Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Int J Mol Sci ; 22(18)2021 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-34575977

RESUMEN

Amidst the global shortfalls in blood supply, storage limitations of donor blood and the availability of potential blood substitutes for transfusion applications, society has pivoted towards in vitro generation of red blood cells (RBCs) as a means to solve these issues. Many conventional research studies over the past few decades have found success in differentiating hematopoietic stem and progenitor cells (HSPCs) from cord blood, adult bone marrow and peripheral blood sources. More recently, techniques that involve immortalization of erythroblast sources have also gained traction in tackling this problem. However, the RBCs generated from human induced pluripotent stem cells (hiPSCs) still remain as the most favorable solution due to many of its added advantages. In this review, we focus on the breakthroughs for high-density cultures of hiPSC-derived RBCs, and highlight the major challenges and prospective solutions throughout the whole process of erythropoiesis for hiPSC-derived RBCs. Furthermore, we elaborate on the recent advances and techniques used to achieve cost-effective, high-density cultures of GMP-compliant RBCs, and on their relevant novel applications after downstream processing and purification.


Asunto(s)
Sustitutos Sanguíneos/uso terapéutico , Eritrocitos/citología , Células Madre Hematopoyéticas/citología , Células Madre Pluripotentes Inducidas/citología , Diferenciación Celular/genética , Transfusión de Eritrocitos , Eritropoyesis/genética , Sangre Fetal/citología , Humanos
2.
Cytotherapy ; 21(6): 631-642, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30975604

RESUMEN

In the current emerging trend of using human mesenchymal stromal cell (MSCs) for cell therapy, large quantities of cells are needed for clinical testing. Current methods of culturing cells, using tissue culture flasks or cell multilayer vessels, are proving to be ineffective in terms of cost, space and manpower. Therefore, alternatives such as large-scale industrialized production of MSCs in stirred tank bioreactors using microcarriers (MCs) are needed. Moreover, the development of biodegradable MCs for MSC expansion can streamline the bioprocess by eliminating the need for enzymatic cell harvesting and scaffold seeding for bone-healing therapies. Our previous studies described a process of making regulated density (1.06 g/cm3) porous polycaprolactone biodegradable MCs Light Polycarprolactone (LPCL) (MCs), which were used for expanding MSCs from various sources in stirred suspension culture. Here, we use human early MSCs (heMSCs) expanded on LPCL MCs for evaluation of their osteogenic differentiation potential in vitro as well as their use in vivo calvarial defect treatment in a rat model. In summary, (i) in vitro data show that LPCL MCs can be used to efficiently expand heMSCs in stirred cultures while maintaining surface marker expression; (ii) LPCL MCs can be used as scaffolds for cell transfer for transplantation in vivo; (iii) 50% sub-confluency, mid-logarithmic phase, on LPCL MCs (50% confluent) exhibited higher secretion levels of six cytokines (interleukin [IL]-6, IL-8, Vascular endothelial growth factor (VEGF), Monocyte Chemoattractant Protein-1 (MCP-1), growth-regulated oncogene-α (GRO-α) and stromal cell-derived factor-1α (SDF-1α)) as compared with 100% confluent, stationary phase cultures (100% confluent); (iv) these 50% confluent cultures demonstrated better in vitro osteogenic differentiation capacity as compared with 100% confluent cultures (higher levels of calcium deposition and at earlier stage); the improved bone differentiation capacity of these 50% confluent cultures was also demonstrated at the molecular level by higher expression of early osteoblast genes Runt-related transcription factor 2 (RUNX2), Alkaline phosphatase (ALP), collagen type I, osterix and osteocalcin); and (v) in vivo implantation of biodegradable LPCL MCs covered with 50% heMSCs into rats with calvarial defect demonstrated significantly better bone formation as compared with heMSCs obtained from monolayer cultures (5.1 ± 1.6 mm3 versus 1.3 ± 0.7 mm3). Moreover, the LPCL MCs covered with 50% heMSCs supported better in vivo bone formation compared with 100% confluent culture (2.1 ± 1.3 mm3). Taken together, our study highlights the potential of implanting 50% confluent MSCs propagated on LPCL MCs as optimal for bone regeneration. This methodology allows for the production of large numbers of MSCs in a three-dimensional (3D) stirred reactor, while supporting improved bone healing and eliminating the need for a 3D matrix support scaffold, as traditionally used in bone-healing treatments.


Asunto(s)
Materiales Biocompatibles/química , Regeneración Ósea/fisiología , Técnicas de Cultivo de Célula/métodos , Células Madre Mesenquimatosas/citología , Osteogénesis/fisiología , Animales , Reactores Biológicos , Recuento de Células , Técnicas de Cultivo de Célula/instrumentación , Diferenciación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Citocinas/metabolismo , Humanos , Masculino , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/fisiología , Poliésteres/química , Ratas Desnudas , Cráneo
3.
Cytotherapy ; 19(3): 419-432, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28017598

RESUMEN

Large numbers of human mesenchymal stromal cells (MSCs) used for a variety of applications in tissue engineering and cell therapy can be generated by scalable expansion in a bioreactor using microcarriers (MCs) systems. However, the enzymatic digestion process needed to detach cells from the growth surface can affect cell viability and potentially the potency and differentiation efficiency. Thus, the main aim of our study was to develop biocompatible and biodegradable MCs that can support high MSC yields while maintaining their differentiation capability and potency. After cell expansion, the cells that covered MCs can be directly implanted in vivo without the need for cell harvesting or use of scaffold. Poly-ε-caprolactone (PCL) is known as a biocompatible and biodegradable material. However, it cannot be used for generation of MCs because its high density (1.14 g/cm3) would exclude its applicability for suspension MCs in stirred reactors. In this article, we describe expansion and potency of MSCs propagated on low-density (1.06 g/cm3) porous PCL MCs coated with extracellular matrices (LPCLs) in suspended stirred reactors. Using these LPCLs, cell yields of about 4 × 104 cells/cm2 and 7- to 10-fold increases were obtained using four different MSC lines (bone marrow, cord blood, fetal and Wharton's jelly). These yields were comparable with those obtained using non-degradable MCs (Cytodex 3) and higher than two-dimensional monolayer (MNL) cultures. A fed-batch process, which demonstrated faster cell expansion (4.5 × 104 cells/cm2 in 5 days as compared with 7 days in batch culture) and about 70% reduction in growth media usage, was developed and scaled up from 100-mL spinner flask to 1-L controlled bioreactor. Surface marker expression, trilineage differentiation and clonogenic potential of the MSCs expanded on LPCL were not affected. Cytokine secretion kinetics, which occurred mostly during late logarithmic phase, was usually comparable with that obtained in Cytodex 3 cultures and higher than MNL cultures. In conclusion, biodegradable LPCL can be used to efficiently expand a variety of MSC lines in stirred scalable reactors in a cost-effective manner while maintaining surface markers expression, differentiation capability and high levels of cytokine secretion. This study is the first step in testing these cell-biodegradable porous MC aggregates for tissue engineering and cell therapy, such as bone and cartilage regeneration, or wound healing.


Asunto(s)
Implantes Absorbibles , Técnicas de Cultivo Celular por Lotes/métodos , Proliferación Celular , Citocinas/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Poliésteres/química , Andamios del Tejido/química , Reactores Biológicos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Medios de Cultivo/metabolismo , Dextranos/química , Humanos , Ensayo de Materiales , Microtecnología/instrumentación , Ingeniería de Tejidos/métodos
4.
Langmuir ; 33(12): 3068-3079, 2017 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-28221044

RESUMEN

Polymeric microspheres may serve as microcarrier (MC) matrices, for the expansion of anchorage-dependent stem cells. They require surface properties that promote both initial cell adhesion and the subsequent spreading of cells, which is a prerequisite for successful expansion. When implemented in a three-dimensional culture environment, under agitation, their suspension under low shear rates depends on the MCs having a modest negative buoyancy, with a density of 1.02-1.05 g/cm3. Bioresorbable poly-ε-caprolactone (PCL), with a density of 1.14 g/cm3, requires a reduction in volumetric density, for the microspheres to achieve high cell viability and yields. Uniform-sized droplets, from solutions of PCL dissolved in dichloromethane (DCM), were generated by coaxial microfluidic geometry. Subsequent exposure to ethanol rapidly extracted the DCM solvent, solidifying the droplets and yielding monodisperse microspheres with a porous structure, which was demonstrated to have tunable porosity and a hollow inner core. The variation in process parameters, including the molecular weight of PCL, its concentration in DCM, and the ethanol concentration, served to effectively alter the diffusion flux between ethanol and DCM, resulting in a broad spectrum of volumetric densities of 1.04-1.11 g/cm3. The solidified microspheres are generally covered by a smooth thin skin, which provides a uniform cell culture surface and masks their internal porous structure. When coated with a cationic polyelectrolyte and extracellular matrix protein, monodisperse microspheres with a diameter of approximately 150 µm and densities ranging from 1.05-1.11 g/cm3 are capable of supporting the expansion of human mesenchymal stem cells (hMSCs). Validation of hMSC expansion was carried out with a positive control of commercial Cytodex 3 MCs and a negative control of uncoated low-density PCL MCs. Static culture conditions generated more than 70% cell attachment and similar yields of sixfold cell expansion on all coated MCs, with poor cell attachment and growth on the negative control. Under agitation, coated porous microspheres, with a low density of 1.05 g/cm3, achieved robust cell attachment and resulted in high cell yields of ninefold cell expansion, comparable with those generated by commercial Cytodex 3 MCs.


Asunto(s)
Células Madre Mesenquimatosas/citología , Poliésteres/química , Supervivencia Celular , Humanos , Cloruro de Metileno/química , Microesferas , Estructura Molecular , Tamaño de la Partícula , Porosidad , Propiedades de Superficie
5.
Biochem Biophys Res Commun ; 473(3): 769-73, 2016 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-26385177

RESUMEN

Mesenchymal stromal cells (MSCs) are being investigated for a variety of therapeutic indications. However, current 2D planar technology cannot meet the anticipated demand and a shift to serum-free microcarrier cultures is needed in order to meet the quality and quantity of cells required. Here we summarize several recent attempts to grow cells in such conditions, and identify several variables that affect cell expansion, including tissue source, serum-free medium formulation, microcarrier type and matrix, and agitation regime (continuous versus intermittent). Optimization of these culture conditions will be necessary to ensure success in bioreactor-scale production of MSCs for cell therapies.


Asunto(s)
Técnicas de Cultivo de Célula , Células Madre Mesenquimatosas/citología , Animales , Reactores Biológicos , Proliferación Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Medios de Cultivo/metabolismo , Matriz Extracelular/metabolismo , Humanos , Fenotipo , Resistencia al Corte
6.
Biochem Biophys Res Commun ; 473(3): 764-8, 2016 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-26385176

RESUMEN

Current methods for human pluripotent stem cells (hPSC) expansion and differentiation can be limited in scalability and costly (due to their labor intensive nature). This can limit their use in cell therapy, drug screening and toxicity assays. One of the approaches that can overcome these limitations is microcarrier (MC) based cultures in which cells are expanded as cell/MC aggregates and then directly differentiated as embryoid bodies (EBs) in the same agitated reactor. This integrated process can be scaled up and eliminate the need for some culture manipulation used in common monolayer and EBs cultures. This review describes the principles of such microcarriers based integrated hPSC expansion and differentiation process, and parameters that can affect its efficiency (such as MC type and extracellular matrix proteins coatings, cell/MC aggregates size, and agitation). Finally examples of integrated process for generation cardiomyocytes (CM) and neural progenitor cells (NPC) as well as challenges to be solved are described.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Neuronas/metabolismo , Células Madre Pluripotentes/citología , Técnicas de Cultivo de Célula , Proliferación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Humanos , Miocitos Cardíacos/citología , Células Madre/citología
7.
Cytotherapy ; 18(6): 740-53, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27173750

RESUMEN

BACKGROUND AIMS: Cartilage tissue engineering with human mesenchymal stromal cells (hMSC) is promising for allogeneic cell therapy. To achieve large-scale hMSC propagation, scalable microcarrier-based cultures are preferred over conventional static cultures on tissue culture plastic. Yet it remains unclear how microcarrier cultures affect hMSC chondrogenic potential, and how this potential is distinguished from that of tissue culture plastic. Hence, our study aims to compare the chondrogenic potential of human early MSC (heMSC) between microcarrier-spinner and tissue culture plastic cultures. METHODS: heMSC expanded on either collagen-coated Cytodex 3 microcarriers in spinner cultures or tissue culture plastic were harvested for chondrogenic pellet differentiation with empirically determined chondrogenic inducer bone morphogenetic protein 2 (BMP2). Pellet diameter, DNA content, glycosaminoglycan (GAG) and collagen II production, histological staining and gene expression of chondrogenic markers including SOX9, S100ß, MMP13 and ALPL, were investigated and compared in both conditions. RESULTS: BMP2 was the most effective chondrogenic inducer for heMSC. Chondrogenic pellets generated from microcarrier cultures developed larger pellet diameters, and produced more DNA, GAG and collagen II per pellet with greater GAG/DNA and collagen II/DNA ratios compared with that of tissue culture plastic. Moreover, they induced higher expression of chondrogenic genes (e.g., S100ß) but not of hypertrophic genes (e.g., MMP13 and ALPL). A similar trend showing enhanced chondrogenic potential was achieved with another microcarrier type, suggesting that the mechanism is due to the agitated nature of microcarrier cultures. CONCLUSIONS: This is the first study demonstrating that scalable microcarrier-spinner cultures enhance the chondrogenic potential of heMSC, supporting their use for large-scale cell expansion in cartilage cell therapy.


Asunto(s)
Cartílago/metabolismo , Técnicas de Cultivo de Célula , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Condrogénesis/fisiología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Ingeniería de Tejidos/métodos , Fosfatasa Alcalina/biosíntesis , Proteína Morfogenética Ósea 2/farmacología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Condrocitos/citología , Colágeno/metabolismo , ADN/análisis , ADN/metabolismo , Glicosaminoglicanos/metabolismo , Humanos , Metaloproteinasa 13 de la Matriz/biosíntesis , Subunidad beta de la Proteína de Unión al Calcio S100/biosíntesis , Factor de Transcripción SOX9/biosíntesis , Trasplante Homólogo
8.
Biomed Microdevices ; 17(6): 105, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26458560

RESUMEN

The generation of liquefied poly-ɛ-caprolactone (PCL) droplets by means of a microfluidic device results in uniform-sized microspheres, which are validated as microcarriers for human embryonic stem cell culture. Formed droplet size and size distribution, as well as the resulting PCL microsphere size, are correlated with the viscosity and flow rate ratio of the dispersed (Q d) and continuous (Q c) phases. PCL in dichloromethane increases its viscosity with concentration and molecular weight. Higher viscosity and Q d/Q c lead to the formation of larger droplets, within two observed formation modes: dripping and jetting. At low viscosity of dispersed phase and Q d/Q c, the microfluidic device is operated in dripping mode, which generates droplets and microspheres with greater size uniformity. Solutions with lower molecular weight PCL have lower viscosity, resulting in a wider concentration range for the dripping mode. When coated with extracellular matrix (ECM) proteins, the fabricated PCL microspheres are demonstrated capable of supporting the expansion of human embryonic stem cells.


Asunto(s)
Células Madre Embrionarias Humanas/citología , Microesferas , Poliésteres/química , Adhesión Celular , Proliferación Celular , Células Cultivadas , Materiales Biocompatibles Revestidos/química , Proteínas de la Matriz Extracelular/química , Humanos , Peso Molecular , Tamaño de la Partícula , Viscosidad
10.
Cell Prolif ; : e13256, 2022 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-36574589

RESUMEN

OBJECTIVES: Induced pluripotent stem cells (iPSCs) generated by monolayer cultures is plagued by low efficiencies, high levels of manipulation and operator unpredictability. We have developed a platform, reprogramming, expansion, and differentiation on Microcarriers, to solve these challenges. MATERIALS AND METHODS: Five sources of human somatic cells were reprogrammed, selected, expanded and differentiated in microcarriers suspension cultures. RESULTS: Improvement of transduction efficiencies up to 2 times was observed. Accelerated reprogramming in microcarrier cultures was 7 days faster than monolayer, providing between 30 and 50-fold more clones to choose from fibroblasts, peripheral blood mononuclear cells, T cells and CD34+ stem cells. This was observed to be due to an earlier induction of genes (ß-catenin, E-cadherin and EpCAM) on day 4 versus monolayer cultures which occurred on days 14 or later. Following that, faster induction and earlier stabilization of pluripotency genes occurred during the maturation phase of reprogramming. Integrated expansion without trypsinization and efficient differentiation, without embryoid bodies formation, to the three germ-layers, cardiomyocytes and haematopoietic stem cells were further demonstrated. CONCLUSIONS: Our method can solve the inherent problems of conventional monolayer cultures. It is highly efficient, cell dissociation free, can be operated with lower labor, and allows testing of differentiation efficiency without trypsinization and generation of embryoid bodies. It is also amenable to automation for processing more samples in a small footprint, alleviating many challenges of manual monolayer selection.

11.
Cell Prolif ; 55(8): e13218, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35289971

RESUMEN

OBJECTIVES: Large-scale generation of universal red blood cells (RBCs) from O-negative (O-ve) human induced pluripotent stem cells (hiPSCs) holds the potential to alleviate worldwide shortages of blood and provide a safe and secure year-round supply. Mature RBCs and reticulocytes, the immature counterparts of RBCs generated during erythropoiesis, could also find important applications in research, for example in malaria parasite infection studies. However, one major challenge is the lack of a high-density culture platform for large-scale generation of RBCs in vitro. MATERIALS AND METHODS: We generated 10 O-ve hiPSC clones and evaluated their potential for mesoderm formation and erythroid differentiation. We then used a perfusion bioreactor system to perform studies with high-density cultures of erythroblasts in vitro. RESULTS: Based on their tri-lineage (and specifically mesoderm) differentiation potential, we isolated six hiPSC clones capable of producing functional erythroblasts. Using the best performing clone, we demonstrated the small-scale generation of high-density cultures of erythroblasts in a perfusion bioreactor system. After process optimization, we were able to achieve a peak cell density of 34.7 million cells/ml with 92.2% viability in the stirred bioreactor. The cells expressed high levels of erythroblast markers, showed oxygen carrying capacity, and were able to undergo enucleation. CONCLUSIONS: This study demonstrated a scalable platform for the production of functional RBCs from hiPSCs. The perfusion culture platform we describe here could pave the way for large volume-controlled bioreactor culture for the industrial generation of high cell density erythroblasts and RBCs.


Asunto(s)
Células Madre Pluripotentes Inducidas , Reactores Biológicos , Diferenciación Celular , Células Clonales , Eritrocitos , Eritropoyesis , Humanos , Perfusión
12.
Stem Cells ; 27(9): 2114-25, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19522013

RESUMEN

Insight into the regulation of core transcription factors is important for a better understanding of the molecular mechanisms that control self-renewal and pluripotency of human ESCs (hESCs). However, the transcriptional regulation of NANOG itself in hESCs has largely been elusive. We established a NANOG promoter luciferase reporter assay as a fast read-out for indicating the pluripotent status of hESCs. From the functional cDNA screens and NANOG promoter characterization, we successfully identified a zinc finger transcription factor KLF4 and a homeodomain transcription factor PBX1 as two novel transcriptional regulators that maintain the pluripotent and undifferentiated state of hESCs. We showed that both KLF4 and PBX1 mRNA and protein expression were downregulated during hESC differentiation. In addition, overexpression of KLF4 and PBX1 upregulated NANOG promoter activity and also the endogenous NANOG protein expression in hESCs. Direct binding of KLF4 on NANOG proximal promoter and PBX1 on a new upstream enhancer and proximal promoter were confirmed by chromatin immunoprecipitation and electrophoretic mobility shift assay. Knockdown of KLF4/PBX1 or mutation of KLF4/PBX1 binding motifs significantly downregulated NANOG promoter activity. We also showed that specific members of the SP/KLF and PBX family are functionally redundant at the NANOG promoter and that KLF4 and PBX1 cooperated with OCT4 and SOX2, and transactivated synergistically the NANOG promoter activity. Our results show two novel upstream transcription activators of NANOG that are functionally important for the self-renewal of hESC and provide new insights into the expanded regulatory circuitry that maintains hESC pluripotency.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Western Blotting , Línea Celular , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Ensayo de Cambio de Movilidad Electroforética , Células Madre Embrionarias , Proteínas de Homeodominio/genética , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Proteína Homeótica Nanog , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Stem Cell Res ; 44: 101738, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32109723

RESUMEN

Articular cartilage defects are one of the major challenges in orthopedic and trauma surgery. However, the poor ability of cartilage to self-repair has motivated efforts to engineer replacement tissues, and human mesenchymal stem cells (MSC), which have an extensive proliferation potential and can undergo chondrogenesis, have emerged as a promising cell source. In this review, we attempt to provide a brief overview of MSC isolation, characterization, current manufacturing platforms using various bioreactors, in vitro differentiation, and sealant-based or scaffold-based implantation.


Asunto(s)
Cartílago Articular , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Diferenciación Celular , Condrogénesis , Humanos , Ingeniería de Tejidos
14.
Stem Cell Res Ther ; 11(1): 118, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-32183888

RESUMEN

BACKGROUND: The production of large quantities of cardiomyocyte is essential for the needs of cellular therapies. This study describes the selection of a human-induced pluripotent cell (hiPSC) line suitable for production of cardiomyocytes in a fully integrated bioprocess of stem cell expansion and differentiation in microcarrier stirred tank reactor. METHODS: Five hiPSC lines were evaluated first for their cardiac differentiation efficiency in monolayer cultures followed by their expansion and differentiation compatibility in microcarrier (MC) cultures under continuous stirring conditions. RESULTS: Three cell lines were highly cardiogenic but only one (FR202) of them was successfully expanded on continuous stirring MC cultures. FR202 was thus selected for cardiac differentiation in a 22-day integrated bioprocess under continuous stirring in a stirred tank bioreactor. In summary, we integrated a MC-based hiPSC expansion (phase 1), CHIR99021-induced cardiomyocyte differentiation step (phase 2), purification using the lactate-based treatment (phase 3) and cell recovery step (phase 4) into one process in one bioreactor, under restricted oxygen control (< 30% DO) and continuous stirring with periodic batch-type media exchanges. High density of undifferentiated hiPSC (2 ± 0.4 × 106 cells/mL) was achieved in the expansion phase. By controlling the stirring speed and DO levels in the bioreactor cultures, 7.36 ± 1.2 × 106 cells/mL cardiomyocytes with > 80% Troponin T were generated in the CHIR99021-induced differentiation phase. By adding lactate in glucose-free purification media, the purity of cardiomyocytes was enhanced (> 90% Troponin T), with minor cell loss as indicated by the increase in sub-G1 phase and the decrease of aggregate sizes. Lastly, we found that the recovery period is important for generating purer and functional cardiomyocytes (> 96% Troponin T). Three independent runs in a 300-ml working volume confirmed the robustness of this process. CONCLUSION: A streamlined and controllable platform for large quantity manufacturing of pure functional atrial, ventricular and nodal cardiomyocytes on MCs in conventional-type stirred tank bioreactors was established, which can be further scaled up and translated to a good manufacturing practice-compliant production process, to fulfill the quantity requirements of the cellular therapeutic industry.


Asunto(s)
Células Madre Pluripotentes Inducidas , Reactores Biológicos , Técnicas de Cultivo de Célula , Diferenciación Celular , Células Cultivadas , Humanos , Miocitos Cardíacos
15.
Biotechnol Bioeng ; 102(3): 923-34, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18853410

RESUMEN

Genome-scale flux analysis of Escherichia coli DH5alpha growth in a complex medium was performed to investigate the relationship between the uptake of various nutrients and their metabolic outcomes. During the exponential growth phase, we observed a sequential consumption order of serine, aspartate and glutamate in the complex medium as well as the complete consumption of key carbohydrate nutrients, glucose and trehalose. Based on the consumption and production rates of the measured metabolites, constraints-based flux analysis of a genome-scale E. coli model was then conducted to elucidate their utilization in the metabolism. The in silico analysis revealed that the cell exploited biosynthetic precursors taken up directly from the complex medium, through growth-related anabolic pathways. This suggests that the cell could be functioning in an energetically more efficient manner by reducing the energy needed to produce amino acids. The in silico simulation also allowed us to explain the observed rapid consumption of serine: excessively consumed external serine from the complex medium was mainly converted into pyruvate and glycine, which in turn, led to the acetate accumulation. The present work demonstrates the application of an in silico modeling approach to characterizing microbial metabolism under complex medium condition. This work further illustrates the use of in silico genome-scale analysis for developing better strategies related to improving microbial growth and enhancing the productivity of desirable metabolites.


Asunto(s)
Biología Computacional/métodos , Escherichia coli/crecimiento & desarrollo , Escherichia coli/metabolismo , Acetatos/metabolismo , Aminoácidos/metabolismo , Reactores Biológicos , Simulación por Computador , Medios de Cultivo , Escherichia coli/genética , Genoma Bacteriano , Glucosa/metabolismo , Redes y Vías Metabólicas/fisiología , Modelos Biológicos , Serina/metabolismo
16.
Biotechnol Appl Biochem ; 52(Pt 1): 53-9, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18380624

RESUMEN

Well-characterized derivatives of Escherichia coli K12 such as DH5alpha are the host strains commonly used for plasmid DNA production. Owing to the prospective clinical demand for large quantities of plasmid DNA for gene therapy and DNA vaccination, existing plasmid production processes need to be optimized to attain higher plasmid yields. To date, nearly all production optimization efforts are focused on media or fermentation process design. Although there has been a simple empirical evaluation of the available host strains, there is a lack of systematic effort at engineering these host strains for improved plasmid DNA production. In view of this, we engineered DH5alpha WT (wild-type) cells carrying a DNA vaccine plasmid by knocking out the fruR (fructose repressor) [also known as the Cra (catabolite repressor activator)] global regulator gene and evaluated the growth and plasmid yields of these P+ (plasmid-bearing) fruR cells (fruR-knockout cells) during fed-batch cultures with exponential feeding. The P+ fruR cells showed a more rapid accumulation of plasmid DNA towards the end of the fed-batch cultures compared with the P+ WT cells. As a result, the specific plasmid yield of the P+ fruR cells was 21% higher than that of the P+ WT cells [19.2 versus 15.9 mg/g DCW (dry cell weight)]. These results demonstrate that, from an initial high-yielding fermentation process, the knockout of the fruR global regulator gene in E. coli DH5alpha further improves plasmid yields during fed-batch culture.


Asunto(s)
Proteínas de Escherichia coli/genética , Escherichia coli/genética , Técnicas de Inactivación de Genes/métodos , Plásmidos/biosíntesis , Proteínas Represoras/genética , Electroforesis en Gel de Agar , Escherichia coli/crecimiento & desarrollo , Espectrometría de Fluorescencia
17.
Biotechnol Lett ; 31(5): 653-8, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19169887

RESUMEN

An automated vision system, TeratomEye, was developed for the identification of three representative tissue types: muscle, gut and neural epithelia which are commonly found in teratomas formed from human embryonic stem cells. Muscle tissue, a common structure was identified with an accuracy of 90.3% with high specificity and sensitivity greater than 90%. Gut epithelia were identified with an accuracy of 87.5% with specificity and sensitivity greater than 80%. Neural epithelia which were the most difficult structures to distinguish gave an accuracy of 47.6%. TeratomEye is therefore useful for the automated identification of differentiated tissues in teratoma sections.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Mucosa Intestinal/citología , Células Musculares/citología , Células Neuroepiteliales/citología , Técnicas Citológicas/métodos , Humanos
18.
Biotechnol Adv ; 36(8): 2118-2128, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30273713

RESUMEN

In vitro generation of red blood cells (RBCs) has the potential to circumvent the shortfalls in global demand for blood for transfusion applications. The conventional approach for RBC generation has been from differentiation of hematopoietic stem cells (HSCs) derived from cord blood, adult bone marrow or peripheral blood. More recently, RBCs have been generated from human induced pluripotent stem cells (hiPSCs) as well as from immortalized adult erythroid progenitors. In this review, we highlight the recent advances to RBC generation from these different approaches and discuss the challenges and new strategies that can potentially make large-scale in vitro generation of RBCs a feasible approach.


Asunto(s)
Técnicas de Cultivo de Célula , Eritrocitos , Medicina Transfusional , Animales , Diferenciación Celular , Sangre Fetal/citología , Células Madre Hematopoyéticas , Humanos , Células Madre Pluripotentes Inducidas/citología , Ratones
19.
Biotechnol J ; 13(4): e1700567, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29330927

RESUMEN

Anticipated shortages in donated blood supply have prompted investigation of alternative approaches for in vitro production of red blood cells (RBCs), such as expansion of conditional immortalization erythroid progenitors. However, there is a bioprocessing challenge wherein factors promoting maximal cell expansion and growth-limiting inhibitory factors are yet to be investigated. The authors use an erythroblast cell line (ImEry) derived from immortalizing CD71+CD235a+ erythroblast from adult peripheral blood for optimization of expansion culture conditions. Design of experiments (DOE) is used in media formulation to explore relationships and interactive effects between factors which affect cell expansion. Our in-house optimized medium formulation produced significantly higher cell densities (3.62 ± 0.055) × 106 cells mL-1 , n = 3) compared to commercial formulations (2.07 ± 0.055) × 106 cells mL-1 , n = 3; at 209 h culture). Culture media costs per unit of blood is shown to have a 2.96-3.09 times cost reduction. As a proof of principle for scale up, ImEry are expanded in a half-liter stirred-bioreactor under controlled settings. Growth characteristics, metabolic, and molecular profile of the cells are evaluated. ImEry has identical O2 binding capacity to adult erythroblasts. Amino acid supplementation results in further yield improvements. The study serves as a first step for scaling up erythroblast expansion in controlled bioreactors.


Asunto(s)
Técnicas de Cultivo Celular por Lotes/métodos , Medio de Cultivo Libre de Suero/química , Eritroblastos/citología , Reactores Biológicos , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Eritroblastos/química , Humanos , Proteínas Proto-Oncogénicas c-myc/genética , Proteína bcl-X/genética
20.
J Biotechnol ; 130(3): 320-8, 2007 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-17544536

RESUMEN

The maintenance of undifferentiated human embryonic stem cells (hESC) requires feeder cells, either in co-culture or feeder-free with conditioned medium (CM) from the feeders. In this study, we compared the CM of a supporting primary mouse embryonic feeder (MEF) and an isogenic but non-supporting MEF line (DeltaE-MEF) in order to gain an insight to the differential expression profile of secreted factors. Using two-dimensional electrophoresis (2-DE) and matrix-assisted laser desorption/ionization-time of flight (MALDI) tandem mass spectrometry, 13 protein identities were found to be downregulated in DeltaE-MEF compared to MEF, of which 4 were found to be soluble factors and 3 proteins were membrane-associated or related to the extracellular matrix. In addition, four other proteins were identified to be differentially expressed in MEF-CM using high pressure liquid chromatography (HPLC) and cytokine arrays. In functional experiments where CM was replaced with six of the factors identified, hESC were able to proliferate for five continuous passages whilst maintaining 68-82% and 74-98% expression of pluripotent markers, Oct-4 and Tra-1-60, respectively. Using proteomic tools, important proteins from CM that supports hESC culture have been identified, which when replaced with recombinant proteins, continue to support undifferentiated hESC growth in a feeder-free culture platform.


Asunto(s)
Células Madre Embrionarias/metabolismo , Proteínas/análisis , Animales , Células Cultivadas , Medios de Cultivo Condicionados , Medio de Cultivo Libre de Suero , Citocinas/metabolismo , Electroforesis en Gel Bidimensional , Células Madre Embrionarias/citología , Fibroblastos/citología , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Ratones , Factores de Transcripción de Octámeros/metabolismo , Proteínas/genética , Proteínas Recombinantes/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA