51
|
Cell membrane engineering with synthetic materials: Applications in cell spheroids, cellular glues and microtissue formation. Acta Biomater 2019; 90:21-36. [PMID: 30986529 DOI: 10.1016/j.actbio.2019.04.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 03/26/2019] [Accepted: 04/03/2019] [Indexed: 12/16/2022]
Abstract
Biologically inspired materials with tunable bio- and physicochemical properties provide an essential framework to actively control and support cellular behavior. Cell membrane remodeling approaches benefit from the advances in polymer science and bioconjugation methods, which allow for the installation of un-/natural molecules and particles on the cells' surface. Synthetically remodeled cells have superior properties and are under intense investigation in various therapeutic scenarios as cell delivery systems, bio-sensing platforms, injectable biomaterials and bioinks for 3D bioprinting applications. In this review article, recent advances in the field of cell surface remodeling via bio-chemical means and the potential biomedical applications of these emerging cell hybrids are discussed. STATEMENT OF SIGNIFICANCE: Recent advances in bioconjugation methods, controlled/living polymerizations, microfabrication techniques and 3D printing technologies have enabled researchers to probe specific cellular functions and cues for therapeutic and research purposes through the formation of cell spheroids and polymer-cell chimeras. This review article highlights recent non-genetic cell membrane engineering strategies towards the fabrication of cellular ensembles and microtissues with interest in 3D in vitro modeling, cell therapeutics and tissue engineering. From a wider perspective, these approaches may provide a roadmap for future advances in cell therapies which will expedite the clinical use of cells, thereby improving the quality and accessibility of disease treatments.
Collapse
|
52
|
Lu M, Peng L, Ming X, Wang X, Cui A, Li Y, Wang X, Meng D, Sun N, Xiang M, Chen S. Enhanced wound healing promotion by immune response-free monkey autologous iPSCs and exosomes vs. their allogeneic counterparts. EBioMedicine 2019; 42:443-457. [PMID: 30926422 PMCID: PMC6491387 DOI: 10.1016/j.ebiom.2019.03.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 02/08/2019] [Accepted: 03/04/2019] [Indexed: 12/20/2022] Open
Abstract
Background Comparing non-inbred autologous and allogeneic induced pluripotent stem cells (iPSCs) and their secreted subcellular products among non-human primates is critical for choosing optimal iPSC products for human clinical trials. Methods iPSCs were induced from skin fibroblastic cells of adult male rhesus macaques belonging to four unrelated consanguineous families. Teratoma generativity, host immune response, and skin wound healing promotion were evaluated subsequently. Findings All autologous, but no allogeneic, iPSCs formed teratomas, whereas all allogeneic, but no autologous, iPSCs caused lymphocyte infiltration. Macrophages were not detectable in any wound. iPSCs expressed significantly more MAMU A and E of the major histocompatibility complex (MHC) class I but not more other MHC genetic alleles than parental fibroblastic cells. All topically disseminated autologous and allogeneic iPSCs, and their exosomes accelerated skin wound healing, as demonstrated by wound closure, epithelial coverage, collagen deposition, and angiogenesis. Allogeneic iPSCs and their exosomes were less effective and viable than their autologous counterparts. Some iPSCs differentiated into new endothelial cells and all iPSCs lost their pluripotency in 14 days. Exosomes increased cell viability of injured epidermal, endothelial, and fibroblastic cells in vitro. Although exosomes contained some mRNAs of pluripotent factors, they did not impart pluripotency to host cells. Interpretation Although all of the autologous and allogeneic iPSCs and exosomes accelerated wound healing, allogeneic iPSC exosomes were the preferred choice for “off-the shelf” iPSC products, owing to their mass-production, with no concern of teratoma formation. Fund National Natural Science Foundation of China and National Key R&D Program of China.
Autologous but not allogeneic rhesus macaque iPSCs generate teratomas. Non-inbred allogeneic but not autologous iPSCs attract lymphocytes. Both autologous and allogeneic iPSCs and their exosomes promote wound healing. Both autologous iPSCs and exosomes promote better wound healing than their allogeneic counterparts. iPSCs lost their pluripotency in vivo. Exosomes did not impart pluripotency to host cells.
Collapse
Affiliation(s)
- Meng Lu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Lu Peng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xu Ming
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xiaokai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Anfeng Cui
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yijun Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xinhong Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Dan Meng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Ning Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Meng Xiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Sifeng Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
53
|
Abati E, Bresolin N, Comi G, Corti S. Advances, Challenges, and Perspectives in Translational Stem Cell Therapy for Amyotrophic Lateral Sclerosis. Mol Neurobiol 2019; 56:6703-6715. [DOI: 10.1007/s12035-019-1554-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 03/13/2019] [Indexed: 12/13/2022]
|
54
|
Li M, Sun S, Dangelmajer S, Zhang Q, Wang J, Hu F, Dong F, Kahlert UD, Zhu M, Lei T. Exploiting tumor-intrinsic signals to induce mesenchymal stem cell-mediated suicide gene therapy to fight malignant glioma. Stem Cell Res Ther 2019; 10:88. [PMID: 30867058 PMCID: PMC6417183 DOI: 10.1186/s13287-019-1194-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/19/2019] [Accepted: 02/25/2019] [Indexed: 12/13/2022] Open
Abstract
Background Human mesenchymal stem cell (MSC)-based tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) gene delivery is regarded as an effective treatment for glioblastoma (GBM). However, adverse-free target site homing of the delivery vehicles to the tumor microsatellite nests is challenging, leading to erroneously sustained released of this suicide protein into the normal brain parenchyma; therefore, limiting off-target cytotoxicity and controlled expression of the suicide inductor is a prerequisite for the safe use of therapeutic stem cells. Methods Utilizing the intrinsic expression profile of GBM and its elevated expression of TGF-β relative to normal brain tissue, we sought to engineer human adipose-derived MSCs (hAMSC-SBE4-TRAIL) which augment the expression of TRAIL under the trigger of TGF-β signaling. We validated our therapeutic technology in a series of functional in vitro and in vivo assays using primary patient-derived GBM models. Results Our current findings show that these biologic delivery vehicles have high tumor tropism efficacy and expression TRAIL gene under the trigger of TGF-β-secreting GBMs, as well as avoid unspecific TRAIL secretion into normal brain tissue. hAMSC-SBE4-TRAIL inhibited the proliferation and induced apoptosis in experimental GBMs both in vitro and in vivo. In addition, our improved platform of engineered MSCs significantly decreased the tumor volume and prolonged survival time in a murine model of GBM. Conclusions Our results on the controlled release of suicide inductor TRAIL by exploiting an endogenous tumor signaling pathway demonstrate a significant improvement for the clinical utility of stem cell-mediated gene delivery to treat brain cancers. Harvesting immune-compatible MSCs from patients’ fat by minimally invasive procedures further highlights the clinical potential of this approach in the vision of applicability in a personalized manner. The hAMSC-SBE4-TRAIL exhibit great curative efficacy and are a promising cell-based treatment option for GBM to be validated in clinical exploration. Electronic supplementary material The online version of this article (10.1186/s13287-019-1194-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Man Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.,Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Shoujia Sun
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Sean Dangelmajer
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Quan Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
| | - Junwen Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
| | - Feng Hu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
| | - Fangyong Dong
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
| | - Ulf D Kahlert
- Department of Neurosurgery, University Medical Center Düsseldorf, German Cancer Consortium (DKTK), Essen/Dusseldorf, Germany
| | - Mingxin Zhu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China.
| | - Ting Lei
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
| |
Collapse
|
55
|
Amniotic cells share clusters of differentiation of fibroblasts and keratinocytes, influencing their ability to proliferate and aid in wound healing while impairing their angiogenesis capability. Eur J Pharmacol 2019; 854:167-178. [PMID: 30826324 DOI: 10.1016/j.ejphar.2019.02.043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/22/2019] [Accepted: 02/26/2019] [Indexed: 12/19/2022]
Abstract
An alternative to cultured skin cell grafts usage in burn treatment is the graft of allogenic stem cells. We verified whether amniotic stem cells are better than the present therapeutic standard: grafts of autologous keratinocytes and fibroblasts along with autologous adipose-derived stem cells, and whether amniotic stem cells can support the growth of autologous keratinocytes and fibroblasts in the culture. The study was performed on the material from 18 amnia. Skin cells were obtained from 3 patients. In order to assess the influence of stem cells on keratinocytes and fibroblasts, the following experiments were performed: impact on viability and cell cycle, wound healing capability, angiogenesis capability, influence on the proliferation speed and capability to differentiate into skin cells. We demonstrated that human amniotic membrane-derived mesenchymal stem cells (hAMMSCs) share amniotic proteins with skin cells. Amniotic stem cells may replace skin fibroblasts in grafts due to the close similarity in their surface antigens, with significantly larger proliferative potential and ability to stimulate wound healing. It was shown that adding amniotic cells to both keratinocytes and fibroblast cultures accelerates directional migration by ≥ 40%. We confirmed in this study the influence of amniotic cells on the proliferation and cell cycle of fibroblasts and keratinocytes. Amniotic stem cells can be successfully used not only as a first choice graft but also to replace 3T3 line cells, supporting the proliferation of the cells during the culturing, as well as a supplementary graft supporting an autologous graft of keratinocytes and fibroblasts.
Collapse
|
56
|
Saffari_Chaleshtori J, Shafiee SM, Ghatreh-Samani K, Jalilian N. The study of drug resistance properties of ABCG2 (ATP-binding cassette G2) in contact with thymoquinone, gallic acid, and hesperetin antioxidants. JOURNAL OF HERBMED PHARMACOLOGY 2019. [DOI: 10.15171/jhp.2019.17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Introduction: ATP-binding cassette (ABC) transporters are a group of intra membrane proteins that play key roles in the transmission and exchange of vital compounds on both sides of the membrane. These proteins can specially transport anti-cancer drugs out of cancer cells. ABCG2 is a member of this family that is extremely expressed in many cancers. This study, aims to evaluate the binding affinity of three antioxidants thymoquinone (TQ), gallic acid (GA), and hesperetin (HP) to ABCG2 compared with an anti-cancer drug, mitoxantrone (Mit), to export cells. Methods: The PDB file of ABCG2 was obtained from the protein data bank server (http://www.rcsb.org) with ID: 5NJ3. After 200 stages of molecular docking running on ABCG2 protein in AutoDock v.4.2 software, the amino acids involved in the binding site of each compound were identified using the LigPlot+ software. Results: HP had the lowest (-6.36 kcal/mol) and GA had the highest (-3.93 kcal/mol) binding energy in comparison with Mit (-0.06 kcal/mol) for binding to ABCG2. Effective concentration required to perform the reaction between ABCG2 was higher in GA (1.31 mM) than TQ (42.69 μM) and HP (21.74 μM). GA, HP, and TQ formed 17, 18, and 22 hydrogen and hydrophobic bonds at the binding site of ABCG2. Conclusion: It seems that GA has the lowest affinity to make contact with ABCG2 binding site. So, GA tends to remain in the cell but TQ and HP tend to leave the cell easily via ABCG2 transporter.
Collapse
Affiliation(s)
- Javad Saffari_Chaleshtori
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sayed Mohammad Shafiee
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Keihan Ghatreh-Samani
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Narges Jalilian
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
57
|
Kurtz A, Elsallab M, Sanzenbacher R, Abou-El-Enein M. Linking Scattered Stem Cell-Based Data to Advance Therapeutic Development. Trends Mol Med 2019; 25:8-19. [DOI: 10.1016/j.molmed.2018.10.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/20/2018] [Accepted: 10/22/2018] [Indexed: 02/07/2023]
|
58
|
Abstract
This review briefly describes the last decades of experimental work on the thymus. Given the histological complexity of this organ, the multiple embryological origins of its cellular components and its role in carefully regulating T lymphocyte maturation and function, methods to dissect and understand this complexity have been developed through the years. The possibility to study ex vivo the thymus organ function has been achieved by developing Fetal Thymus Organ Cultures (FTOC). Subsequently, the combination of organ disaggregation and reaggregation in vitro represented by Reaggregate Thymus Organ cultures (RTOC) allowed mixing cellular components from different genetic backgrounds. Moreover, RTOC allowed dissecting the different stromal and hematological components to study the interactions between Major Histocompatibility Complex (MHC) molecules and the T-cell receptors during thymocytes selection. In more recent years, prospective isolation of stromal cells and thymocytes at different stages of development made it possible to explore and elucidate the molecular and cellular players in both the developing and adult thymus. Finally, the appearance of novel cell sources such as embryonic stem (ES) cells and more recently induced pluripotent stem (iPS) cells has opened new scenarios in modelling thymus development and regeneration strategies. Most of the work described was carried out in rodents and the current challenge is to develop equivalent or even more informative assays and tools in entirely human model systems.
Collapse
|
59
|
Erikainen S, Pickersgill M, Cunningham-Burley S, Chan S. Patienthood and participation in the digital era. Digit Health 2019; 5:2055207619845546. [PMID: 31041112 PMCID: PMC6482654 DOI: 10.1177/2055207619845546] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 03/28/2019] [Indexed: 12/15/2022] Open
Abstract
The 'digital era' of informatics and knowledge integration has changed the roles and experiences of patients, research participants and health consumers. No longer figured (merely) as passive recipients of healthcare services or as beneficiaries of top-down biomedical information, individuals are increasingly seen as active contributors in healthcare and research. They are positioned into multiple roles that are experienced simultaneously by those who access and co-produce digital content that can easily be transformed into data. This is contextualised by 'big data' technologies that have altered biomedicine, enabling collation and analysis of myriad data from digitised records to personal mobile data. Social media facilitate new formations of communities and knowledge enacted online, while novel kinds of commercial value emerge from digital networks that enable health data commodification. In this paper, we draw from exemplary digital era shifts towards participatory medicine to cast light on the rapprochements between patienthood, participation and consumption, and we explore how these rapprochements are mediated by, and materialise through, the use of participatory digital technologies and big data. We argue that there is a need to use new conceptual tools that account for the multiple roles and experiences of patient-participant-consumers that co-emerge through digital technologies. We must also ethically re-assess the rights and responsibilities of individuals in the digital era, and the implications of digital era changes for the future of biomedicine and healthcare.
Collapse
Affiliation(s)
- Sonja Erikainen
- Centre for Biomedicine, Self and Society, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Old Medical School, UK
| | - Martyn Pickersgill
- Centre for Biomedicine, Self and Society, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Old Medical School, UK
| | - Sarah Cunningham-Burley
- Centre for Biomedicine, Self and Society, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Old Medical School, UK
| | - Sarah Chan
- Centre for Biomedicine, Self and Society, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Old Medical School, UK
| |
Collapse
|
60
|
Liang K, Bae KH, Kurisawa M. Recent advances in the design of injectable hydrogels for stem cell-based therapy. J Mater Chem B 2019. [DOI: 10.1039/c9tb00485h] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The recent advances in the design of injectable hydrogels for stem cell delivery, especially for in vivo applications, are overviewed in this review.
Collapse
Affiliation(s)
- Kun Liang
- Institute of Bioengineering and Nanotechnology
- Singapore 138669
- Singapore
| | - Ki Hyun Bae
- Institute of Bioengineering and Nanotechnology
- Singapore 138669
- Singapore
| | - Motoichi Kurisawa
- Institute of Bioengineering and Nanotechnology
- Singapore 138669
- Singapore
| |
Collapse
|
61
|
Sane MS, Misra N, Mousa OM, Czop S, Tang H, Khoo LT, Jones CD, Mustafi SB. Cytokines in umbilical cord blood-derived cellular product: a mechanistic insight into bone repair. Regen Med 2018; 13:881-898. [PMID: 30346891 PMCID: PMC6439518 DOI: 10.2217/rme-2018-0102] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/04/2018] [Indexed: 12/12/2022] Open
Abstract
AIM Umbilical cord blood (UCB) finds frequent applications in regenerative medicine. We evaluated the role of cytokines present in a uniquely processed, UCB-derived cellular allograft product (UCBp). MATERIALS & METHODS Luminex multiplex assay and standard cell biology methods were employed. RESULTS Study with allografts from 33 donors identified 44 quantifiable cytokines in the UCBp derived conditioned media (CM). The UCBp-CM elevated proliferation and migration rates of mesenchymal stem cells (MSCs) and bone marrow stromal cells. Moreover, UCBp-CM induced secretion of VEGF-A and osteoprotegerin, which promoted angiogenesis of endothelial cells and positively influenced the osteogenic differentiation of MSCs, respectively. CONCLUSION Cytokines in UCBp stimulate cellular processes important for bone regeneration, making UCBp an excellent candidate for potential applications in orthopedic procedures like bone non-union and spinal fusion.
Collapse
Affiliation(s)
- Mukta S Sane
- Department of Research & Development, Burst Biologics, Boise, ID 83705, USA
| | - Neha Misra
- Department of Research & Development, Burst Biologics, Boise, ID 83705, USA
| | - Omid Mohammad Mousa
- Department of Research & Development, Burst Biologics, Boise, ID 83705, USA
- Department of Regulatory & Medical Affairs, Burst Biologics, Boise, ID 83705, USA
- Biomolecular Research Center, Boise State University, Boise, ID, 83725, USA
| | - Steve Czop
- Department of Regulatory & Medical Affairs, Burst Biologics, Boise, ID 83705, USA
| | - Huiyuan Tang
- Department of Research & Development, Burst Biologics, Boise, ID 83705, USA
| | - Larry T Khoo
- The Spine Clinic of Los Angeles, Good Samaritan Hospital, University of Southern California, Los Angeles, CA 90017, USA
| | - Christopher D Jones
- Department of Research & Development, Burst Biologics, Boise, ID 83705, USA
- Department of Regulatory & Medical Affairs, Burst Biologics, Boise, ID 83705, USA
| | | |
Collapse
|
62
|
Generation of Progesterone-Responsive Endometrial Stromal Fibroblasts from Human Induced Pluripotent Stem Cells: Role of the WNT/CTNNB1 Pathway. Stem Cell Reports 2018; 11:1136-1155. [PMID: 30392973 PMCID: PMC6234962 DOI: 10.1016/j.stemcr.2018.10.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 10/01/2018] [Accepted: 10/02/2018] [Indexed: 12/26/2022] Open
Abstract
Defective endometrial stromal fibroblasts (EMSFs) contribute to uterine factor infertility, endometriosis, and endometrial cancer. Induced pluripotent stem cells (iPSCs) derived from skin or bone marrow biopsies provide a patient-specific source that can be differentiated to various cells types. Replacement of abnormal EMSFs is a potential novel therapeutic approach for endometrial disease; however, the methodology or mechanism for differentiating iPSCs to EMSFs is unknown. The uterus differentiates from the intermediate mesoderm (IM) to form coelomic epithelium (CE) followed by the Müllerian duct (MD). Here, we successfully directed the differentiation of human iPSCs (hiPSCs) through IM, CE, and MD to EMSFs under molecularly defined embryoid body culture conditions using specific hormonal treatments. Activation of CTNNB1 was essential for expression of progesterone receptor that mediated the final differentiation step of EMSFs before implantation. These hiPSC-derived tissues illustrate the potential for iPSC-based endometrial regeneration for future cell-based treatments.
We developed a molecularly defined system for differentiating hiPSCs to EMSFs hiPSC-derived EMSFs undergo decidualization in response to hormonal stimulation D14 embryoid bodies recapitulate the molecular signature of primary EMSFs The WNT/CTNNB1 pathway is required for induction of EMSF from hiPSCs
Collapse
|
63
|
Ji Y, Zhang P, Xing Y, Jia L, Zhang Y, Jia T, Wu X, Zhao B, Xu X. Effect of 1α, 25-dihydroxyvitamin D3 on the osteogenic differentiation of human periodontal ligament stem cells and the underlying regulatory mechanism. Int J Mol Med 2018; 43:167-176. [PMID: 30365053 PMCID: PMC6257868 DOI: 10.3892/ijmm.2018.3947] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 10/17/2018] [Indexed: 12/30/2022] Open
Abstract
1α, 25-dihydroxyvitamin D3 (1,25-D3), an active vitamin D metabolite, is a well-known regulator of osteogenic differentiation. However, how 1,25-D3 regulates osteogenic differentiation in human periodontal ligament stem cells (hPDLSCs) remains to be fully elucidated. The present study aimed to clarify this issue through well-controlled in vitro experiments. After hPDLSCs were treated with 1,25-D3, immunofluorescence and western blotting were used to detect the expression of vitamin D receptor; Cell Counting Kit-8 and western blotting were used to assay the cell proliferation ability. Alkaline phosphatase staining, Alizarin Red staining and western blotting were used to detect the osteogenic differentiation. It was found that treating hPDLSCs with 1,25-D3: i) Inhibited cell proliferation; ii) promoted osteogenic differentiation; iii) upregulated the expression of transcriptional coactivator with PDZ-binding motif (TAZ), an important downstream effector of Hippo signaling that has been demonstrated to be involved in the osteogenic differentiation of stem/progenitor cells; and iv) that co-treatment of TAZ-overexpressing hPDLSCs with 1,25-D3 synergistically stimulated the expression of osteogenic markers. These results suggested that the induction of osteogenic differentiation promoted by 1,25-D3 in hPDLSCs involves, at least in part, the action of TAZ.
Collapse
Affiliation(s)
- Yawen Ji
- School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Panpan Zhang
- School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yixiao Xing
- School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Linglu Jia
- School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yunpeng Zhang
- School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Tingting Jia
- School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xuan Wu
- School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Bin Zhao
- School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xin Xu
- School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
64
|
O'Neill CL, McLoughlin KJ, Chambers SEJ, Guduric-Fuchs J, Stitt AW, Medina RJ. The Vasoreparative Potential of Endothelial Colony Forming Cells: A Journey Through Pre-clinical Studies. Front Med (Lausanne) 2018; 5:273. [PMID: 30460233 PMCID: PMC6232760 DOI: 10.3389/fmed.2018.00273] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/05/2018] [Indexed: 12/24/2022] Open
Abstract
For over a decade various cell populations have been investigated for their vasoreparative potential. Cells with the capacity to promote blood vessel regeneration are commonly known as endothelial progenitor cells (EPCs); although such a definition is currently considered too simple for the complexity of cell populations involved in the reparative angiogenic process. A subset of EPCs called endothelial colony forming cells (ECFCs) have emerged as a suitable candidate for cytotherapy, primarily due to their clonogenic progenitor characteristics, unequivocal endothelial phenotype, and inherent ability to promote vasculogenesis. ECFCs can be readily isolated from human peripheral and cord blood, expanded ex vivo and used to revascularize ischemic tissues. These cells have demonstrated efficacy in several in vivo preclinical models such as the ischemic heart, retina, brain, limb, lung and kidney. This review will summarize the current pre-clinical evidence for ECFC cytotherapy and discuss their potential for clinical application.
Collapse
Affiliation(s)
- Christina L O'Neill
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Kiran J McLoughlin
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Sarah E J Chambers
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Jasenka Guduric-Fuchs
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Alan W Stitt
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Reinhold J Medina
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
65
|
Erkoc P, Yasa IC, Ceylan H, Yasa O, Alapan Y, Sitti M. Mobile Microrobots for Active Therapeutic Delivery. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800064] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Pelin Erkoc
- Physical Intelligence Department; Max Planck Institute for Intelligent; Systems 70569 Stuttgart Germany
| | - Immihan C. Yasa
- Physical Intelligence Department; Max Planck Institute for Intelligent; Systems 70569 Stuttgart Germany
| | - Hakan Ceylan
- Physical Intelligence Department; Max Planck Institute for Intelligent; Systems 70569 Stuttgart Germany
| | - Oncay Yasa
- Physical Intelligence Department; Max Planck Institute for Intelligent; Systems 70569 Stuttgart Germany
| | - Yunus Alapan
- Physical Intelligence Department; Max Planck Institute for Intelligent; Systems 70569 Stuttgart Germany
| | - Metin Sitti
- Physical Intelligence Department; Max Planck Institute for Intelligent; Systems 70569 Stuttgart Germany
| |
Collapse
|
66
|
He XT, Wang J, Li X, Yin Y, Sun HH, Chen FM. The Critical Role of Cell Homing in Cytotherapeutics and Regenerative Medicine. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800098] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Xiao-Tao He
- State Key Laboratory of Military Stomatology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- National Clinical Research Center for Oral Diseases; Department of Periodontology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- Shaanxi Engineering Research Center for Dental Materials, and Advanced Manufacture; Biomaterials Unit; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| | - Jia Wang
- State Key Laboratory of Military Stomatology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- Shaanxi Engineering Research Center for Dental Materials, and Advanced Manufacture; Biomaterials Unit; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| | - Xuan Li
- State Key Laboratory of Military Stomatology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- National Clinical Research Center for Oral Diseases; Department of Periodontology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- Shaanxi Engineering Research Center for Dental Materials, and Advanced Manufacture; Biomaterials Unit; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| | - Yuan Yin
- State Key Laboratory of Military Stomatology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- Shaanxi Engineering Research Center for Dental Materials, and Advanced Manufacture; Biomaterials Unit; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| | - Hai-Hua Sun
- National Clinical Research Center for Oral Diseases; Department of Periodontology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| | - Fa-Ming Chen
- State Key Laboratory of Military Stomatology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- National Clinical Research Center for Oral Diseases; Department of Periodontology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- Shaanxi Engineering Research Center for Dental Materials, and Advanced Manufacture; Biomaterials Unit; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| |
Collapse
|
67
|
Zhao L, Hu C, Zhang P, Jiang H, Chen J. Novel preconditioning strategies for enhancing the migratory ability of mesenchymal stem cells in acute kidney injury. Stem Cell Res Ther 2018; 9:225. [PMID: 30139368 PMCID: PMC6108125 DOI: 10.1186/s13287-018-0973-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Acute kidney injury (AKI) remains a worldwide public health issue due to its increasing incidence, significant mortality, and lack of specific target-orientated therapy. Developments in mesenchymal stem cell (MSC) research make MSCs a promising candidate for AKI management but relevant clinical trials show confusing results (NCT00733876, NCT01602328). One primary cause of the limited therapeutic effect may result from poor engraftment of transplanted cells. To solve this problem, investigators have developed a series of preconditioning strategies to improve MSC engraftment in animal AKI models. In this review, we summarize these previous studies, providing an integrated and updated view of different preconditioning strategies aimed at promoting the therapeutic effect of MSCs in AKI patients.
Collapse
Affiliation(s)
- Lingfei Zhao
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang Province, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Chenxia Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Ping Zhang
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang Province, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Hua Jiang
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang Province, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jianghua Chen
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China. .,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang Province, People's Republic of China. .,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
68
|
Bauer G, Elsallab M, Abou-El-Enein M. Concise Review: A Comprehensive Analysis of Reported Adverse Events in Patients Receiving Unproven Stem Cell-Based Interventions. Stem Cells Transl Med 2018; 7:676-685. [PMID: 30063299 PMCID: PMC6127222 DOI: 10.1002/sctm.17-0282] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 05/18/2018] [Accepted: 05/22/2018] [Indexed: 02/06/2023] Open
Abstract
The promise of stem cell (SC) therapies to restore functions of damaged tissues and organs brings enormous hope to patients, their families, loved ones, and caregivers. However, limits may exist for which indications SC therapies might be useful, efficacious, and safe. Applications of innovative therapies within regulatory boundaries and within the framework of controlled clinical trials are the norm in the scientific and medical community; such a system minimizes patient risk by setting a clear and acceptable safety and efficacy profile for new therapeutics before marketing authorization. This careful clinical validation approach often takes time, which patients suffering from terminal or debilitating diseases do not have. Not validated, unproven stem cell interventions (SCI) that promise a working treatment or cure for severe diseases have therefore found their way into the patient community, and providers of such treatments often take advantage of the public's willingness to pay large amounts of money for the misguided hope of a reliable recovery from their illnesses. We conducted a review of scientific publications, clinical case reports, and mass media publications to assess the reported cases and safety incidents associated with unproven SCI. The review also analyzes the main factors that were identified as contributing to the emergence and global rise of the “stem cell tourism” phenomenon. stemcellstranslationalmedicine2018;1–10
Collapse
Affiliation(s)
- Gerhard Bauer
- University of California Davis, Institute For Regenerative Cures (IRC), Sacramento, California, USA
| | - Magdi Elsallab
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charite' - Universitatsmedizin Berlin, Berlin, Germany
| | - Mohamed Abou-El-Enein
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charite' - Universitatsmedizin Berlin, Berlin, Germany
| |
Collapse
|
69
|
Touré SB, Kleiderman E, Knoppers BM. Bridging stem cell research and medicine: a learning health system. Regen Med 2018; 13:741-752. [PMID: 30043682 DOI: 10.2217/rme-2017-0129] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Stem cells may not systematically obey traditional Phase I-IV clinical translation models. In response, various actors have suggested that stem cell-based medical innovation models could catalyze translation instead. Accordingly, calls were made to adopt more permissive approaches to stem cell translation. Yet, the Phase I-IV paradigm remains the standard within the scientific community. This article seeks to advance the stalemated discussions by proposing an alternative model for consideration. In it, we argue that a stem cell-based learning health system may be able to reconcile these two models. Centered on the acceleration of evidence and knowledge flow, a stem cell-based learning health system could maximize patient retention and data follow-up, thereby promoting inclusive system learning and improvement.
Collapse
Affiliation(s)
- Seydina B Touré
- Centre of Genomics & Policy, Department of Human Genetics, McGill University, Montréal, QC H3A 0G1, Canada
| | - Erika Kleiderman
- Centre of Genomics & Policy, Department of Human Genetics, McGill University, Montréal, QC H3A 0G1, Canada
| | - Bartha M Knoppers
- Centre of Genomics & Policy, Department of Human Genetics, McGill University, Montréal, QC H3A 0G1, Canada
| |
Collapse
|
70
|
Suh N, Lee EB. Antioxidant effects of selenocysteine on replicative senescence in human adipose-derived mesenchymal stem cells. BMB Rep 2018; 50:572-577. [PMID: 29065969 PMCID: PMC5720471 DOI: 10.5483/bmbrep.2017.50.11.174] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Indexed: 11/30/2022] Open
Abstract
In most clinical applications, human mesenchymal stem cells (hMSCs) are expanded in large scale before their administration. Prolonged culture in vitro results in cellular senescence-associated phenotypes, including accumulation of reactive oxygen species (ROS) and decreased cell viabilities. Profiling of stem cell-related genes during in vitro expansion revealed that numerous canonical pathways were significantly changed. To determine the effect of selenocysteine (Sec), a rare amino acid found in several antioxidant enzymes, on the replicative senescence in hMSCs, we treated senescent hMSCs with Sec. Supplementation of Sec in the culture medium in late-passage hMSCs reduced ROS levels and improved the survival of hMSCs. In addition, a subset of key antioxidant genes and Sec-containing selenoproteins showed increased mRNA levels after Sec treatment. Furthermore, ROS metabolism and inflammation pathways were predicted to be downregulated. Taken together, our results suggest that Sec has antioxidant effects on the replicative senescence of hMSCs.
Collapse
Affiliation(s)
- Nayoung Suh
- Department of Pharmaceutical Engineering, Soon Chun Hyang University, Asan 31538, Korea
| | - Eun-Bi Lee
- Department of Pharmaceutical Engineering, Soon Chun Hyang University, Asan 31538, Korea
| |
Collapse
|
71
|
von Wunster B, Bailey S, Wilkins A, Marks DI, Scolding NJ, Rice CM. Advising patients seeking stem cell interventions for multiple sclerosis. Pract Neurol 2018; 18:472-476. [PMID: 29848512 DOI: 10.1136/practneurol-2018-001956] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2018] [Indexed: 12/29/2022]
Abstract
Given the intuitive potential of stem cell therapy and limitations of current treatment options for progressive multiple sclerosis (MS), it is not surprising that patients consider undertaking significant clinical and financial risks to access stem cell transplantation. However, while increasing evidence supports autologous haematopoietic stem cell transplantation (AHSCT) in aggressive relapsing-remitting MS, interventions employing haematopoietic or other stem cells should otherwise be considered experimental and recommended only in the context of a properly regulated clinical study. Understandably, most neurologists are unfamiliar with AHSCT procedures and the specific requirements for quality assurance and safety standards, as well as post-procedure precautions and follow-up. Consequently they may feel ill-equipped to advise patients. Here, we highlight important points for discussion in consultations with patients considering stem cell 'tourism' for MS.
Collapse
Affiliation(s)
- Beatrice von Wunster
- Clinical Neurosciences, Translational Health Sciences, University of Bristol, Bristol, UK.,School of Medicine, Vita-Salute san Raffaele University, Milan, Italy
| | - Steven Bailey
- Clinical Neurosciences, Translational Health Sciences, University of Bristol, Bristol, UK.,Bristol and Avon MS Unit, Bristol Brain Centre, North Bristol NHS Trust, Southmead Hospital, Bristol, UK
| | - Alastair Wilkins
- Clinical Neurosciences, Translational Health Sciences, University of Bristol, Bristol, UK.,Bristol and Avon MS Unit, Bristol Brain Centre, North Bristol NHS Trust, Southmead Hospital, Bristol, UK
| | - David I Marks
- Department of Haematology, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Neil J Scolding
- Clinical Neurosciences, Translational Health Sciences, University of Bristol, Bristol, UK.,Bristol and Avon MS Unit, Bristol Brain Centre, North Bristol NHS Trust, Southmead Hospital, Bristol, UK
| | - Claire M Rice
- Clinical Neurosciences, Translational Health Sciences, University of Bristol, Bristol, UK.,Bristol and Avon MS Unit, Bristol Brain Centre, North Bristol NHS Trust, Southmead Hospital, Bristol, UK
| |
Collapse
|
72
|
Duncan T, Lowe A, Sidhu K, Sachdev P, Lewis T, Lin RCY, Sytnyk V, Valenzuela M. Replicable Expansion and Differentiation of Neural Precursors from Adult Canine Skin. Stem Cell Reports 2018; 9:557-570. [PMID: 28793248 PMCID: PMC5550271 DOI: 10.1016/j.stemcr.2017.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 07/07/2017] [Accepted: 07/10/2017] [Indexed: 11/28/2022] Open
Abstract
Repopulation of brain circuits by neural precursors is a potential therapeutic strategy for neurodegenerative disorders; however, choice of cell is critical. Previously, we introduced a two-step culture system that generates a high yield of neural precursors from small samples of adult canine skin. Here, we probe their gene and protein expression profiles in comparison with dermal fibroblasts and brain-derived neural stem cells and characterize their neuronal potential. To date, we have produced >50 skin-derived neural precursor (SKN) lines. SKNs can be cultured in a highly replicable fashion and uniformly express a panel of identifying markers. Upon differentiation, they self-upregulate neural specification genes, generating neurons with basic electrophysiological functionality. This unique population of neural precursors, derived from mature skin, overcomes many of the practical issues that have limited clinical translation of alternative cell types. Easily accessible, neuronally committed, and patient specific, SKNs may have potential for the treatment of brain disorders.
Collapse
Affiliation(s)
- Thomas Duncan
- Regenerative Neuroscience Group, Brain and Mind Centre, University of Sydney, Sydney, NSW 2050, Australia
| | - Aileen Lowe
- Regenerative Neuroscience Group, Brain and Mind Centre, University of Sydney, Sydney, NSW 2050, Australia; Stem Cell Laboratory, University of New South Wales, Sydney, NSW 2031, Australia
| | - Kuldip Sidhu
- Stem Cell Laboratory, University of New South Wales, Sydney, NSW 2031, Australia
| | - Perminder Sachdev
- Centre for Healthy Brain Ageing, University of New South Wales, Sydney, NSW 2031, Australia; School of Psychiatry, University of New South Wales, Sydney, NSW 2052, Australia
| | - Trevor Lewis
- School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Ruby C Y Lin
- School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Vladimir Sytnyk
- School of Biotechnology and Biomolecular Science, University of New South Wales, Sydney, NSW 2052, Australia
| | - Michael Valenzuela
- Regenerative Neuroscience Group, Brain and Mind Centre, University of Sydney, Sydney, NSW 2050, Australia.
| |
Collapse
|
73
|
Pharmacological activation of TAZ enhances osteogenic differentiation and bone formation of adipose-derived stem cells. Stem Cell Res Ther 2018. [PMID: 29514703 PMCID: PMC5842656 DOI: 10.1186/s13287-018-0799-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Adipose-derived stem cells (ADSCs) are an attractive cell source for bone tissue engineering and have great potential for bone regeneration and defect repair. The transcriptional coactivator with PDZ-binding motif (TAZ) has been demonstrated to modulate osteogenic and adipogenic differentiation of mesenchymal stem cells. However, its roles during ADSC differentiation and therapeutic potentials for bone regeneration have as yet not been well established. Methods TAZ expression was measured during osteogenic differentiation of ADSCs in vitro. Both loss-of-function and gain-of-function approaches by TAZ knockdown or enforced overexpression were utilized to determine its functions during osteogenic differentiation of ADSCs. TM-25659, a chemical activator of TAZ, was used to determine whether pharmacological activation of TAZ in ADSCs enhanced osteogenic differentiation in vitro and bone formation in animal models. The molecular mechanisms underlying TAZ in promoting osteogenesis of ADSCs were also explored. Results Increased TAZ expression was observed during osteogenic differentiation of human ADSCs. TAZ knockdown resulted in compromised osteogenic differentiation and enhanced adipogenic differentiation of ADSCs. In contrast, enforced TAZ overexpression yielded increased osteogenic differentiation and bone regeneration in vivo, and impaired adipogenic differentiation of ADSCs. Pharmacological activation of TAZ by its chemical activator TM-25659 facilitated osteogenic differentiation of ADSCs. Noticeably, transient treatment of ADSCs with TM-25659 or intraperitoneal injection of TM-25659 significantly enhanced bone regeneration of ADSCs loaded with porous β-TCP in vivo. Mechanistically, TM-25659 exposure significantly promoted TAZ phosphorylation and nuclear translocation, and potentiated the assembly of the TAZ-Runx2 complex. Subsequently, the TAZ-Runx2 complex was further recruited to the promoter of osteocalcin and in turn enhanced its transcription. Conclusions Our findings indicate that TAZ is a key mediator that promotes ADSC commitment to the osteoblast lineage. Pharmacological activation of TAZ in ADSCs might become a feasible and promising approach to enhance bone regeneration and repair. Electronic supplementary material The online version of this article (10.1186/s13287-018-0799-z) contains supplementary material, which is available to authorized users.
Collapse
|
74
|
Zaman RT, Tuerkcan S, Mahmoudi M, Saito T, Yang PC, Chin FT, McConnell MV, Xing L. Imaging cellular pharmacokinetics of 18F-FDG and 6-NBDG uptake by inflammatory and stem cells. PLoS One 2018; 13:e0192662. [PMID: 29462173 PMCID: PMC5819797 DOI: 10.1371/journal.pone.0192662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 01/26/2018] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Myocardial infarction (MI) causes significant loss of cardiomyocytes, myocardial tissue damage, and impairment of myocardial function. The inability of cardiomyocytes to proliferate prevents the heart from self-regeneration. The treatment for advanced heart failure following an MI is heart transplantation despite the limited availability of the organs. Thus, stem-cell-based cardiac therapies could ultimately prevent heart failure by repairing injured myocardium that reverses cardiomyocyte loss. However, stem-cell-based therapies lack understanding of the mechanisms behind a successful therapy, including difficulty tracking stem cells to provide information on cell migration, proliferation and differentiation. In this study, we have investigated the interaction between different types of stem and inflammatory cells and cell-targeted imaging molecules, 18F-FDG and 6-NBDG, to identify uptake patterns and pharmacokinetics in vitro. METHODS Macrophages (both M1 and M2), human induced pluripotent stem cells (hiPSCs), and human amniotic mesenchymal stem cells (hAMSCs) were incubated with either 18F-FDG or 6-NBDG. Excess radiotracer and fluorescence were removed and a 100 μm-thin CdWO4 scintillator plate was placed on top of the cells for radioluminescence microscopy imaging of 18F-FDG uptake, while no scintillator was needed for fluorescence imaging of 6-NBDG uptake. Light produced following beta decay was imaged with a highly sensitive inverted microscope (LV200, Olympus) and an Electron Multiplying Charge-Couple Device (EM-CCD) camera. Custom-written software was developed in MATLAB for image processing. RESULTS The average cellular activity of 18F-FDG in a single cell of hAMSCs (0.670±0.028 fCi/μm2, P = 0.001) was 20% and 36% higher compared to uptake in hiPSCs (0.540±0.026 fCi/μm2, P = 0.003) and macrophages (0.430±0.023 fCi/μm2, P = 0.002), respectively. hAMSCs exhibited the slowest influx (0.210 min-1) but the fastest efflux (0.327 min-1) rate compared to the other tested cell lines for 18F-FDG. This cell line also has the highest phosphorylation but exhibited the lowest rate of de-phosphorylation. The uptake pattern for 6-NBDG was very different in these three cell lines. The average cellular activity of 6-NBDG in a single cell of macrophages (0.570±0.230 fM/μm2, P = 0.004) was 38% and 14% higher compared to hiPSCs (0.350±0.160 fM/μm2, P = 0.001) and hAMSCs (0.490±0.028 fM/μm2, P = 0.006), respectively. The influx (0.276 min-1), efflux (0.612 min-1), phosphorylation (0.269 min-1), and de-phosphorylation (0.049 min-1) rates were also highest for macrophages compared to the other two tested cell lines. CONCLUSION hAMSCs were found to be 2-3× more sensitive to 18F-FDG molecule compared to hiPSCs/macrophages. However, macrophages exhibited the most sensitivity towards 6-NBDG. Based on this result, hAMSCs targeted with 18F-FDG could be more suitable for understanding the mechanisms behind successful therapy for treating MI patients by gathering information on cell migration, proliferation and differentiation.
Collapse
Affiliation(s)
- Raiyan T. Zaman
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
- Department of Radiation Oncology, Division of Medical Physics, Stanford University School of Medicine, Stanford, CA, United States of America
- * E-mail:
| | - Silvan Tuerkcan
- Department of Radiation Oncology, Division of Medical Physics, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Morteza Mahmoudi
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Toshinobu Saito
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Phillip C. Yang
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Frederick T. Chin
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Michael V. McConnell
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Lei Xing
- Department of Radiation Oncology, Division of Medical Physics, Stanford University School of Medicine, Stanford, CA, United States of America
| |
Collapse
|
75
|
Abstract
The history of stem cell therapies is one of a limited number of clinical applications despite a vast therapeutic potential. Major breakthroughs in stem cell research have not yet enjoyed clinical success—all stem cell therapies bar hematopoietic stem cell transplantations remain experimental. With the increased risk of organ failure and neurodegenerative disease associated with our ability to push the boundaries of life expectancy comes an increased pressure to pioneer novel stem cell-based therapeutic approaches. We conclude that the failure of such therapies to achieve clinical translation stems from the polarising effect of the ethical debate around their use. The intractability of the ethical debate is double edged: legislators not only have placed tighter restrictions on certain stem cell therapies, but do so in favour of less controversial cells which will have worse outcomes for patients. It is by considering this relationship between the politics, ethics and science of stem cells that the reasons for the currently limited clinical significance of stem cell therapies be realised.
Collapse
Affiliation(s)
- Jordan Poulos
- University College London Medical School, London, UK.
| |
Collapse
|
76
|
The survey on cellular and tissue-engineered therapies in Europe and neighboring Eurasian countries in 2014 and 2015. Cytotherapy 2018; 20:1-20. [DOI: 10.1016/j.jcyt.2017.08.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 08/09/2017] [Accepted: 08/09/2017] [Indexed: 01/28/2023]
|
77
|
Chen X, Harkness L, Jia Z, Prowse A, Monteiro MJ, Gray PP. Methods for Expansion of Three-Dimensional Cultures of Human Embryonic Stem Cells Using a Thermoresponsive Polymer. Tissue Eng Part C Methods 2017; 24:146-157. [PMID: 29239281 DOI: 10.1089/ten.tec.2017.0331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) are viewed as promising candidates for applications in regenerative medicine and therapy due to their proliferative and pluripotent properties. However, obtaining clinically significant numbers of hPSCs remains a limiting factor and impedes their use in therapeutic applications. Conventionally, hPSCs are cultured on two-dimensional surfaces coated with a suitable substrate, such as Matrigel™. This method, however, requires a large surface area to generate sufficient cell numbers to meet clinical needs and is therefore impractical as a manufacturing platform for cell expansion. In addition, the use of enzymes for cell detachment and small molecule inhibitors to increase plating efficiency may impact future cell behavior when used for routine subculturing. In this study, we describe a protocol to generate and maintain hPSC aggregates in a three-dimensional suspension culture by utilizing thermoresponsive nanobridges. The property of the polymer used in the nanobridges enables passaging and expansion through a temperature change in combination with mechanically applied shear to dissociate aggregates; thus, we eliminate the need of enzymes or small molecules for cell dissociation and viability, respectively. Utilizing this platform, maintenance of human embryonic stem cells for three continuous passages demonstrated high expression levels in key pluripotent markers.
Collapse
Affiliation(s)
- Xiaoli Chen
- 1 Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland , Brisbane, Australia
| | - Linda Harkness
- 1 Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland , Brisbane, Australia
| | - Zhongfan Jia
- 1 Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland , Brisbane, Australia
| | - Andrew Prowse
- 2 The Garvan Institute of Medical Research , Sydney, Australia
| | - Michael J Monteiro
- 1 Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland , Brisbane, Australia
| | - Peter P Gray
- 1 Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland , Brisbane, Australia
| |
Collapse
|
78
|
Abstract
Every year 13.3 million people suffer acute kidney injury (AKI), which is associated with a high risk of death or development of long-term chronic kidney disease (CKD) in a substantial percentage of patients besides other organ dysfunctions. To date, the mortality rate per year for AKI exceeds 50 % at least in patients requiring early renal replacement therapy and is higher than the mortality for breast and prostate cancer, heart failure and diabetes combined.Until now, no effective treatments able to accelerate renal recovery and improve survival post AKI have been developed. In search of innovative and effective strategies to foster the limited regeneration capacity of the kidney, several studies have evaluated the ability of mesenchymal stem cells (MSCs) of different origin as an attractive therapeutic tool. The results obtained in several models of AKI and CKD document that MSCs have therapeutic potential in repair of renal injury, preserving renal function and structure thus prolonging animal survival through differentiation-independent pathways. In this chapter, we have summarized the mechanisms underlying the regenerative processes triggered by MSC treatment, essentially due to their paracrine activity. The capacity of MSC to migrate to the site of injury and to secrete a pool of growth factors and cytokines with anti-inflammatory, mitogenic, and immunomodulatory effects is described. New modalities of cell-to-cell communication via the release of microvesicles and exosomes by MSCs to injured renal cells will also be discussed. The translation of basic experimental data on MSC biology into effective care is still limited to preliminary phase I clinical trials and further studies are needed to definitively assess the efficacy of MSC-based therapy in humans.
Collapse
Affiliation(s)
- Marina Morigi
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126, Bergamo, Italy.
| | - Cinzia Rota
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126, Bergamo, Italy
| | - Giuseppe Remuzzi
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126, Bergamo, Italy
- Unit of Nephrology and Dialysis, A.O. Papa Giovanni XXIII, 24127, Bergamo, Italy
| |
Collapse
|
79
|
Li X, He X, Yin Y, Wu R, Tian B, Chen F. Administration of signalling molecules dictates stem cell homing for in situ regeneration. J Cell Mol Med 2017; 21:3162-3177. [PMID: 28767189 PMCID: PMC5706509 DOI: 10.1111/jcmm.13286] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 05/29/2017] [Indexed: 12/13/2022] Open
Abstract
Ex vivo-expanded stem cells have long been a cornerstone of biotherapeutics and have attracted increasing attention for treating intractable diseases and improving tissue regeneration. However, using exogenous cellular materials to develop restorative treatments for large numbers of patients has become a major concern for both economic and safety reasons. Advances in cell biological research over the past two decades have expanded the potential for using endogenous stem cells during wound healing processes, and in particular, recent insight into stem cell movement and homing has prompted regenerative research and therapy based on recruiting endogenous cells. Inspired by the natural healing process, artificial administration of specific chemokines as signals systemically or at the injury site, typically using biomaterials as vehicles, is a state-of-the-art strategy that potentiates stem cell homing and recreates an anti-inflammatory and immunomodulatory microenvironment to enhance in situ tissue regeneration. However, pharmacologically coaxing endogenous stem cells to act as therapeutics in the field of biomedicine remains in the early stages; its efficacy is limited by the lack of innovative methodologies for chemokine presentation and release. This review describes how to direct the homing of endogenous stem cells via the administration of specific signals, with a particular emphasis on targeted signalling molecules that regulate this homing process, to enhance in situ tissue regeneration. We also provide an outlook on and critical considerations for future investigations to enhance stem cell recruitment and harness the reparative potential of these recruited cells as a clinically relevant cell therapy.
Collapse
Affiliation(s)
- Xuan Li
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral DiseasesDepartment of PeriodontologySchool of StomatologyFourth Military Medical UniversityXi'anChina
| | - Xiao‐Tao He
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral DiseasesDepartment of PeriodontologySchool of StomatologyFourth Military Medical UniversityXi'anChina
| | - Yuan Yin
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral DiseasesDepartment of PeriodontologySchool of StomatologyFourth Military Medical UniversityXi'anChina
| | - Rui‐Xin Wu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral DiseasesDepartment of PeriodontologySchool of StomatologyFourth Military Medical UniversityXi'anChina
| | - Bei‐Min Tian
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral DiseasesDepartment of PeriodontologySchool of StomatologyFourth Military Medical UniversityXi'anChina
| | - Fa‐Ming Chen
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral DiseasesDepartment of PeriodontologySchool of StomatologyFourth Military Medical UniversityXi'anChina
| |
Collapse
|
80
|
Baldari S, Di Rocco G, Piccoli M, Pozzobon M, Muraca M, Toietta G. Challenges and Strategies for Improving the Regenerative Effects of Mesenchymal Stromal Cell-Based Therapies. Int J Mol Sci 2017; 18:2087. [PMID: 28974046 PMCID: PMC5666769 DOI: 10.3390/ijms18102087] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/18/2017] [Accepted: 09/28/2017] [Indexed: 12/11/2022] Open
Abstract
Cell-based therapies have the potential to revolutionize current treatments for diseases with high prevalence and related economic and social burden. Unfortunately, clinical trials have made only modest improvements in restoring normal function to degenerating tissues. This limitation is due, at least in part, to the death of transplanted cells within a few hours after transplant due to a combination of mechanical, cellular, and host factors. In particular, mechanical stress during implantation, extracellular matrix loss upon delivery, nutrient and oxygen deprivation at the recipient site, and host inflammatory response are detrimental factors limiting long-term transplanted cell survival. The beneficial effect of cell therapy for regenerative medicine ultimately depends on the number of administered cells reaching the target tissue, their viability, and their promotion of tissue regeneration. Therefore, strategies aiming at improving viable cell engraftment are crucial for regenerative medicine. Here we review the major factors that hamper successful cell engraftment and the strategies that have been studied to enhance the beneficial effects of cell therapy. Moreover, we provide a perspective on whether mesenchymal stromal cell-derived extracellular vesicle delivery, as a cell-free regenerative approach, may circumvent current cell therapy limitations.
Collapse
Affiliation(s)
- Silvia Baldari
- Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute, via E. Chianesi 53, Rome 00144, Italy.
| | - Giuliana Di Rocco
- Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute, via E. Chianesi 53, Rome 00144, Italy.
| | - Martina Piccoli
- Stem Cells and Regenerative Medicine Laboratory, Foundation Institute of Pediatric Research "Città della Speranza", corso Stati Uniti 4, Padova 35127, Italy.
| | - Michela Pozzobon
- Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, Padova 35128, Italy.
| | - Maurizio Muraca
- Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, Padova 35128, Italy.
| | - Gabriele Toietta
- Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute, via E. Chianesi 53, Rome 00144, Italy.
| |
Collapse
|
81
|
Devireddy LR, Boxer L, Myers MJ, Skasko M, Screven R. Questions and Challenges in the Development of Mesenchymal Stromal/Stem Cell-Based Therapies in Veterinary Medicine. TISSUE ENGINEERING PART B-REVIEWS 2017; 23:462-470. [DOI: 10.1089/ten.teb.2016.0451] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Lax R. Devireddy
- Division of Applied Veterinary Research, Center for Veterinary Medicine, US Food and Drug Administration, Laurel, Maryland
| | - Lynne Boxer
- Office of New Animal Drug Evaluation, Center for Veterinary Medicine, US Food and Drug Administration, Rockville, Maryland
| | - Michael J. Myers
- Division of Applied Veterinary Research, Center for Veterinary Medicine, US Food and Drug Administration, Laurel, Maryland
| | - Mark Skasko
- Office of New Animal Drug Evaluation, Center for Veterinary Medicine, US Food and Drug Administration, Rockville, Maryland
| | - Rudell Screven
- Division of Applied Veterinary Research, Center for Veterinary Medicine, US Food and Drug Administration, Laurel, Maryland
| |
Collapse
|
82
|
Regulation of Osteogenic Differentiation of Placental-Derived Mesenchymal Stem Cells by Insulin-Like Growth Factors and Low Oxygen Tension. Stem Cells Int 2017; 2017:4576327. [PMID: 29138637 PMCID: PMC5613461 DOI: 10.1155/2017/4576327] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 07/20/2017] [Indexed: 01/20/2023] Open
Abstract
Placental mesenchymal stem cells (PMSCs) are multipotent cells that can differentiate in vitro to multiple lineages, including bone. Insulin-like growth factors (IGFs, IGF-1 and IGF-2) participate in maintaining growth, survival, and differentiation of many stem cells, including osteoprogenitors. Low oxygen tension (PO2) can maintain stem cell multipotency and impede osteogenic differentiation. In this study, we investigated whether PMSC osteogenic differentiation is influenced by low PO2 and by IGFs. Our results indicated that low PO2 decreased osteogenic markers RUNX2 and OPN; however, re-exposure to higher oxygen tension (room air) restored differentiation. IGFs, especially IGF-1, triggered an earlier expression of RUNX2 and enhanced OPN and mineralization. RUNX2 was phosphorylated in room air and augmented by IGFs. IGF-1 receptor (IGF-1R) was increased in low PO2 and reduced by IGFs, while insulin receptor (IR) was increased in differentiating PMSCs and enhanced by IGF-1. Low PO2 and IGFs maintained higher IR-A which was switched to IR-B in room air. PI3K/AKT was required for osteogenic differentiation, while MEK/ERK was required to repress an RUNX2 and OPN increase in low PO2. Therefore, IGFs, specifically IGF-1, trigger the earlier onset of osteogenic differentiation in room air, whereas, reversibly, low PO2 impedes complete differentiation by maintaining higher multipotency and lower differentiation markers.
Collapse
|
83
|
McKee C, Chaudhry GR. Advances and challenges in stem cell culture. Colloids Surf B Biointerfaces 2017; 159:62-77. [PMID: 28780462 DOI: 10.1016/j.colsurfb.2017.07.051] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 07/04/2017] [Accepted: 07/22/2017] [Indexed: 12/12/2022]
Abstract
Stem cells (SCs) hold great promise for cell therapy, tissue engineering, and regenerative medicine as well as pharmaceutical and biotechnological applications. They have the capacity to self-renew and the ability to differentiate into specialized cell types depending upon their source of isolation. However, use of SCs for clinical applications requires a high quality and quantity of cells. This necessitates large-scale expansion of SCs followed by efficient and homogeneous differentiation into functional derivatives. Traditional methods for maintenance and expansion of cells rely on two-dimensional (2-D) culturing techniques using plastic culture plates and xenogenic media. These methods provide limited expansion and cells tend to lose clonal and differentiation capacity upon long-term passaging. Recently, new approaches for the expansion of SCs have emphasized three-dimensional (3-D) cell growth to mimic the in vivo environment. This review provides a comprehensive compendium of recent advancements in culturing SCs using 2-D and 3-D techniques involving spheroids, biomaterials, and bioreactors. In addition, potential challenges to achieve billion-fold expansion of cells are discussed.
Collapse
Affiliation(s)
- Christina McKee
- Department of Biological Sciences , Oakland University, Rochester, MI, 48309, USA; OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA
| | - G Rasul Chaudhry
- Department of Biological Sciences , Oakland University, Rochester, MI, 48309, USA; OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA.
| |
Collapse
|
84
|
|
85
|
Lysaght T, Kerridge IH, Sipp D, Porter G, Capps BJ. Ethical and Regulatory Challenges with Autologous Adult Stem Cells: A Comparative Review of International Regulations. JOURNAL OF BIOETHICAL INQUIRY 2017; 14:261-273. [PMID: 28247202 DOI: 10.1007/s11673-017-9776-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 10/24/2016] [Indexed: 06/06/2023]
Abstract
Cell and tissue-based products, such as autologous adult stem cells, are being prescribed by physicians across the world for diseases and illnesses that they have neither been approved for or been demonstrated as safe and effective in formal clinical trials. These doctors often form part of informal transnational networks that exploit differences and similarities in the regulatory systems across geographical contexts. In this paper, we examine the regulatory infrastructure of five geographically diverse but socio-economically comparable countries with the aim of identifying similarities and differences in how these products are regulated and governed within clinical contexts. We find that while there are many subtle technical differences in how these regulations are implemented, they are sufficiently similar that it is difficult to explain why these practices appear more prevalent in some countries and not in others. We conclude with suggestions for how international governance frameworks might be improved to discourage the exploitation of vulnerable patient populations while enabling innovation in the clinical application of cellular therapies.
Collapse
Affiliation(s)
- Tamra Lysaght
- Centre for Biomedical Ethics, National University of Singapore, Level 2 Block MD11, Clinical Research Centre, 10 Medical Drive, 117576, Singapore, Singapore.
| | - Ian H Kerridge
- Centre for Values, Ethics and the Law in Medicine, University of Sydney, Medical Foundations Building K25, Sydney, NSW, 2006, Australia
| | - Douglas Sipp
- RIKEN Centre for Developmental Biology, 2-2-3 Minatojima-minamimachi Chuou-ku, Kobe, 650-004, Japan
| | - Gerard Porter
- School of Law, Edinburgh University, Old College, South Bridge, Edinburgh, EH8 9YL, Scotland
| | - Benjamin J Capps
- Department of Bioethics, Dalhousie University, 5849 University Avenue, Room C-315, CRC Bldg, PO Box 15000, Halifax, NS, B3H 4R2, Canada
| |
Collapse
|
86
|
Li CL, Leng Y, Zhao B, Gao C, Du FF, Jin N, Lian QZ, Xu SY, Yan GL, Xia JJ, Zhuang GH, Fu QL, Qi ZQ. Human iPSC-MSC-Derived Xenografts Modulate Immune Responses by Inhibiting the Cleavage of Caspases. Stem Cells 2017; 35:1719-1732. [PMID: 28520232 DOI: 10.1002/stem.2638] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 04/05/2017] [Accepted: 04/20/2017] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells (MSCs) negatively modulate immune properties. Induced pluripotent stem cells (iPSCs)-derived MSCs are alternative source of MSCs. However, the effects of iPSC-MSCs on T cells phenotypes in vivo remain unclear. We established an iPSC-MSC-transplanted host versus graft reaction mouse model using subcapsular kidney injection. Th1, Th2, regulatory T cells (Treg), and Th17 phenotypes and their cytokines were investigated in vivo and in vitro. The role of caspases and the soluble factors involved in the effects of MSCs were examined. We found that iPSC-MSC grafts led to more cell survival and less infiltration of inflammatory cells in mice. iPSC-MSC transplantation inhibited T cell proliferation, decreased Th1 and Th2 phenotypes and cytokines, upregulated Th17 and Treg subsets. Moreover, iPSC-MSCs inhibited the cleavage of caspases 3 and 8 and inhibition of caspases downregulated Th1, Th2 responses and upregulated Th17, Treg responses. Soluble factors were determined using protein array and TGF-β1/2/3, IL-10, and MCP-1 were found to be highly expressed in iPSC-MSCs. The administration of the soluble factors decreased Th1/2 response, upregulated Treg response and inhibited the cleavage of caspases. Our results demonstrate that iPSC-MSCs regulate T cell responses as a result of a combined action of the above soluble factors secreted by iPSC-MSCs. These factors suppress T cell responses by inhibiting the cleavage of caspases. These data provide a novel immunomodulatory mechanism for the underlying iPSC-MSC-based immunomodulatory effects on T cell responses. Stem Cells 2017;35:1719-1732.
Collapse
Affiliation(s)
- Cheng-Lin Li
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen, Fujian, People's Republic of China.,Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yun Leng
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen, Fujian, People's Republic of China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, Fujian, People's Republic of China.,Xiamen Key Laboratory of Regenerative Medicine, Xiamen, Fujian, People's Republic of China
| | - Bin Zhao
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen, Fujian, People's Republic of China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, Fujian, People's Republic of China.,Xiamen Key Laboratory of Regenerative Medicine, Xiamen, Fujian, People's Republic of China
| | - Chang Gao
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen, Fujian, People's Republic of China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, Fujian, People's Republic of China.,Xiamen Key Laboratory of Regenerative Medicine, Xiamen, Fujian, People's Republic of China
| | - Fei-Fei Du
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen, Fujian, People's Republic of China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, Fujian, People's Republic of China.,Xiamen Key Laboratory of Regenerative Medicine, Xiamen, Fujian, People's Republic of China
| | - Ning Jin
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen, Fujian, People's Republic of China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, Fujian, People's Republic of China.,Xiamen Key Laboratory of Regenerative Medicine, Xiamen, Fujian, People's Republic of China
| | - Qi-Zhou Lian
- Department of Ophthalmology, and Department of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, People's Republic of China
| | - Shuang-Yue Xu
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen, Fujian, People's Republic of China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, Fujian, People's Republic of China.,Xiamen Key Laboratory of Regenerative Medicine, Xiamen, Fujian, People's Republic of China
| | - Guo-Liang Yan
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen, Fujian, People's Republic of China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, Fujian, People's Republic of China.,Xiamen Key Laboratory of Regenerative Medicine, Xiamen, Fujian, People's Republic of China
| | - Jun-Jie Xia
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen, Fujian, People's Republic of China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, Fujian, People's Republic of China.,Xiamen Key Laboratory of Regenerative Medicine, Xiamen, Fujian, People's Republic of China
| | - Guo-Hong Zhuang
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen, Fujian, People's Republic of China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, Fujian, People's Republic of China.,Xiamen Key Laboratory of Regenerative Medicine, Xiamen, Fujian, People's Republic of China
| | - Qing-Ling Fu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhong-Quan Qi
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen, Fujian, People's Republic of China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, Fujian, People's Republic of China.,Xiamen Key Laboratory of Regenerative Medicine, Xiamen, Fujian, People's Republic of China
| |
Collapse
|
87
|
A Hyper-Crosslinked Carbohydrate Polymer Scaffold Facilitates Lineage Commitment and Maintains a Reserve Pool of Proliferating Cardiovascular Progenitors. Transplant Direct 2017; 3:e153. [PMID: 28573188 PMCID: PMC5441984 DOI: 10.1097/txd.0000000000000667] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 02/12/2017] [Indexed: 12/17/2022] Open
Abstract
Background Cardiovascular progenitor cells (CPCs) have been cultured on various scaffolds to resolve the challenge of cell retention after transplantation and to improve functional outcome after cell-based cardiac therapy. Previous studies have reported successful culture of fully differentiated cardiomyocytes on scaffolds of various types, and ongoing efforts are focused on optimizing the mix of cardiomyocytes and endothelial cells as well as on the identification of a source of progenitors capable of reversing cardiovascular damage. A scaffold culture that fosters cell differentiation into cardiomyocytes and endothelial cells while maintaining a progenitor reserve would benefit allogeneic cell transplantation. Methods Isl-1 + c-Kit + CPCs were isolated as clonal populations from human and sheep heart tissue. After hyper-crosslinked carbohydrate polymer scaffold culture, cells were assessed for differentiation, intracellular signaling, cell cycling, and growth factor/chemokine expression using real time polymerase chain reaction, flow cytometry, immunohistochemistry, and calcium staining. Results Insulin-like growth factor 1, hepatocyte growth factor, and stromal cell derived factor 1α paracrine factors were induced, protein kinase B signaling was activated, extracellular signal-regulated kinase phosphorylation was reduced and differentiation into both cardiomyocytes and endothelial cells was induced by scaffold-based cell culture. Interestingly, movement of CPCs out of the G1 phase of the cell cycle and increased expression of pluripotency genes PLOU5F1 (Oct4) and T (Brachyury) within a portion of the cultured population occurred, which suggests the maintenance of a progenitor population. Two-color immunostaining and 3-color fluorescence-activated cell sorting analysis confirmed the presence of both Isl-1 expressing undifferentiated cells and differentiated cells identified by troponin T and von Willebrand factor expression. Ki-67 labeling verified the presence of proliferating cells that remained in situ alongside the differentiated functional derivatives. Conclusions Cloned Isl-1 + c-kit + CPCs maintained on a hyper-cross linked polymer scaffold retain dual potential for proliferation and differentiation, providing a scaffold-based stem cell source for transplantation of committed and proliferating cardiovascular progenitors for functional testing in preclinical models of cell-based repair.
Collapse
|
88
|
Liu Y, Zhang C, Li Z, Wang C, Jia J, Gao T, Hildebrandt G, Zhou D, Bondada S, Ji P, St Clair D, Liu J, Zhan C, Geiger H, Wang S, Liang Y. Latexin Inactivation Enhances Survival and Long-Term Engraftment of Hematopoietic Stem Cells and Expands the Entire Hematopoietic System in Mice. Stem Cell Reports 2017; 8:991-1004. [PMID: 28330618 PMCID: PMC5390104 DOI: 10.1016/j.stemcr.2017.02.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 02/09/2017] [Accepted: 02/10/2017] [Indexed: 12/20/2022] Open
Abstract
Natural genetic diversity offers an important yet largely untapped resource to decipher the molecular mechanisms regulating hematopoietic stem cell (HSC) function. Latexin (Lxn) is a negative stem cell regulatory gene identified on the basis of genetic diversity. By using an Lxn knockout mouse model, we found that Lxn inactivation in vivo led to the physiological expansion of the entire hematopoietic hierarchy. Loss of Lxn enhanced the competitive repopulation capacity and survival of HSCs in a cell-intrinsic manner. Gene profiling of Lxn-null HSCs showed altered expression of genes enriched in cell-matrix and cell-cell interactions. Thrombospondin 1 (Thbs1) was a potential downstream target with a dramatic downregulation in Lxn-null HSCs. Enforced expression of Thbs1 restored the Lxn inactivation-mediated HSC phenotypes. This study reveals that Lxn plays an important role in the maintenance of homeostatic hematopoiesis, and it may lead to development of safe and effective approaches to manipulate HSCs for clinical benefit.
Collapse
Affiliation(s)
- Yi Liu
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Cuiping Zhang
- Department of Toxicology and Cancer Biology, University of Kentucky, Health Sciences Research Building Room 340, 1095 V.A. Drive, Lexington, KY 40536, USA
| | - Zhenyu Li
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Chi Wang
- Department of Cancer Biostatistics, University of Kentucky, Lexington, KY 40536, USA
| | - Jianhang Jia
- Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Tianyan Gao
- Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Gerhard Hildebrandt
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Daohong Zhou
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Subbarao Bondada
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| | - Peng Ji
- Department of Pathology, Northwestern University, Chicago, IL 60611, USA
| | - Daret St Clair
- Department of Toxicology and Cancer Biology, University of Kentucky, Health Sciences Research Building Room 340, 1095 V.A. Drive, Lexington, KY 40536, USA
| | - Jinze Liu
- Department of Computer Science, University of Kentucky, Lexington, KY 40536, USA
| | - Changguo Zhan
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Hartmut Geiger
- Cincinnati Children's Hospital Medical Center, Experimental Hematology and Cancer Biology, Cincinnati, OH 45229, USA; Institute for Molecular Medicine, University of Ulm, 89081 Ulm, Germany
| | - Shuxia Wang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Ying Liang
- Department of Toxicology and Cancer Biology, University of Kentucky, Health Sciences Research Building Room 340, 1095 V.A. Drive, Lexington, KY 40536, USA.
| |
Collapse
|
89
|
DNA repair and replication links to pluripotency and differentiation capacity of pig iPS cells. PLoS One 2017; 12:e0173047. [PMID: 28253351 PMCID: PMC5333863 DOI: 10.1371/journal.pone.0173047] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 02/14/2017] [Indexed: 01/05/2023] Open
Abstract
Pigs are proposed to be suitable large animal models for test of the efficacy and safety of induced pluripotent stem cells (iPSCs) for stem cell therapy, but authentic pig ES/iPS cell lines with germline competence are rarely produced. The pathways or signaling underlying the defective competent pig iPSCs remain poorly understood. By improving induction conditions using various small chemicals, we generated pig iPSCs that exhibited high pluripotency and differentiation capacity that can contribute to chimeras. However, their potency was reduced with increasing passages by teratoma formation test, and correlated with declined expression levels of Rex1, an important marker for naïve state. By RNA-sequencing analysis, genes related to WNT signaling were upregulated and MAPK signaling and TGFβ pathways downregulated in pig iPSCs compared to fibroblasts, but they were abnormally expressed during passages. Notably, pathways involving in DNA repair and replication were upregulated at early passage, but downregulated in iPSCs during prolonged passage in cluster with fibroblasts. Our data suggests that reduced DNA repair and replication capacity links to the instability of pig iPSCs. Targeting these pathways may facilitate generation of truly pluripotent pig iPSCs, with implication in translational studies.
Collapse
|
90
|
The Roles of Insulin-Like Growth Factors in Mesenchymal Stem Cell Niche. Stem Cells Int 2017; 2017:9453108. [PMID: 28298931 PMCID: PMC5337393 DOI: 10.1155/2017/9453108] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 12/22/2016] [Accepted: 01/18/2017] [Indexed: 12/12/2022] Open
Abstract
Many tissues contain adult mesenchymal stem cells (MSCs), which may be used in tissue regeneration therapies. However, the MSC availability in most tissues is limited which demands expansion in vitro following isolation. Like many developing cells, the state of MSCs is affected by the surrounding microenvironment, and mimicking this natural microenvironment that supports multipotent or differentiated state in vivo is essential to understand for the successful use of MSC in regenerative therapies. Many researchers are, therefore, optimizing cell culture conditions in vitro by altering growth factors, extracellular matrices, chemicals, oxygen tension, and surrounding pH to enhance stem cells self-renewal or differentiation. Insulin-like growth factors (IGFs) system has been demonstrated to play an important role in stem cell biology to either promote proliferation and self-renewal or enhance differentiation onset and outcome, depending on the cell culture conditions. In this review, we will describe the importance of IGFs, IGF-1 and IGF-2, in development and in the MSC niche and how they affect the pluripotency or differentiation towards multiple lineages of the three germ layers.
Collapse
|
91
|
Farajkhoda T. An overview on ethical considerations in stem cell research in Iran and ethical recommendations: A review. Int J Reprod Biomed 2017; 15:67-74. [PMID: 28462397 PMCID: PMC5405218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Conducting research on the stem cell lines might bring some worthy good to public. Human Stem Cells (hSCs) research has provided opportunities for scientific progresses and new therapies, but some complex ethical matters should be noticed to ensure that stem cell research is carried out in an ethically appropriate manner. The aim of this review article is to discuss the importance of stem cell research, code of ethics for stem cell research in Iran and ethical recommendation. Generation of stem cells for research from human embryo or adult stem cells, saving, maintenance and using of them are the main ethical, legal and jurisprudence concerns in Iran. Concerns regarding human reproduction or human cloning, breach of human dignity, genetic manipulation and probability of tumorogenisity are observed in adult/somatic stem cells. Destruction of embryo to generate stem cell is an important matter in Iran. In this regards, obtaining stem cell from donated frozen embryos through infertility treatment that would be discarded is an acceptable solution in Iran for generation of embryo for research. Ethical, legal, and jurisprudence strategies for using adult/somatic stem cells are determination of ownership of stem cells, trade prohibition of human body, supervision on bio banks and information of Oversight Committee on Stem Cell Research. Recommendations to handle ethical issues for conducting stem cell research are well-designed studies, compliance codes of ethics in biomedical research (specifically codes of ethics on stem cell research, codes of ethics on clinical trials studies and codes of ethics on animals studies), appropriate collaboration with ethics committees and respecting of rights of participants (including both of human and animal rights) in research. In addition, there is a necessity for extending global networks of bioethics for strengthening communications within organizations at both the regional and international level, strengthening legislation systems, designing and establishing convenient collaborative educational courses at different levels.
Collapse
|
92
|
Canine Adipose Derived Mesenchymal Stem Cells Transcriptome Composition Alterations: A Step towards Standardizing Therapeutic. Stem Cells Int 2017; 2017:4176292. [PMID: 28246532 PMCID: PMC5299202 DOI: 10.1155/2017/4176292] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 12/19/2016] [Accepted: 01/04/2017] [Indexed: 01/09/2023] Open
Abstract
Although canine adipose derived stem cells (cASCs) morphology characteristics and differentiation ability are well documented, transcriptome alterations of undifferentiated cASCs during ex vivo cultivation remain unknown. Here we demonstrate, for the first time, the transcriptome composition of isolated cASCs in undifferentiated state originating from six donors. Transcriptome changes were monitored during ex vivo cultivation between passage 3 (P3) and P5, which are mostly used in therapy. Influence of donors' age in given passage number on transcriptome composition was also investigated. Cultivation from P3 to P5 resulted in 16 differentially expressed genes with cooverexpression of pluripotency and self-renewal transcription factors genes SOX2 and POU5F1 dominant in old donors' cells. Furthermore, cASCs demonstrated upregulation of IL-6 in young and old donors' cells. In addition, ex vivo cultivation of cASCs revealed well-known morphological alterations accompanied with decrease in expression of CD90 and CD44 markers in P4 and higher monitored by flow cytometry and successful osteo- and chondrodifferentiation but inefficient adipodifferentiation in P3. Our results revealed the impact of ex vivo cultivation on nature of cells. Correlation of transcriptome changes with secretome composition is needed and its further impact on therapeutic potential of cASCs remains to be evaluated in clinical trials.
Collapse
|
93
|
Sardo VL, Ferguson W, Erikson GA, Topol EJ, Baldwin KK, Torkamani A. Influence of donor age on induced pluripotent stem cells. Nat Biotechnol 2017; 35:69-74. [PMID: 27941802 PMCID: PMC5505172 DOI: 10.1038/nbt.3749] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 11/14/2016] [Indexed: 12/14/2022]
Abstract
Induced pluripotent stem cells (iPSCs) are being pursued as a source of cells for autologous therapies, many of which will be aimed at aged patients. To explore the impact of age on iPSC quality, we produced iPSCs from blood cells of 16 donors aged 21-100. We find that iPSCs from older donors retain an epigenetic signature of age, which can be reduced through passaging. Clonal expansion via reprogramming also enables the discovery of somatic mutations present in individual donor cells, which are missed by bulk sequencing methods. We show that exomic mutations in iPSCs increase linearly with age, and all iPSC lines analyzed carry at least one gene-disrupting mutation, several of which have been associated with cancer or dysfunction. Unexpectedly, elderly donors (>90 yrs) harbor fewer mutations than predicted, likely due to a contracted blood progenitor pool. These studies establish that donor age is associated with an increased risk of abnormalities in iPSCs and will inform clinical development of reprogramming technology.
Collapse
Affiliation(s)
- Valentina Lo Sardo
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, California
| | - William Ferguson
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Galina A. Erikson
- The Scripps Translational Science Institute, Scripps Health and The Scripps Research Institute, La Jolla, California
| | - Eric J Topol
- The Scripps Translational Science Institute, Scripps Health and The Scripps Research Institute, La Jolla, California
| | - Kristin K Baldwin
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Ali Torkamani
- The Scripps Translational Science Institute, Scripps Health and The Scripps Research Institute, La Jolla, California
| |
Collapse
|
94
|
Chen F, Yao H, Wang M, Yu B, Liu Q, Li J, He Z, Hu YP. Suppressing Pitx2 inhibits proliferation and promotes differentiation of iHepSCs. Int J Biochem Cell Biol 2016; 80:154-162. [PMID: 27697592 DOI: 10.1016/j.biocel.2016.09.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 09/18/2016] [Accepted: 09/29/2016] [Indexed: 01/05/2023]
Abstract
Induced hepatic stem cells (iHepSCs) have great potential as donors for liver cell therapy due to their abilities for self-renewal and bi-potential differentiation. However, the molecular mechanism regulating proliferation and differentiation of iHepSCs is poorly understood. In this study, we provide evidence that the homeodomain transcription factor, Pitx2, is essential to maintain iHepSCs stem cell characteristics. Suppressing Pitx2 expression in iHepSCs by lentivirus mediated specific shRNA markedly reduced the expression of the hepatic stem cell-associated genes (Lgr5, EpCAM, and Sox9) with concomitant inhibition of proliferation by blocking the G1/S phase transition, and these phenotypic changes were reversed upon re-expression of Pitx2. Pitx2 knockdown also resulted in up-regulation of the p53-induced Cdk inhibitor p21, and down-regulation of its downstream effector CDK2-Cyclin E kinase complex. Furthermore, we observed that iHepSCs were more efficiently induced to differentiate into both hepatocytes and cholangiocytes when Pitx2 expression was suppressed, as compared to unmanipulated iHepSCs. These findings reveal that Pitx2 expression may be leveraged to control the status of iHepSCs during expansion in vitro to provide a strategy for further application of iHepSCs in liver cell therapy.
Collapse
Affiliation(s)
- Fei Chen
- Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, PR China
| | - Hao Yao
- Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, PR China
| | - Minjun Wang
- Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, PR China
| | - Bing Yu
- Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, PR China
| | - Qinggui Liu
- Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, PR China
| | - Jianxiu Li
- Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, PR China
| | - Zhiying He
- Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, PR China
| | - Yi-Ping Hu
- Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, PR China.
| |
Collapse
|
95
|
Zhang Y, Wang L, Feng Z, Cheng H, McGuire TF, Ding Y, Cheng T, Gao Y, Xie XQ. StemCellCKB: An Integrated Stem Cell-Specific Chemogenomics KnowledgeBase for Target Identification and Systems-Pharmacology Research. J Chem Inf Model 2016; 56:1995-2004. [PMID: 27643925 DOI: 10.1021/acs.jcim.5b00748] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Given the capacity of self-renewal and multilineage differentiation, stem cells are promising sources for use in regenerative medicines as well as in the clinical treatment of certain hematological malignancies and degenerative diseases. Complex networks of cellular signaling pathways largely determine stem cell fate and function. Small molecules that modulate these pathways can provide important biological and pharmacological insights. However, it is still challenging to identify the specific protein targets of these compounds, to explore the changes in stem cell phenotypes induced by compound treatment and to ascertain compound mechanisms of action. To facilitate stem cell related small molecule study and provide a better understanding of the associated signaling pathways, we have constructed a comprehensive domain-specific chemogenomics resource, called StemCellCKB ( http://www.cbligand.org/StemCellCKB/ ). This new cloud-computing platform describes the chemical molecules, genes, proteins, and signaling pathways implicated in stem cell regulation. StemCellCKB is also implemented with web applications designed specifically to aid in the identification of stem cell relevant protein targets, including TargetHunter, a machine-learning algorithm for predicting small molecule targets based on molecular fingerprints, and HTDocking, a high-throughput docking module for target prediction and systems-pharmacology analyses. We have systematically tested StemCellCKB to verify data integrity. Target-prediction accuracy has also been validated against the reported known target/compound associations. This proof-of-concept example demonstrates that StemCellCKB can (1) accurately predict the macromolecular targets of existing stem cell modulators and (2) identify novel small molecules capable of probing stem cell signaling mechanisms, for use in systems-pharmacology studies. StemCellCKB facilitates the exploration and exchange of stem cell chemogenomics data among members of the broader research community.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; National Center of Excellence for Computational Drug Abuse Research; Drug Discovery Institute; Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania 15260, United States.,Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin 300020, P. R. China
| | - Lirong Wang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; National Center of Excellence for Computational Drug Abuse Research; Drug Discovery Institute; Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania 15260, United States
| | - Zhiwei Feng
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; National Center of Excellence for Computational Drug Abuse Research; Drug Discovery Institute; Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania 15260, United States
| | - Haizi Cheng
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; National Center of Excellence for Computational Drug Abuse Research; Drug Discovery Institute; Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania 15260, United States
| | - Terence Francis McGuire
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; National Center of Excellence for Computational Drug Abuse Research; Drug Discovery Institute; Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania 15260, United States
| | - Yahui Ding
- Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin 300020, P. R. China
| | - Tao Cheng
- Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin 300020, P. R. China
| | - Yingdai Gao
- Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin 300020, P. R. China
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; National Center of Excellence for Computational Drug Abuse Research; Drug Discovery Institute; Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
96
|
Reproducible preparation of spheroids of pancreatic hormone positive cells from human iPS cells: An in vitro study. Biochim Biophys Acta Gen Subj 2016; 1860:2008-16. [DOI: 10.1016/j.bbagen.2016.05.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 04/18/2016] [Accepted: 05/09/2016] [Indexed: 01/13/2023]
|
97
|
Comenge J, Fragueiro O, Sharkey J, Taylor A, Held M, Burton NC, Park BK, Wilm B, Murray P, Brust M, Lévy R. Preventing Plasmon Coupling between Gold Nanorods Improves the Sensitivity of Photoacoustic Detection of Labeled Stem Cells in Vivo. ACS NANO 2016; 10:7106-7116. [PMID: 27308890 DOI: 10.1021/acsnano.6b03246] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Gold nanorods are excellent contrast agents for imaging technologies which rely on near-infrared absorption such as photoacoustic imaging. For cell tracking applications, the cells of interest are labeled with the contrast agent prior to injection. However, after uptake into cells by endocytosis, the confinement and high concentration in endosomes leads to plasmon band broadening and reduced absorbance. This would limit the potential of multispectral optoacoustic tomography in terms of spectral processing and, consequently, sensitivity. Here, we show that steric hindrance provided by silica coating of the nanorods leads to the preservation of their spectral properties and improved photoacoustic sensitivity. This strategy allowed the detection and monitoring of as few as 2 × 10(4) mesenchymal stem cells in mice over a period of 15 days with a high spatial resolution. Importantly, the silica-coated nanorods did not affect the viability or differentiation potential of the transplanted mesenchymal stem cells.
Collapse
Affiliation(s)
- Joan Comenge
- Institute of Integrative Biology, University of Liverpool , Liverpool L69 7ZB, United Kingdom
| | - Oihane Fragueiro
- Department of Chemistry, University of Liverpool , Liverpool L69 7ZD, United Kingdom
| | - Jack Sharkey
- Institute of Translational Medicine, University of Liverpool , Liverpool L69 3BX, United Kingdom
| | - Arthur Taylor
- Institute of Translational Medicine, University of Liverpool , Liverpool L69 3BX, United Kingdom
| | - Marie Held
- Institute of Integrative Biology, University of Liverpool , Liverpool L69 7ZB, United Kingdom
| | - Neal C Burton
- iThera Medical GmbH , Zielstattstrasse 13, 81379 Munich, Germany
| | - Brian Kevin Park
- Institute of Translational Medicine, University of Liverpool , Liverpool L69 3BX, United Kingdom
| | - Bettina Wilm
- Institute of Translational Medicine, University of Liverpool , Liverpool L69 3BX, United Kingdom
| | - Patricia Murray
- Institute of Translational Medicine, University of Liverpool , Liverpool L69 3BX, United Kingdom
| | - Mathias Brust
- Department of Chemistry, University of Liverpool , Liverpool L69 7ZD, United Kingdom
| | - Raphaël Lévy
- Institute of Integrative Biology, University of Liverpool , Liverpool L69 7ZB, United Kingdom
| |
Collapse
|
98
|
Gao M, Yao H, Dong Q, Zhang H, Yang Z, Yang Y, Zhu J, Xu M, Xu R. Tumourigenicity and Immunogenicity of Induced Neural Stem Cell Grafts Versus Induced Pluripotent Stem Cell Grafts in Syngeneic Mouse Brain. Sci Rep 2016; 6:29955. [PMID: 27417157 PMCID: PMC4945932 DOI: 10.1038/srep29955] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 06/24/2016] [Indexed: 01/08/2023] Open
Abstract
Along with the development of stem cell-based therapies for central nervous system (CNS) disease, the safety of stem cell grafts in the CNS, such as induced pluripotent stem cells (iPSCs) and induced neural stem cells (iNSCs), should be of primary concern. To provide scientific basis for evaluating the safety of these stem cells, we determined their tumourigenicity and immunogenicity in syngeneic mouse brain. Both iPSCs and embryonic stem cells (ESCs) were able to form tumours in the mouse brain, leading to tissue destruction along with immune cell infiltration. In contrast, no evidence of tumour formation, brain injury or immune rejection was observed with iNSCs, neural stem cells (NSCs) or mesenchymal stem cells (MSCs). With the help of gene ontology (GO) enrichment analysis, we detected significantly elevated levels of chemokines in the brain tissue and serum of mice that developed tumours after ESC or iPSC transplantation. Moreover, we also investigated the interactions between chemokines and NF-κB signalling and found that NF-κB activation was positively correlated with the constantly rising levels of chemokines, and vice versa. In short, iNSC grafts, which lacked any resulting tumourigenicity or immunogenicity, are safer than iPSC grafts.
Collapse
Affiliation(s)
- Mou Gao
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
- Affiliated Bayi Brain hospital, General Hospital of PLA Army, Beijing 100700, China
| | - Hui Yao
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
- Affiliated Bayi Brain hospital, General Hospital of PLA Army, Beijing 100700, China
| | - Qin Dong
- Department of Neurology, Fu Xing Hospital, Capital Medical University, Beijing 100038, China
| | - Hongtian Zhang
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
- Affiliated Bayi Brain hospital, General Hospital of PLA Army, Beijing 100700, China
| | - Zhijun Yang
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
- Affiliated Bayi Brain hospital, General Hospital of PLA Army, Beijing 100700, China
| | - Yang Yang
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
- Affiliated Bayi Brain hospital, General Hospital of PLA Army, Beijing 100700, China
| | - Jianwei Zhu
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
- Affiliated Bayi Brain hospital, General Hospital of PLA Army, Beijing 100700, China
| | - Minhui Xu
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
- Affiliated Bayi Brain hospital, General Hospital of PLA Army, Beijing 100700, China
| | - Ruxiang Xu
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
- Affiliated Bayi Brain hospital, General Hospital of PLA Army, Beijing 100700, China
| |
Collapse
|
99
|
Roura S, Pujal JM, Gálvez-Montón C, Bayes-Genis A. Quality and exploitation of umbilical cord blood for cell therapy: Are we beyond our capabilities? Dev Dyn 2016; 245:710-7. [PMID: 27043849 DOI: 10.1002/dvdy.24408] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/03/2016] [Accepted: 03/29/2016] [Indexed: 02/06/2023] Open
Abstract
There is increasing interest in identifying novel stem cell sources for application in emerging cell therapies. In this context, umbilical cord blood (UCB) shows great promise in multiple clinical settings. The number of UCB banks has therefore increased worldwide, with the objective of preserving potentially life-saving cells that are usually discarded after birth. After a rather long and costly processing procedure, the resultant UCB-derived cell products are cryopreserved until transplantation to patients. However, in many cases, only a small proportion of administered cells engraft successfully. Thus, can we do any better regarding current UCB-based therapeutic approaches? Here we discuss concerns about the use of UCB that are not critically pondered by researchers, clinicians, and banking services, including wasting samples with small volumes and the need for more reliable quality and functional controls to ensure the biological activity of stem cells and subsequent engraftment and treatment efficacy. Finally, we appeal for collaborative agreements between research institutions and UCB banks in order to redirect currently discarded small-volume UCB units for basic and clinical research purposes. Developmental Dynamics 245:710-717, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Santiago Roura
- Heart Failure and Cardiac Regeneration (ICREC) Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Crta.Can Ruti-Camí Escoles s/n, 08916, Badalona, Spain
- Center of Regenerative Medicine in Barcelona, c/ Dr. Aiguader 88, 08003, Barcelona, Spain
| | - Josep Maria Pujal
- Cell Processing Laboratory, Edifici Giroemprèn, Pic de Peguera 11, Parc Científic i Tecnològic Universitat de Girona, 17003, Girona, Spain
| | - Carolina Gálvez-Montón
- Heart Failure and Cardiac Regeneration (ICREC) Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Crta.Can Ruti-Camí Escoles s/n, 08916, Badalona, Spain
| | - Antoni Bayes-Genis
- Heart Failure and Cardiac Regeneration (ICREC) Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Crta.Can Ruti-Camí Escoles s/n, 08916, Badalona, Spain
- Cardiology Service, Germans Trias i Pujol University Hospital, Crta.Can Ruti-Camí Escoles s/n, 08916, Badalona, Spain
- Department of Medicine, Crta. Can Ruti-Camí Escoles s/n, Universitat Autònoma de Barcelona, 08916, Badalona, Spain
| |
Collapse
|
100
|
Roura S, Gálvez-Montón C, Bayes-Genis A. Fibrin, the preferred scaffold for cell transplantation after myocardial infarction? An old molecule with a new life. J Tissue Eng Regen Med 2016; 11:2304-2313. [PMID: 27061269 DOI: 10.1002/term.2129] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/13/2015] [Accepted: 12/10/2015] [Indexed: 12/12/2022]
Abstract
Fibrin is a topical haemostat, sealant and tissue glue, which consists of concentrated fibrinogen and thrombin. It has broad medical and research uses. Recently, several studies have shown that engineered patches comprising mixtures of biological or synthetic materials and progenitor cells showed therapeutic promise for regenerating damaged tissues. In that context, fibrin maintains cell adherence at the site of injury, where cells are required for tissue repair, and offers a nurturing environment that protects implanted cells without interfering with their expected benefit. Here we review the past, present and future uses of fibrin, with a focus on its use as a scaffold material for cardiac repair. Fibrin patches filled with regenerative cells can be placed over the scarring myocardium; this methodology avoids many of the drawbacks of conventional cell-infusion systems. Advantages of using fibrin also include extraction from the patient's blood, an easy readjustment and implantation procedure, increase in viability and early proliferation of delivered cells, and benefits even with the patch alone. In line with this, we discuss the numerous preclinical studies that have used fibrin-cell patches, the practical issues inherent in their generation, and the necessary process of scaling-up from animal models to patients. In the light of the data presented, fibrin stands out as a valuable biomaterial for delivering cells to damaged tissue and for promoting beneficial effects. However, before the fibrin scaffold can be translated from bench to bedside, many issues must be explored further, including suboptimal survival and limited migration of the implanted cells to underlying ischaemic myocardium. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Santiago Roura
- Heart Failure and Cardiac Regeneration (ICREC) Research Programme, Germans Trias i Pujol Health Science Research Institute, Badalona, Barcelona, Spain.,Center of Regenerative Medicine in Barcelona, Barcelona, Spain
| | - Carolina Gálvez-Montón
- Heart Failure and Cardiac Regeneration (ICREC) Research Programme, Germans Trias i Pujol Health Science Research Institute, Badalona, Barcelona, Spain
| | - Antoni Bayes-Genis
- Heart Failure and Cardiac Regeneration (ICREC) Research Programme, Germans Trias i Pujol Health Science Research Institute, Badalona, Barcelona, Spain.,Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Spain
| |
Collapse
|