSCs. This could be a dominant direction in future for stem cells based therapy or tissue engineering for various diseases. Therefore, the isolation, manipulation, and differentiation of MSCs are very important steps to make meaningful use of MSCs for disease treatments. In this chapter, we have described a method of isolating MSC from human bone marrow, and how to culture and differentiate them in vitro. We have also provided research methods on how to use MSCs in an in vitro model and how to observe MSC biological response on the surface of nano-scaled materials.
Collapse
Affiliation(s)
- Yinghong Zhou
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | | | | | | |
Collapse
|
100
|
Wuchter P, Wagner W, Ho AD. Mesenchymal Stem Cells – An Oversimplified Nomenclature for Extremely Heterogeneous Progenitors. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|