Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Nat Cell Biol ; 24(5): 659-671, 2022 05.
Article in English | MEDLINE | ID: mdl-35550611

ABSTRACT

Heart regeneration is an unmet clinical need, hampered by limited renewal of adult cardiomyocytes and fibrotic scarring. Pluripotent stem cell-based strategies are emerging, but unravelling cellular dynamics of host-graft crosstalk remains elusive. Here, by combining lineage tracing and single-cell transcriptomics in injured non-human primate heart biomimics, we uncover the coordinated action modes of human progenitor-mediated muscle repair. Chemoattraction via CXCL12/CXCR4 directs cellular migration to injury sites. Activated fibroblast repulsion targets fibrosis by SLIT2/ROBO1 guidance in organizing cytoskeletal dynamics. Ultimately, differentiation and electromechanical integration lead to functional restoration of damaged heart muscle. In vivo transplantation into acutely and chronically injured porcine hearts illustrated CXCR4-dependent homing, de novo formation of heart muscle, scar-volume reduction and prevention of heart failure progression. Concurrent endothelial differentiation contributed to graft neovascularization. Our study demonstrates that inherent developmental programmes within cardiac progenitors are sequentially activated in disease, enabling the cells to sense and counteract acute and chronic injury.


Subject(s)
Nerve Tissue Proteins , Pluripotent Stem Cells , Animals , Cell Differentiation , Cicatrix/pathology , Cicatrix/prevention & control , Fibrosis , Humans , Myocardium/pathology , Myocytes, Cardiac/pathology , Pluripotent Stem Cells/pathology , Receptors, Immunologic , Swine
2.
Regen Med ; 11(5): 483-92, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27404768

ABSTRACT

This paper summarizes the proceedings of a workshop held at Trinity Hall, Cambridge to discuss comparability and includes additional information and references to related information added subsequently to the workshop. Comparability is the need to demonstrate equivalence of product after a process change; a recent publication states that this 'may be difficult for cell-based medicinal products'. Therefore a well-managed change process is required which needs access to good science and regulatory advice and developers are encouraged to seek help early. The workshop shared current thinking and best practice and allowed the definition of key research questions. The intent of this report is to summarize the key issues and the consensus reached on each of these by the expert delegates.


Subject(s)
Pluripotent Stem Cells/transplantation , Regenerative Medicine , Biotechnology/methods , Biotechnology/trends , Humans , Manufacturing and Industrial Facilities , Regenerative Medicine/legislation & jurisprudence , Regenerative Medicine/methods , Regenerative Medicine/trends , United Kingdom
4.
Philos Trans R Soc Lond B Biol Sci ; 370(1680): 20150017, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26416686

ABSTRACT

Cell therapies offer the promise of treating and altering the course of diseases which cannot be addressed adequately by existing pharmaceuticals. Cell therapies are a diverse group across cell types and therapeutic indications and have been an active area of research for many years but are now strongly emerging through translation and towards successful commercial development and patient access. In this article, we present a description of a classification of cell therapies on the basis of their underlying technologies rather than the more commonly used classification by cell type because the regulatory path and manufacturing solutions are often similar within a technology area due to the nature of the methods used. We analyse the progress of new cell therapies towards clinical translation, examine how they are addressing the clinical, regulatory, manufacturing and reimbursement requirements, describe some of the remaining challenges and provide perspectives on how the field may progress for the future.


Subject(s)
Cell- and Tissue-Based Therapy/classification , Animals , Biotechnology/trends , Cell Line, Transformed , Cell- and Tissue-Based Therapy/economics , Cell- and Tissue-Based Therapy/trends , Cellular Reprogramming Techniques , Genetic Vectors , Humans , RNA Editing , Tissue Engineering , Translational Research, Biomedical/trends
5.
Curr Stem Cell Rep ; 1(2): 110-117, 2015.
Article in English | MEDLINE | ID: mdl-26225325

ABSTRACT

The development of induced pluripotent stem cells offers the possibility of the scalable manufacture of cellular therapies for regenerative medicine. Moreover, donors can be selected on the basis of major transplant antigen systems to match the widest possible number of recipients worldwide, reducing the likely risk of immunological rejection and the degree of immune suppression or tolerance required. If such cell lines are to be broadly available, there will need to be mutual recognition of common standards across different jurisdictions. Extensive international collaboration will be required around issues such as determination of the optimal homozygous human leukocyte antigens (HLA) panel, donor selection, screening and consent, good manufacturing practice (GMP), standards and quality control and regulatory legislation. The challenges in establishing a global GMP induced pluripotent stem cell (iPSC) haplobank are formidable. We argue that now is the time to attempt to reach international agreement around common standards for GMP iPSC manufacture before the field develops in a fragmented manner.

6.
Regen Med ; 10(2): 169-79, 2015.
Article in English | MEDLINE | ID: mdl-25835481

ABSTRACT

Cell Therapy Clinical Trial and Preclinical Research databases have been established by the Cell Therapy Catapult to document current and future cell therapy clinical trials in the UK. We identified 41 ongoing trials in April 2014, an increase of seven trials from April 2013. In addition, we identified 45 late-stage preclinical research projects. The majority of the clinical trials are early phase, primarily led by academic groups. The leading therapeutic areas are cancer, cardiology and neurology. The trends in the UK are also seen globally. As the field matures, more later phase and commercial studies will emerge and the challenges will likely evolve into how to manufacture sufficient cell quantities, manage complex logistics for multi-center trials and control cost.


Subject(s)
Cell Transplantation/methods , Cell- and Tissue-Based Therapy/trends , Clinical Trials as Topic , Financing, Government , Geography , Humans , Immunotherapy , Multicenter Studies as Topic , Neoplasms/immunology , Neoplasms/therapy , Research Design , United Kingdom
7.
Sci Rep ; 4: 4670, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24751527

ABSTRACT

To overcome the limitations and misjudgments of conventional prediction of arrhythmic cardiotoxicity, we have developed an on-chip in vitro predictive cardiotoxicity assay using cardiomyocytes derived from human stem cells employing a constructive spatiotemporal two step measurement of fluctuation (short-term variability; STV) of cell's repolarization and cell-to-cell conduction time, representing two origins of lethal arrhythmia. Temporal STV of field potential duration (FPD) showed a potential to predict the risks of lethal arrhythmia originated from repolarization dispersion for false negative compounds, which was not correctly predicted by conventional measurements using animal cells, even for non-QT prolonging clinical positive compounds. Spatial STV of conduction time delay also unveiled the proarrhythmic risk of asynchronous propagation in cell networks, whose risk cannot be correctly predicted by single-cell-based measurements, indicating the importance of the spatiotemporal fluctuation viewpoint of in vitro cell networks for precise prediction of lethal arrhythmia reaching clinical assessment such as thorough QT assay.


Subject(s)
Cardiotoxicity , Drug Evaluation, Preclinical , Microchip Analytical Procedures , Myocytes, Cardiac/drug effects , Cell Communication/drug effects , Cell Culture Techniques , Humans , In Vitro Techniques , Lab-On-A-Chip Devices , Myocytes, Cardiac/metabolism
8.
Stem Cells Transl Med ; 3(4): 433-47, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24604283

ABSTRACT

Human induced pluripotent stem cells (iPSCs) are potential cell sources for regenerative medicine; however, clinical applications of iPSCs are restricted because of undesired genomic modifications associated with most reprogramming protocols. We show, for the first time, that chondrocytes from autologous chondrocyte implantation (ACI) donors can be efficiently reprogrammed into iPSCs using a nonintegrating method based on mRNA delivery, resulting in footprint-free iPSCs (no genome-sequence modifications), devoid of viral factors or remaining reprogramming molecules. The search for universal allogeneic cell sources for the ACI regenerative treatment has been difficult because making chondrocytes with high matrix-forming capacity from pluripotent human embryonic stem cells has proven challenging and human mesenchymal stem cells have a predisposition to form hypertrophic cartilage and bone. We show that chondrocyte-derived iPSCs can be redifferentiated in vitro into cartilage matrix-producing cells better than fibroblast-derived iPSCs and on par with the donor chondrocytes, suggesting the existence of a differentiation bias toward the somatic cell origin and making chondrocyte-derived iPSCs a promising candidate universal cell source for ACI. Whole-genome single nucleotide polymorphism array and karyotyping were used to verify the genomic integrity and stability of the established iPSC lines. Our results suggest that RNA-based technology eliminates the risk of genomic integrations or aberrations, an important step toward a clinical-grade cell source for regenerative medicine such as treatment of cartilage defects and osteoarthritis.


Subject(s)
Cartilage/metabolism , Cell Dedifferentiation , Chondrocytes/metabolism , Induced Pluripotent Stem Cells/metabolism , Cartilage/cytology , Cells, Cultured , Chondrocytes/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism
9.
Methods Mol Biol ; 997: 239-51, 2013.
Article in English | MEDLINE | ID: mdl-23546761

ABSTRACT

Human pluripotent stem cells provide unique possibilities for in vitro studies of human cells in basic research, disease modeling as well as in industrial applications. By introducing relevant genome engineering technology, and thereby creating, for example, reporter cell lines, one will facilitate and improve safety pharmacology, toxicity testing, and can help the scientists to better understand pathological processes in humans. This review discusses how the merger of these two fields, human pluripotent stem cells and genome engineering, form extremely powerful tools and how they have been implemented already within the scientific community. In sharp contrast to immortalized human cell lines, which are both easy to expand and very simple to transfect, the genetically modified pluripotent stem cell line can be directed to a specific cell lineage and provide the user with highly relevant information. We highlight some of the challenges the field had to solve and how new technology advancements has removed the early bottlenecks.


Subject(s)
Drug Evaluation, Preclinical/methods , Pluripotent Stem Cells/metabolism , Animals , Cells, Cultured , Drug Discovery , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Genetic Diseases, Inborn/drug therapy , Humans , Neurodegenerative Diseases/drug therapy , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/pathology , Staining and Labeling
10.
PLoS One ; 8(1): e55319, 2013.
Article in English | MEDLINE | ID: mdl-23383153

ABSTRACT

Mesenchymal stroma cells (MSCs) have a high potential for novel cell therapy approaches in clinical transplantation. Commonly used bone marrow-derived MSCs (BM-MSCs), however, have a restricted proliferative capacity and cultures are difficult to standardize. Recently developed human embryonic stem cell-derived mesenchymal stroma cells (hES-MSCs) might represent an alternative and unlimited source of hMSCs. We therefore compared human ES-cell-derived MSCs (hES-MP002.5 cells) to normal human bone marrow-derived MSCs (BM-MSCs). hES-MP002.5 cells had lower yet reasonable CFU-F capacity compared with BM-MSC (8±3 versus 29±13 CFU-F per 100 cells). Both cell types showed similar immunophenotypic properties, i.e. cells were positive for CD105, CD73, CD166, HLA-ABC, CD44, CD146, CD90, and negative for CD45, CD34, CD14, CD31, CD117, CD19, CD 271, SSEA-4 and HLA-DR. hES-MP002.5 cells, like BM-MSCs, could be differentiated into adipocytes, osteoblasts and chondrocytes in vitro. Neither hES-MP002.5 cells nor BM-MSCs homed to the bone marrow of immune-deficient NSG mice following intravenous transplantation, whereas intra-femoral transplantation into NSG mice resulted in engraftment for both cell types. In vitro long-term culture-initiating cell assays and in vivo co-transplantation experiments with cord blood CD34+ hematopoietic cells demonstrated furthermore that hES-MP002.5 cells, like BM-MSCs, possess potent stroma support function. In contrast to BM-MSCs, however, hES-MP002.5 cells showed no or only little activity in mixed lymphocyte cultures and phytohemagglutinin (PHA) lymphocyte stimulation assays. In summary, ES-cell derived MSCs might be an attractive unlimited source for stroma transplantation approaches without suppressing immune function.


Subject(s)
Embryonic Stem Cells/cytology , Hematopoiesis/physiology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Culture Techniques , Colony-Forming Units Assay , Cytokines/metabolism , Fluorescent Antibody Technique , Humans , Immunophenotyping , Mesenchymal Stem Cell Transplantation/trends , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Mice
11.
Acta Ophthalmol ; 91(2): 127-30, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22280565

ABSTRACT

PURPOSE: The aim of this study was to investigate whether cells originating from human embryonic stem cells (hESCs) could be successfully transplanted onto a partially wounded human cornea. A second aim was to study the ability of the transplanted cells to differentiate into corneal epithelial-like cells. METHODS: Spontaneously, differentiated hESCs were transplanted onto a human corneal button (without limbus) with the epithelial layer partially removed. The cells were cultured on Bowman's membrane for up to 9 days, and the culture dynamics documented in a time-lapse system. As the transplanted cells originated from a genetically engineered hESC line, they all expressed green fluorescent protein, which facilitated their identification during the culture experiments, tissue preparation and analysis. To detect any differentiation into human corneal epithelial-like cells, we analysed the transplanted cells by immunohistochemistry using antibodies specific for CK3, CK15 and PAX6. RESULTS: The transplanted cells established and expanded on Bowman's membrane, forming a 1-4 cell layer surrounded by host corneal epithelial cells. Expression of the corneal marker PAX6 appeared 3 days after transplantation, and after 6 days, the cells were expressing both PAX6 and CK3. CONCLUSION: This shows that it is possible to transplant cells originating from hESCs onto Bowman's membrane with the epithelial layer partially removed and to get these cells to establish, grow and differentiate into corneal epithelial-like cells in vitro.


Subject(s)
Corneal Injuries , Embryonic Stem Cells/transplantation , Eye Injuries/therapy , Stem Cell Transplantation , Wounds, Nonpenetrating/therapy , Cell Differentiation , Cell Proliferation , Cells, Cultured , Embryonic Stem Cells/cytology , Epithelium, Corneal/cytology , Epithelium, Corneal/metabolism , Eye Injuries/metabolism , Eye Proteins/metabolism , Fluorescent Antibody Technique, Indirect , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Humans , Keratin-15/metabolism , Keratin-3/metabolism , Limbus Corneae/cytology , Limbus Corneae/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Tissue Embedding , Tissue Fixation , Wounds, Nonpenetrating/metabolism
12.
Tissue Eng Part A ; 19(1-2): 175-87, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22924642

ABSTRACT

Bone tissue engineering represents a promising strategy to obviate bone deficiencies, allowing the ex vivo construction of bone substitutes with unprecedented potential in the clinical practice. Considering that in the human body cells are constantly stimulated by chemical and mechanical stimuli, the use of bioreactor is emerging as an essential factor for providing the proper environment for the reproducible and large-scale production of the engineered substitutes. Human mesenchymal stem cells (hMSCs) are experimentally relevant cells but, regardless the encouraging results reported after culture under dynamic conditions in bioreactors, show important limitations for tissue engineering applications, especially considering their limited proliferative potential, loss of functionality following protracted expansion, and decline in cellular fitness associated with aging. On the other hand, we previously demonstrated that human embryonic stem cell-derived mesodermal progenitors (hES-MPs) hold great potential to provide a homogenous and unlimited source of cells for bone engineering applications. Based on prior scientific evidence using different types of stem cells, in the present study we hypothesized that dynamic culture of hES-MPs in a packed bed/column bioreactor had the potential to affect proliferation, expression of genes involved in osteogenic differentiation, and matrix mineralization, therefore resulting in increased bone-like tissue formation. The reported findings suggest that hES-MPs constitute a suitable alternative cell source to hMSCs and hold great potential for the construction of bone substitutes for tissue engineering applications in clinical settings.


Subject(s)
Batch Cell Culture Techniques/instrumentation , Bioreactors , Bone Development/physiology , Embryonic Stem Cells/cytology , Mesoderm/cytology , Organ Culture Techniques/instrumentation , Osteogenesis/physiology , Cell Differentiation , Cells, Cultured , Equipment Design , Equipment Failure Analysis , Humans , Mechanotransduction, Cellular/physiology , Mesenchymal Stem Cells
13.
Methods Mol Biol ; 873: 53-68, 2012.
Article in English | MEDLINE | ID: mdl-22528348

ABSTRACT

This chapter describes the principles for derivation and maintenance of human embryonic stem cells. Detailed protocols are outlined and researchers who are generally skilled in mammalian cell culture should be able to repeat the processes successfully. Further, the protocols are intended for scientists who do not have access to advanced IVF equipment and therefore cannot perform, e.g. assisted hatching. In addition to derivation, we also discuss characterisation and banking of hES cells.


Subject(s)
Cell Culture Techniques/methods , Embryonic Stem Cells/cytology , Blastocyst/cytology , Humans
14.
Nat Biotechnol ; 29(12): 1132-44, 2011 Nov 27.
Article in English | MEDLINE | ID: mdl-22119741

ABSTRACT

The International Stem Cell Initiative analyzed 125 human embryonic stem (ES) cell lines and 11 induced pluripotent stem (iPS) cell lines, from 38 laboratories worldwide, for genetic changes occurring during culture. Most lines were analyzed at an early and late passage. Single-nucleotide polymorphism (SNP) analysis revealed that they included representatives of most major ethnic groups. Most lines remained karyotypically normal, but there was a progressive tendency to acquire changes on prolonged culture, commonly affecting chromosomes 1, 12, 17 and 20. DNA methylation patterns changed haphazardly with no link to time in culture. Structural variants, determined from the SNP arrays, also appeared sporadically. No common variants related to culture were observed on chromosomes 1, 12 and 17, but a minimal amplicon in chromosome 20q11.21, including three genes expressed in human ES cells, ID1, BCL2L1 and HM13, occurred in >20% of the lines. Of these genes, BCL2L1 is a strong candidate for driving culture adaptation of ES cells.


Subject(s)
Embryonic Stem Cells/cytology , Growth/genetics , Induced Pluripotent Stem Cells/cytology , RNA-Binding Proteins/metabolism , bcl-X Protein/metabolism , Cell Differentiation/genetics , Cell Line , Chromosomes, Human, Pair 20/genetics , Clonal Evolution/genetics , DNA Methylation , Ethnicity/genetics , Gene Expression Regulation, Developmental , Genetic Variation , Genotype , Humans , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Polymorphism, Single Nucleotide , RNA-Binding Proteins/genetics , Selection, Genetic/genetics , bcl-X Protein/genetics
15.
Cytotechnology ; 63(3): 227-37, 2011 May.
Article in English | MEDLINE | ID: mdl-21409453

ABSTRACT

Pluripotent human embryonic stem cell (hESC) lines are a promising model system in developmental and tissue regeneration research. Differentiation of hESCs towards the three germ layers and finally tissue specific cell types is often performed through the formation of embryoid bodies (EBs) in suspension or hanging droplet culture systems. However, these systems are inefficient regarding embryoid body (EB) formation, structural support to the EB and long term differentiation capacity. The present study investigates if agarose, as a semi solid matrix, can facilitate EB formation and support differentiation of hESC lines. The results showed that agarose culture is able to enhance EB formation efficiency with 10% and increase EB growth by 300%. The agarose culture system was able to maintain expression of the three germ layers over 8 weeks of culture. All of the four hESC lines tested developed EBs in the agarose system although with a histological heterogeneity between cell lines as well as within cell lines. In conclusion, a 3-D agarose culture of spherical hESC colonies improves EB formation and growth in a cost effective, stable and non-laborious technique.

16.
J Tissue Eng Regen Med ; 5(3): 180-90, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20718035

ABSTRACT

For more than a decade, human mesenchymal stem cells (hMSCs) have been used in bone tissue-engineering research. More recently some of the focus in this field has shifted towards the use of embryonic stem cells. While it is well known that hMSCs are able to form bone when implanted subcutaneously in immune-deficient mice, the osteogenic potential of embryonic stem cells has been mainly assessed in vitro. Therefore, we performed a series of studies to compare the in vitro and in vivo osteogenic capacities of human and mouse embryonic stem cells to those of hMSCs. Embryonic and mesenchymal stem cells showed all characteristic signs of osteogenic differentiation in vitro when cultured in osteogenic medium, including the deposition of a mineralized matrix and expression of genes involved in osteogenic differentiation. As such, based on the in vitro results, osteogenic ES cells could not be discriminated from osteogenic hMSCs. Nevertheless, although osteogenic hMSCs formed bone upon implantation, osteogenic cells derived from both human and mouse embryonic stem cells did not form functional bone, indicated by absence of osteocytes, bone marrow and lamellar bone. Although embryonic stem cells show all signs of osteogenic differentiation in vitro, it appears that, in contrast to mesenchymal stem cells, they do not possess the ability to form bone in vivo when a similar culture method and osteogenic differentiation protocol was applied.


Subject(s)
Bone Marrow Cells/cytology , Embryonic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Osteocytes/cytology , Osteogenesis/physiology , Alkaline Phosphatase/metabolism , Animals , Calcification, Physiologic/drug effects , Calcium Phosphates/pharmacology , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/ultrastructure , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/enzymology , Mice , Osteocytes/drug effects , Osteogenesis/drug effects , Spectrum Analysis, Raman
17.
Tissue Eng Part A ; 16(11): 3427-40, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20653416

ABSTRACT

Human mesenchymal stem cells (hMSCs) represent a promising source of cells for bone tissue engineering. However, their low frequencies and limited proliferation restrict their clinical utility. An alternative is the use of human embryonic stem cells (hESCs), but labor-intensive expansion with the need for coating support limits their clinical use. We have previously derived a cell line from hESCs denoted matrix-free growth (MFG)-hESC that are independent of coating support for expansion, and we here compare its osteogenic capacity to that of hMSCs. Microarray analysis of hMSCs and MFG-hESCs revealed differential expression of genes involved in ossification. MFG-hESCs have significantly higher expression of secreted phosphoprotein 1 (SPP1) during osteogenic differentiation, whereas the opposite was true for alkaline phosphatase (ALPL), transforming growth factor, beta 1 (TGFB2), runt-related transcription factor 2 (RUNX2), and forkhead box C1 (FOXC1), as well as the activity of the ALPL enzyme, demonstrating that these two cell types differentiate into the osteogenic lineage using different signaling pathways. von Kossa staining, time-of-flight secondary ion mass spectrometry, and measurement of calcium and phosphate in the extracellular matrix demonstrated a superior ability of the MFG-hESCs to produce a mineralized matrix compared to hMSCs. The superior ability of the MFG-hESCs to form mineralized matrix compared to hMSCs demonstrates that MFG-hESCs are a promising alternative to the use of adult stem cells in future bone regenerative applications.


Subject(s)
Embryonic Stem Cells/cytology , Extracellular Matrix/metabolism , Mesenchymal Stem Cells/cytology , Osteogenesis/physiology , Adolescent , Adult , Alkaline Phosphatase/metabolism , Biomarkers/metabolism , Calcification, Physiologic/genetics , Cell Line , Cell Proliferation , Cluster Analysis , Embryonic Stem Cells/enzymology , Flow Cytometry , Gene Expression Regulation , Humans , Mesenchymal Stem Cells/enzymology , Oligonucleotide Array Sequence Analysis , Osteogenesis/genetics , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Secondary Ion
18.
J Biotechnol ; 148(4): 208-15, 2010 Aug 02.
Article in English | MEDLINE | ID: mdl-20600380

ABSTRACT

The successful transfer of human embryonic stem cell (hESC) technology and cellular products into clinical and industrial applications needs to address issues of automation, standardization and the generation of relevant cell numbers of high quality. In this study, we combined microcarrier technology and controlled stirred tank bioreactors, to develop an efficient and scalable system for expansion of pluripotent hESCs. We demonstrate the importance of controlling pO(2) at 30% air saturation to improve hESCs growth. This concentration allowed for a higher energetic cell metabolism, increased growth rate and maximum cell concentration in contrast to 5% pO(2) where a shift to anaerobic metabolism was observed, decreasing cell expansion 3-fold. Importantly, the incorporation of an automated perfusion system in the bioreactor enhanced culture performance and allowed the continuous addition of small molecules assuring higher cell concentrations for a longer time period. The expanded hESCs retained their undifferentiated phenotype and pluripotency. Our results show, for the first time, that the use of controlled bioreactors is critical to ensure the production of high quality hESCs. When compared to the standard colony culture, our strategy improves the final yield of hESCs by 12-fold, providing a potential bioprocess to be transferred to clinical and industrial applications.


Subject(s)
Bioreactors , Models, Biological , Oxygen/metabolism , Perfusion/instrumentation , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Cell Proliferation , Cell Survival , Cells, Cultured , Computer Simulation , Equipment Design , Humans , Perfusion/methods
19.
Tissue Eng Part A ; 16(11): 3413-26, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20536357

ABSTRACT

INTRODUCTION: Human mesenchymal stem cells (hMSCs) are promising candidates for bone engineering and regeneration with a considerable number of experimental successes reported over the last years. However, hMSCs show several limitations for tissue engineering applications, which can be overcome by using human embryonic stem cell-derived mesodermal progenitors (hES-MPs). The aim of this study was to investigate and compare the osteogenic differentiation potential of hMSCs and hES-MPs. MATERIALS AND METHODS: The osteogenic differentiation and mineralization behavior of both cell types were evaluated at passage 5, 10, 15, and 20. Expression of COL1A1, RUNX2, OPN, and OC was evaluated by reverse transcription (RT)-polymerase chain reaction, whereas mineralization was examined by photospectrometry, von Kossa staining, and time-of-flight secondary ion mass spectrometry. The immunoprofile of both cell types was investigated by flow cytometry. RESULTS: We demonstrated that, under proper stimulation, hES-MPs undergo osteogenic differentiation and exhibit significantly increased mineralization ability compared to hMSCs after protracted expansion. hES-MPs were also found to express lower amount of human leukocyte antigens class II proteins. CONCLUSIONS: The high osteogenic ability of hES-MPs, together with low expression of human leukocyte antigens class II, makes these cells an attractive alternative for bulk production of cells for bone engineering applications.


Subject(s)
Embryonic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Mesoderm/cytology , Osteogenesis , Tissue Engineering , Adipogenesis/genetics , Adolescent , Adult , Alkaline Phosphatase/metabolism , Calcification, Physiologic/physiology , Calcium/metabolism , Cell Separation , Embryonic Stem Cells/enzymology , Flow Cytometry , Gene Expression Regulation , HLA Antigens/metabolism , Humans , Mesenchymal Stem Cells/enzymology , Mesoderm/enzymology , Osteogenesis/genetics , Phosphates/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Secondary Ion , Staining and Labeling , Tissue Donors
20.
Tissue Eng Part A ; 16(7): 2161-82, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20136402

ABSTRACT

Adult stem cells, such as human mesenchymal stem cells (hMSCs), show limited proliferative capacity and, after long-term culture, lose their differentiation capacity and are therefore not an optimal cell source for tissue engineering. Human embryonic stem cells (hESCs) constitute an important new resource in this field, but one major drawback is the risk of tumor formation in the recipients. One alternative is to use progenitor cells derived from hESCs that are more lineage restricted but do not form teratomas. We have recently derived a cell line from hESCs denoted hESC-derived mesodermal progenitors (hES-MPs), and here, using genome-wide microarray analysis, we report that the process of hES-MPs derivation results in a significantly altered expression of hESC characteristic genes to an expression level highly similar to that of hMSCs. However, hES-MPs displayed a significantly higher proliferative capacity and longer telomeres. The hES-MPs also displayed lower expression of HLA class II proteins before and after interferon-gamma treatment, indicating that these cells may somewhat be immunoprivileged and potentially used for HLA-incompatible transplantation. The hES-MPs are thus an appealing alternative to hMSCs in tissue engineering applications and stem-cell-based therapies for mesodermal tissues.


Subject(s)
Cell Shape , Embryonic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Mesoderm/cytology , Tissue Engineering/methods , Adolescent , Cell Line , Cell Proliferation , Flow Cytometry , Gene Expression Regulation , Gene Regulatory Networks , Histocompatibility Antigens/immunology , Humans , Immunohistochemistry , Oligonucleotide Array Sequence Analysis , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Telomerase/metabolism , Telomere/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...