Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Food Sci ; 89(2): 851-865, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38174744

RESUMEN

Cell-based meat technology provides an effective method to meet the demand for meat, while also posing a huge challenge to the expansion of myoblasts. It is difficult to develop serum-free medium suitable for long-term culture and large-scale expansion of myoblasts, which causes limited understanding of myoblasts expansion. Therefore, this study used C2C12 myoblasts as model cells and developed a serum-free medium for large-scale expansion of myoblasts in vitro using the Plackett-Burman design. The serum-free medium can support short-term proliferation and long-term passage of C2C12 myoblasts, while maintaining myogenic differentiation potential well, which is comparable to those of growth medium containing 10% fetal bovine serum. Based on the C2C12 myoblasts microcarriers serum-free culture system established in this study, the actual expansion folds of myoblasts can reach 43.55 folds after 7 days. Moreover, cell-based meat chunks were preliminarily prepared using glutamine transaminase and edible pigments. The research results provide reference for serum-free culture and large-scale expansion of myoblasts in vitro, laying the foundation for cell-based meat production. PRACTICAL APPLICATION: This study developed a serum-free medium suitable for long-term passage of myoblasts and established a microcarrier serum-free culture system for myoblasts, which is expected to solve the problem of serum-free culture and large-scale expansion of myoblasts in cell culture meat production.


Asunto(s)
Técnicas de Cultivo de Célula , Carne in Vitro , Proliferación Celular , Técnicas de Cultivo de Célula/métodos , Mioblastos , Diferenciación Celular
2.
Stem Cells Cloning ; 16: 61-73, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37790697

RESUMEN

Purpose: Two-dimensional (2D)-based cell culture systems, limited by their inherent heterogeneity and scalability, are a bottleneck in the production of high-quality cells for downstream biomedical applications. Finding the optimal conditions for large-scale stem cell culture while maintaining good cellular status is challenging. The aim of this study was to assess the effects of three-dimensional (3D) culture on the viability, proliferation, self-renewal, and differentiation of human induced pluripotent stem cells (IPSCs). Patients and Methods: Various culture conditions were evaluated to determine the optimal conditions to maintain the viability and proliferation of human IPSCs in a 3D environment: static versus dynamic culture, type of adhesion protein added to alginate (Matrigel™ versus gelatin), and the addition of Y-27632t on long-term 3D culture. The proliferation ability of the cells was evaluated via the MTS proliferation assay; the expression levels of the pluripotency markers Nanog and Oct3/4, PAX6 as an ectoderm marker, and laminin-5 and fibronectin as markers of extracellular matrix synthesis were assessed; and HIF1α and HIF2α levels were measured using quantitative reverse transcription polymerase chain reaction. Results: Using a high-aspect-ratio vessel bioreactor with a gentle, low-sheer, and low-turbulence environment with sufficient oxygenation and effective mass transfer of nutrients and waste, we verified its ability to promote cell proliferation and self-renewal. The findings showed that human IPSCs have the ability to maintain pluripotency in a feeder-free system and by inhibiting ROCK signaling and using hypoxia to improve single-cell viability in 3D culture. Furthermore, these cells demonstrated increased self-renewal and proliferation when inoculated as single cells in 3D alginate beads by adding RI during the culture period. Conclusion: Dynamic 3D culture is desirable for the large-scale expansion of undifferentiated human IPSCs.

3.
Tissue Eng Part C Methods ; 29(9): 424-437, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37395490

RESUMEN

Allogeneic chondrocyte therapies need to be developed to allow more individuals to be treated with a cell therapy for cartilage repair and to reduce the burden and cost of the current two-stage autologous procedures. Upscale manufacture of chondrocytes using a bioreactor could help provide an off-the-shelf allogeneic chondrocyte therapy with many doses being produced in a single manufacturing run. In this study, we assess a good manufacturing practice-compliant hollow-fiber bioreactor (Quantum®) for adult chondrocyte manufacture. Chondrocytes were isolated from knee arthroplasty-derived cartilage (n = 5) and expanded in media supplemented with 10% fetal bovine serum (FBS) or 5% human platelet lysate (hPL) on tissue culture plastic (TCP) for a single passage. hPL-supplemented cultures were then expanded in the Quantum bioreactor for a further passage. Matched, parallel cultures in hPL or FBS were maintained on TCP. Chondrocytes from all culture conditions were characterized in terms of growth kinetics, morphology, immunoprofile, chondrogenic potential (chondrocyte pellet assays), and single telomere length analysis. Quantum expansion of chondrocytes resulted in 86.4 ± 38.5 × 106 cells in 8.4 ± 1.5 days, following seeding of 10.2 ± 3.6 × 106 cells. This related to 3.0 ± 1.0 population doublings in the Quantum bioreactor, compared with 2.1 ± 0.6 and 1.3 ± 1.0 on TCP in hPL- and FBS-supplemented media, respectively. Quantum- and TCP-expanded cultures retained equivalent chondropotency and mesenchymal stromal cell marker immunoprofiles, with only the integrin marker, CD49a, decreasing following Quantum expansion. Quantum-expanded chondrocytes demonstrated equivalent chondrogenic potential (as assessed by ability to form and maintain chondrogenic pellets) with matched hPL TCP populations. hPL manufacture, however, led to reduced chondrogenic potential and increased cell surface positivity of integrins CD49b, CD49c, and CD51/61 compared with FBS cultures. Quantum expansion of chondrocytes did not result in shortened 17p telomere length when compared with matched TCP cultures. This study demonstrates that large numbers of adult chondrocytes can be manufactured in the Quantum hollow-fiber bioreactor. This rapid, upscale expansion does not alter chondrocyte phenotype when compared with matched TCP expansion. Therefore, the Quantum provides an attractive method of manufacturing chondrocytes for clinical use. Media supplementation with hPL for chondrocyte expansion may, however, be unfavorable in terms of retaining chondrogenic capacity.


Asunto(s)
Condrocitos , Trasplante de Células Madre Hematopoyéticas , Adulto , Humanos , Cartílago , Células Cultivadas , Matriz Extracelular/metabolismo , Diferenciación Celular , Proliferación Celular
4.
Bioresour Bioprocess ; 10(1): 18, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36915643

RESUMEN

Mesenchymal stem cells (MSCs) are highly important in biomedicine and hold great potential in clinical treatment for various diseases. In recent years, the capabilities of MSCs have been under extensive investigation for practical application. Regarding therapy, the efficacy usually depends on the amount of MSCs. Nevertheless, the yield of MSCs is still limited due to the traditional cultural methods. Herein, we proposed a three-dimensional (3D) scaffold prepared using poly lactic-co-glycolic acid (PLGA) nanofiber with polylysine (PLL) grafting, to promote the growth and proliferation of MSCs derived from the human umbilical cord (hUC-MSCs). We found that the inoculated hUC-MSCs adhered efficiently to the PLGA scaffold with good affinity, fast growth rate, and good multipotency. The harvested cells were ideally distributed on the scaffold and we were able to gain a larger yield than the traditional culturing methods under the same condition. Thus, our cell seeding with a 3D scaffold could serve as a promising strategy for cell proliferation in the large-scale production of MSCs. Moreover, the simplicity and low preparation cost allow this 3D scaffold to extend its potential application beyond cell culture. Supplementary Information: The online version contains supplementary material available at 10.1186/s40643-023-00635-6.

5.
Methods Mol Biol ; 2436: 113-125, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34519979

RESUMEN

Mesenchymal stem cells show great potential in tissue engineering and cell-based therapies. This protocol demonstrates the use of 3D TableTrix® microcarrier tablets for large-scale manufacturing of human umbilical cord mesenchymal stem cells (hUCMSCs) in a 5-L stirred tank bioreactor. 3D TableTrix® microcarrier tablets readily disperse into tens of thousands of porous microcarriers to simplify cell seeding, and 3D FloTrix® vivaSPIN bioreactor could automate, monitor, and control the entire culture process. 3D TableTrix® microcarriers could also be fully dissolved upon adding dissolution reagent to gently harvest the expanded cells at a high recovery rate. With this protocol, more than 109 cells could be produced in a 5-L bioreactor.


Asunto(s)
Técnicas de Cultivo de Célula , Células Madre Mesenquimatosas , Reactores Biológicos , Técnicas de Cultivo de Célula/métodos , Humanos , Comprimidos , Cordón Umbilical
6.
Int J Mol Sci ; 22(7)2021 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-33808241

RESUMEN

Around 40% of the population will suffer at some point in their life a disease involving tissue loss or an inflammatory or autoimmune process that cannot be satisfactorily controlled with current therapies. An alternative for these processes is represented by stem cells and, especially, mesenchymal stem cells (MSC). Numerous preclinical studies have shown MSC to have therapeutic effects in different clinical conditions, probably due to their mesodermal origin. Thereby, MSC appear to play a central role in the control of a galaxy of intercellular signals of anti-inflammatory, regenerative, angiogenic, anti-fibrotic, anti-oxidative stress effects of anti-apoptotic, anti-tumor, or anti-microbial type. This concept forces us to return to the origin of natural physiological processes as a starting point to understand the evolution of MSC therapy in the field of regenerative medicine. These biological effects, demonstrated in countless preclinical studies, justify their first clinical applications, and draw a horizon of new therapeutic strategies. However, several limitations of MSC as cell therapy are recognized, such as safety issues, handling difficulties for therapeutic purposes, and high economic cost. For these reasons, there is an ongoing tendency to consider the use of MSC-derived secretome products as a therapeutic tool, since they reproduce the effects of their parent cells. However, it will be necessary to resolve key aspects, such as the choice of the ideal type of MSC according to their origin for each therapeutic indication and the implementation of new standardized production strategies. Therefore, stem cell science based on an intelligently designed production of MSC and or their derivative products will be able to advance towards an innovative and more personalized medical biotechnology.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas/métodos , Trasplante de Células Madre Mesenquimatosas/tendencias , Células Madre Mesenquimatosas/metabolismo , Animales , Exosomas/metabolismo , Exosomas/trasplante , Humanos , Medicina Regenerativa/métodos , Medicina Regenerativa/tendencias
7.
Neurochem Res ; 45(1): 204-214, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31828497

RESUMEN

Neurodegenerative disorders present a broad group of neurological diseases and remain one of the greatest challenges and burdens to mankind. Maladies like amyotrophic lateral sclerosis, Alzheimer's disease, stroke or spinal cord injury commonly features astroglia involvement (astrogliosis) with signs of inflammation. Regenerative, paracrine and immunomodulatory properties of human mesenchymal stromal cells (hMSCs) could target the above components, thus opening new therapeutic possibilities for regenerative medicine. A special interest should be given to hMSCs derived from the umbilical cord (UC) tissue, due to their origin, properties and lack of ethical paradigms. The aim of this study was to establish standard operating and scale-up good manufacturing practice (GMP) protocols of UC-hMSCs isolation, characterization, expansion and comparison of cells' properties when harvested on T-flasks versus using a large-scale bioreactor system. Human UC-hMSCs, isolated by tissue explant culture technique from Wharton's jelly, were harvested after reaching 75% confluence and cultured using tissue culture flasks. Obtained UC-hMSCs prior/after the cryopreservation and after harvesting in a bioreactor, were fully characterized for "mesenchymness" immunomodulatory, tumorigenicity and genetic stability, senescence and cell-doubling properties, as well as gene expression features. Our study demonstrates an efficient and simple technique for large scale UC-hMSCs expansion. Harvesting of UC-hMSCs' using classic and large scale methods did not alter UC-hMSCs' senescence, genetic stability or in vitro tumorigenicity features. We observed comparable growth and immunomodulatory capacities of fresh, frozen and expanded UC-hMSCs. We found no difference in the ability to differentiate toward adipogenic, osteogenic and chondrogenic lineages between classic and large scale UC-hMSCs expansion methods. Both, methods enabled derivation of genetically stabile cells with typical mesenchymal features. Interestingly, we found significantly increased mRNA expression levels of neural growth factor (NGF) and downregulated insulin growth factor (IGF) in UC-hMSCs cultured in bioreactor, while IL4, IL6, IL8, TGFb and VEGF expression levels remained at the similar levels. A culturing of UC-hMSCs using a large-scale automated closed bioreactor expansion system under the GMP conditions does not alter basic "mesenchymal" features and quality of the cells. Our study has been designed to pave a road toward translation of basic research data known about human UC-MSCs for the future clinical testing in patients with neurological and immunocompromised disorders. An industrial manufacturing of UC-hMSCs next will undergo regulatory approval following advanced therapy medicinal products (ATMP) criteria prior to clinical application and approval to be used in patients.


Asunto(s)
Reactores Biológicos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Enfermedades del Sistema Nervioso/terapia , Cordón Umbilical/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Humanos , Trasplante de Células Madre Mesenquimatosas/tendencias , Enfermedades del Sistema Nervioso/patología , Cordón Umbilical/citología , Cordón Umbilical/trasplante , Gelatina de Wharton/citología , Gelatina de Wharton/fisiología , Gelatina de Wharton/trasplante
8.
Stem Cell Res Ther ; 10(1): 99, 2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30885254

RESUMEN

BACKGROUND: The manufacture of mesenchymal stem/stromal cells (MSCs) for clinical use needs to be cost effective, safe and scaled up. Current methods of expansion on tissue culture plastic are labour-intensive and involve several 'open' procedures. We have used the closed Quantum® hollow fibre bioreactor to expand four cultures each of MSCs derived from bone marrow (BM) and, for the first time, umbilical cords (UCs) and assessed extensive characterisation profiles for each, compared to parallel cultures grown on tissue culture plastic. METHODS: Bone marrow aspirate was directly loaded into the Quantum®, and cells were harvested and characterised at passage (P) 0. Bone marrow cells were re-seeded into the Quantum®, harvested and further characterised at P1. UC-MSCs were isolated enzymatically and cultured once on tissue culture plastic, before loading cells into the Quantum®, harvesting and characterising at P1. Quantum®-derived cultures were phenotyped in terms of immunoprofile, tri-lineage differentiation, response to inflammatory stimulus and telomere length, as were parallel cultures expanded on tissue culture plastic. RESULTS: Bone marrow cell harvests from the Quantum® were 23.1 ± 16.2 × 106 in 14 ± 2 days (P0) and 131 ± 84 × 106 BM-MSCs in 13 ± 1 days (P1), whereas UC-MSC harvests from the Quantum® were 168 ± 52 × 106 UC-MSCs after 7 ± 2 days (P1). Quantum®- and tissue culture plastic-expanded cultures at P1 adhered to criteria for MSCs in terms of cell surface markers, multipotency and plastic adherence, whereas the integrins, CD29, CD49c and CD51/61, were found to be elevated on Quantum®-expanded BM-MSCs. Rapid culture expansion in the Quantum® did not cause shortened telomeres when compared to cultures on tissue culture plastic. Immunomodulatory gene expression was variable between donors but showed that all MSCs upregulated indoleamine 2, 3-dioxygenase (IDO). CONCLUSIONS: The results presented here demonstrate that the Quantum® can be used to expand large numbers of MSCs from bone marrow and umbilical cord tissues for next-generation large-scale manufacturing, without impacting on many of the properties that are characteristic of MSCs or potentially therapeutic. Using the Quantum®, we can obtain multiple MSC doses from a single manufacturing run to treat many patients. Together, our findings support the development of cheaper cell-based treatments.


Asunto(s)
Células de la Médula Ósea/citología , Técnicas de Cultivo de Célula , Diferenciación Celular , Separación Celular , Células Madre Mesenquimatosas/citología , Cordón Umbilical/citología , Adulto , Células de la Médula Ósea/metabolismo , Humanos , Recién Nacido , Masculino , Células Madre Mesenquimatosas/metabolismo , Cordón Umbilical/metabolismo
9.
Biotechnol Bioeng ; 115(7): 1717-1728, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29578573

RESUMEN

With recent advances in biotechnology, mammalian cells are used in biopharmaceutical industries to produce valuable protein therapeutics and investigated as effective therapeutic agents to permanently degenerative diseases in cell based therapy. In these exciting and actively expanding fields, a reliable, efficient, and affordable platform to culture mammalian cells on a large scale is one of the most vital necessities. To produce and maintain a very large population of anchorage-dependent cells, a microcarrier-based stirred tank bioreactor is commonly used. In this approach, the cells are exposed to harmful hydrodynamic shear stress in the bioreactor and the mass transfer rates of nutrients and gases in the bioreactor are often kept below an optimal level to prevent cellular damages from the shear stress. In this paper, a hollow microcarrier (HMC) is presented as a novel solution to protect cells from shear stress in stirred bioreactors, while ensuring sufficient and uniform mass transfer rate of gases and nutrients. HMC is a hollow microsphere and cells are cultured on its inner surface to be protected, while openings on the HMC provide sufficient exchange of media inside the HMC. As a proof of concept, we demonstrated the expansion of fibroblasts, NIH/3T3 and the expansion and cardiac differentiation of human induced pluripotent stem cells, along with detailed numerical analysis. We believe that the developed HMC can be a practical solution to enable large-scale expansion of shear-sensitive anchorage-dependent cells in an industrial scale with stirred bioreactors.


Asunto(s)
Reactores Biológicos , Adhesión Celular , Técnicas de Cultivo de Célula/métodos , Células Inmovilizadas/fisiología , Microesferas , Animales , Biotecnología/métodos , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Ratones , Células 3T3 NIH/fisiología , Tecnología Farmacéutica/métodos
10.
Colloids Surf B Biointerfaces ; 159: 62-77, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28780462

RESUMEN

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.


Asunto(s)
Células Madre/citología , Animales , Materiales Biocompatibles/química , Reactores Biológicos , Técnicas de Cultivo de Célula , Proliferación Celular/fisiología , Humanos , Ingeniería de Tejidos/métodos
11.
Methods Mol Biol ; 1441: 195-202, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27177667

RESUMEN

Recent advances in methods for the ex vivo expansion of human natural killer (NK) cells have facilitated the use of these powerful immune cells in clinical protocols. Further, the ability to genetically modify primary human NK cells following rapid expansion allows targeting and enhancement of their immune function. We have successfully adapted an expansion method for primary NK cells from peripheral blood mononuclear cells or from apheresis products in gas permeable rapid expansion devices (G-Rexes). Here, we describe an optimized protocol for rapid and robust NK cell expansion as well as a method for highly efficient retroviral transduction of these ex vivo expanded cells. These methodologies are good manufacturing practice (GMP) compliant and could be used for clinical-grade product manufacturing.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Asesinas Naturales/citología , Transducción Genética , Eliminación de Componentes Sanguíneos , Proliferación Celular , Trasplante de Células , Células Nutrientes/citología , Células Nutrientes/inmunología , Células Nutrientes/metabolismo , Humanos , Células K562 , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Activación de Linfocitos
12.
J Tissue Eng Regen Med ; 10(2): 108-19, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23495227

RESUMEN

The regenerative potential of mesenchymal stromal or stem cells (MSCs) has generated tremendous interest for treating various degenerative diseases. Regulatory preference is to use a culture medium that is devoid of bovine components for stem cell expansion intended for therapeutic applications. However, a clear choice an alternative to fetal bovine serum (FBS) has not yet emerged. We have screened five different commercially available serum-free media (SFM) for their ability to support the growth and expansion of pre-isolated undifferentiated bone marrow-derived MSCs (BM-MSCs) and compared the results with cells grown in standard FBS-containing medium as control. In addition, based on initial screening results, BD Mosaic™ Mesenchymal Stem Cell Serum-free (BD-SFM) medium was evaluated in large-scale cultures for the performance and culture characteristics of BM-MSCs. Of the five different serum-free media, BD-SFM enhanced BM-MSCs growth and expansion in Cell STACK (CS), but the cell yield per CS-10 was less when compared to the control medium. The characteristics of MSCs were measured in terms of population doubling time (PDT), cell yield and expression of MSC-specific markers. Significant differences were observed between BD-SFM and control medium in terms of population doublings (PDs), cell yield, CFU-F and morphological features, whereas surface phenotype and differentiation potentials were comparable. The BD-SFM-cultured MSCs were also found to retain the differentiation potential, immune-privileged status and immunosuppressive properties inherent to MSCs. Our results suggest that BD-SFM supports large-scale expansion of BM-MSCs for therapeutic use.


Asunto(s)
Células de la Médula Ósea/citología , Separación Celular/métodos , Células Madre Mesenquimatosas/citología , Bioensayo , Diferenciación Celular , Proliferación Celular , Forma de la Célula , Ensayo de Unidades Formadoras de Colonias , Medio de Cultivo Libre de Suero , Humanos , Inmunomodulación , Inmunofenotipificación , Terapia de Inmunosupresión , Cinética , Albúmina Sérica Bovina/metabolismo , Adulto Joven
13.
Philos Trans R Soc Lond B Biol Sci ; 370(1661): 20140040, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25533097

RESUMEN

Anchorage-dependent cells are of great interest for various biotechnological applications. (i) They represent a formidable production means of viruses for vaccination purposes at very large scales (in 1000-6000 l reactors) using microcarriers, and in the last decade many more novel viral vaccines have been developed using this production technology. (ii) With the advent of stem cells and their use/potential use in clinics for cell therapy and regenerative medicine purposes, the development of novel culture devices and technologies for adherent cells has accelerated greatly with a view to the large-scale expansion of these cells. Presently, the really scalable systems--microcarrier/microcarrier-clump cultures using stirred-tank reactors--for the expansion of stem cells are still in their infancy. Only laboratory scale reactors of maximally 2.5 l working volume have been evaluated because thorough knowledge and basic understanding of critical issues with respect to cell expansion while retaining pluripotency and differentiation potential, and the impact of the culture environment on stem cell fate, etc., are still lacking and require further studies. This article gives an overview on critical issues common to all cell culture systems for adherent cells as well as specifics for different types of stem cells in view of small- and large-scale cell expansion and production processes.


Asunto(s)
Adhesión Celular/fisiología , Células Inmovilizadas/fisiología , Células Inmovilizadas/virología , Vacunas Virales , Replicación Viral , Animales , Células Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA