Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cytotherapy ; 26(7): 749-756, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38506771

RESUMEN

BACKGROUND & AIMS: Cell therapies based on mesenchymal stromal cells (MSCs) have gained an increasing therapeutic interest in the context of multiple disorders. Nonetheless, this field still faces important challenges, particularly concerning suitable manufacturing platforms. Here, we aimed at establishing a scalable culture system to expand umbilical cord-derived Wharton's jelly MSC (MSC(WJ)) and their derived extracellular vesicles (EVs) by using dissolvable microcarriers combined with xeno(geneic)-free culture medium. METHODS: MSC(WJ) isolated from three donors were cultured at a starting density of 1 × 106 cells per spinner flask, i.e., 2.8 × 103 cells per cm2 of dissolvable microcarrier surface area. After a 6-day expansion period of MSC(WJ), extracellular vesicles (EVs) were produced for 24 h. RESULTS: Taking advantage of an intermittent agitation regimen, we observed high adhesion rates to the microcarriers (over 90% at 24 h) and achieved 15.8 ± 0.7-fold expansion after 6 days of culture. Notably, dissolution of the microcarriers was achieved through a pectinase-based solution to recover the cell product, reducing the hurdles of downstream processing. MSC identity was validated by detecting the characteristic MSC immunophenotype and by multilineage differentiation assays. Considering the growing interest in MSC-derived EVs, which are known to be mediators of the therapeutic features of MSC, this platform also was evaluated for EV production. Upon a 24-h period of conditioning, secreted EVs were isolated by ultrafiltration followed by anion-exchange chromatography and exhibited the typical cup-shaped morphology, small size distribution (162.6 ± 30.2 nm) and expressed EV markers (CD63, CD9 and syntenin-1). CONCLUSIONS: Taken together, we established a time-effective and robust scalable platform that complies with clinical-grade standards for the dual production of MSC(WJ) and their derived EV.


Asunto(s)
Técnicas de Cultivo de Célula , Diferenciación Celular , Vesículas Extracelulares , Células Madre Mesenquimatosas , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Vesículas Extracelulares/metabolismo , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Proliferación Celular , Cordón Umbilical/citología , Gelatina de Wharton/citología
2.
Mol Carcinog ; 62(6): 845-854, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36994661

RESUMEN

Cutaneous squamous cell carcinoma (CSCC), which develops in response to ultraviolet irradiation exposure, is among the most common cancers. CSCC lesions can be removed by surgical excision, but 4.5% of these cancers reappear as aggressive and therapy-resistant tumors. CSCC tumors display a high mutation burden, and tumor frequency is dramatically increased in immune-suppressed patients, indicating a vital role for the immune system in controlling cancer development. Natural killer cells (NK cells) play a key role in cancer immune surveillance, and recent studies suggest that NK cells from healthy donors can be expanded from peripheral blood for use in therapy. In the present study, we test the ability of ex vivo expanded human NK cells to suppress the CSCC cell cancer phenotype and reduce tumor growth. We expanded human NK cells from multiple healthy donors, in the presence of IL-2, and tested their ability to suppress the CSCC cell cancer phenotype. NK cell treatment produced a dose-dependent reduction in SCC-13 and HaCaT cell spheroid growth and matrigel invasion and induced SCC-13 and HaCaT cell apoptosis as evidenced by increased procaspase 9, procaspase 3, and PARP cleavage. Moreover, two important CSCC cell pro-cancer signaling pathways, YAP1/TAZ/TEAD and MEK1/2-ERK1/2, were markedly reduced. Furthermore, tail-vein injection of NK cells markedly suppressed the growth of SCC-13 xenograft tumors in NSG mice, which was also associated with a reduction in YAP1 and MEK1/2-P levels and enhanced apoptosis. These findings show that NK cell treatment suppresses CSCC cell spheroid formation, invasion, viability, and tumor growth, suggesting NK cell treatment may be a candidate therapy for CSCC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias Cutáneas , Humanos , Animales , Ratones , Supervivencia Celular , Células Asesinas Naturales , Apoptosis
3.
Cytotherapy ; 17(9): 1169-77, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26276001

RESUMEN

Mesenchymal stem/stromal cells (MSCs) have been extensively investigated for their regenerative, immune-modulatory, and wound healing properties. While the laboratory studies have suggested that MSC's have a unique potential for modulating the etiopathology of multiple diseases, the results from clinical trials have not been encouraging or reproducible. One of the explanations for such variability is explained by the "art" of isolating and propagating MSCs. Therefore, establishing more than minimal criteria to define MSC would help understand best protocols to isolate, propagate and deliver MSCs. Developing a calibration standard, a database and a set of functional tests would be a better quality metric for MSCs. In this review, we discuss the importance of selecting a standard, issues associated with coming up with such a standard and how these issues can be mitigated.


Asunto(s)
Separación Celular/normas , Células Madre Mesenquimatosas/citología , Técnicas de Cultivo de Célula , Células Cultivadas , Humanos , Trasplante de Células Madre Mesenquimatosas/métodos , Estándares de Referencia
4.
Biochim Biophys Acta ; 1830(2): 2427-34, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22820019

RESUMEN

BACKGROUND: The epidermis is an important protective barrier that is essential for maintenance of life. Maintaining this barrier requires continuous cell proliferation and differentiation. Moreover, these processes must be balanced to produce a normal epidermis. The stem cells of the epidermis reside in specific locations in the basal epidermis, hair follicle and sebaceous glands and these cells are responsible for replenishment of this tissue. SCOPE OF REVIEW: A great deal of effort has gone into identifying protein epitopes that mark stem cells, in identifying stem cell niche locations, and in understanding how stem cell populations are related. We discuss these studies as they apply to understanding normal epidermal homeostasis and skin cancer. MAJOR CONCLUSIONS: An assortment of stem cell markers have been identified that permit assignment of stem cells to specific regions of the epidermis, and progress has been made in understanding the role of these cells in normal epidermal homeostasis and in conditions of tissue stress. A key finding is the multiple stem cell populations exist in epidermis that give rise to different structures, and that multiple stem cell types may contribute to repair in damaged epidermis. GENERAL SIGNIFICANCE: Understanding epidermal stem cell biology is likely to lead to important therapies for treating skin diseases and cancer, and will also contribute to our understanding of stem cells in other systems. This article is part of a Special Issue entitled Biochemistry of Stem Cells.


Asunto(s)
Células Epidérmicas , Células Madre/citología , Epidermis/metabolismo , Humanos
5.
Biotechnol Bioeng ; 111(6): 1116-27, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24420557

RESUMEN

The large cell doses (>1 × 10(6) cells/kg) used in clinical trials with mesenchymal stem/stromal cells (MSC) will require an efficient production process. Moreover, monitoring and control of MSC ex-vivo expansion is critical to provide a safe and reliable cell product. Bioprocess engineering approaches, such as bioreactor technology, offer the adequate tools to develop and optimize a cost-effective culture system for the rapid expansion of human MSC for cellular therapy. Herein, a xenogeneic (xeno)-free microcarrier-based culture system was successfully established for bone marrow (BM) MSC and adipose tissue-derived stem/stromal cell (ASC) cultivation using a 1L-scale controlled stirred-tank bioreactor, allowing the production of (1.1 ± 0.1) × 10(8) and (4.5 ± 0.2) × 10(7) cells for BM MSC and ASC, respectively, after 7 days. Additionally, the effect of different percent air saturation values (%Airsat ) and feeding regime on the proliferation and metabolism of BM MSC was evaluated. No significant differences in cell growth and metabolic patterns were observed under 20% and 9%Airsat . Also, the three different feeding regimes studied-(i) 25% daily medium renewal, (ii) 25% medium renewal every 2 days, and (iii) fed-batch addition of concentrated nutrients and growth factors every 2 days-yielded similar cell numbers, and only slight metabolic differences were observed. Moreover, the immunophenotype (positive for CD73, CD90 and CD105 and negative for CD31, CD80 and HLA-DR) and multilineage differentiative potential of expanded cells were not affected upon bioreactor culture. These results demonstrated the feasibility of expanding human MSC from different sources in a clinically relevant expansion configuration in a controlled microcarrier-based stirred culture system under xeno-free conditions. The further optimization of this bioreactor culture system will represent a crucial step towards an efficient GMP-compliant clinical-scale MSC production system.


Asunto(s)
Reactores Biológicos , Proliferación Celular , Células Madre Mesenquimatosas/fisiología , Células del Estroma/fisiología , Técnicas de Cultivo de Célula/métodos , Humanos , Inmunofenotipificación
6.
Regen Ther ; 27: 39-47, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38496009

RESUMEN

Induced pluripotent stem cells (iPSCs) have immense potential for use in disease modeling, etiological studies, and drug discovery. However, the current workflow for iPSC generation and maintenance poses challenges particularly during the establishment phase when specialized skills are required. Although three-dimensional culture systems offer scalability for maintaining established iPSCs, the enzymatic dissociation step is complex and time-consuming. In this study, a novel approach was developed to address these challenges by enabling iPSC generation, maintenance, and differentiation without the need for two-dimensional culture or enzymatic dissociation. This streamlined method offers a more convenient workflow, reduces variability and labor for technicians, and opens up avenues for advancements in iPSC research and broader applications.

7.
Front Bioeng Biotechnol ; 12: 1380950, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38846805

RESUMEN

As caretakers of the hematopoietic system, hematopoietic stem cells assure a lifelong supply of differentiated populations that are responsible for critical bodily functions, including oxygen transport, immunological protection and coagulation. Due to the far-reaching influence of the hematopoietic system, hematological disorders typically have a significant impact on the lives of individuals, even becoming fatal. Hematopoietic cell transplantation was the first effective therapeutic avenue to treat such hematological diseases. Since then, key use and manipulation of hematopoietic stem cells for treatments has been aspired to fully take advantage of such an important cell population. Limited knowledge on hematopoietic stem cell behavior has motivated in-depth research into their biology. Efforts were able to uncover their native environment and characteristics during development and adult stages. Several signaling pathways at a cellular level have been mapped, providing insight into their machinery. Important dynamics of hematopoietic stem cell maintenance were begun to be understood with improved comprehension of their metabolism and progressive aging. These advances have provided a solid platform for the development of innovative strategies for the manipulation of hematopoietic stem cells. Specifically, expansion of the hematopoietic stem cell pool has triggered immense interest, gaining momentum. A wide range of approaches have sprouted, leading to a variety of expansion systems, from simpler small molecule-based strategies to complex biomimetic scaffolds. The recent approval of Omisirge, the first expanded hematopoietic stem and progenitor cell product, whose expansion platform is one of the earliest, is predictive of further successes that might arise soon. In order to guarantee the quality of these ex vivo manipulated cells, robust assays that measure cell function or potency need to be developed. Whether targeting hematopoietic engraftment, immunological differentiation potential or malignancy clearance, hematopoietic stem cells and their derivatives need efficient scaling of their therapeutic potency. In this review, we comprehensively view hematopoietic stem cells as therapeutic assets, going from fundamental to translational.

8.
Front Cell Neurosci ; 16: 838217, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35308123

RESUMEN

The demand for large cell numbers for cellular therapies and drug screening applications requires the development of scalable platforms capable of generating high-quality populations of tissue-specific cells derived from human pluripotent stem cells (hPSCs). Here, we studied the ability of Gibco StemScale PSC Suspension Medium to promote the efficient expansion of hPSC cultures as aggregates grown in suspension. We tested human induced pluripotent stem cell (hiPSC) growth in 6-well plates (on orbital shaker platforms) and single-use vertical-wheel bioreactors for a total of three consecutive passages. Up to a 9-fold increase in cell number was observed over 5 days per passage, with a cumulative fold change up to 600 in 15 days. Additionally, we compared neural induction of hiPSCs by using a dual SMAD inhibition protocol with a commercially available neural induction medium, which can potentially yield more than a 30-fold change, including neural progenitor induction and expansion. This system can also be adapted toward the generation of floor plate progenitors, which yields up to an 80-fold change in cell number and generates FOXA2-positive populations. In summary, we developed platforms for hiPSC expansion and neural induction into different brain regions that provide scalability toward producing clinically relevant cell numbers.

9.
Front Cell Dev Biol ; 9: 648472, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33928083

RESUMEN

Human mesenchymal stromal cell (hMSC) therapy has been gaining immense interest in regenerative medicine and quite recently for its immunomodulatory properties in COVID-19 treatment. Currently, the use of hMSCs for various diseases is being investigated in >900 clinical trials. Despite the huge effort, setting up consistent and robust scalable manufacturing to meet regulatory compliance across various global regions remains a nagging challenge. This is in part due to a lack of definitive consensus for quality control checkpoint assays starting from cell isolation to expansion and final release criterion of clinical grade hMSCs. In this review, we highlight the bottlenecks associated with hMSC-based therapies and propose solutions for consistent GMP manufacturing of hMSCs starting from raw materials selection, closed and modular systems of manufacturing, characterization, functional testing, quality control, and safety testing for release criteria. We also discuss the standard regulatory compliances adopted by current clinical trials to broaden our view on the expectations across different jurisdictions worldwide.

10.
Front Immunol ; 12: 732135, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34925314

RESUMEN

Natural killer cells (NK cells) are the first line of the innate immune defense system, primarily located in peripheral circulation and lymphoid tissues. They kill virally infected and malignant cells through a balancing play of inhibitory and stimulatory receptors. In pre-clinical investigational studies, NK cells show promising anti-tumor effects and are used in adoptive transfer of activated and expanded cells, ex-vivo. NK cells express co-stimulatory molecules that are attractive targets for the immunotherapy of cancers. Recent clinical trials are investigating the use of CAR-NK for different cancers to determine the efficiency. Herein, we review NK cell therapy approaches (NK cell preparation from tissue sources, ways of expansion ex-vivo for "off-the-shelf" allogeneic cell-doses for therapies, and how different vector delivery systems are used to engineer NK cells with CARs) for cancer immunotherapy.


Asunto(s)
Células Alogénicas/inmunología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Inmunoterapia Adoptiva/métodos , Células Asesinas Naturales/inmunología , Neoplasias/terapia , Receptores Quiméricos de Antígenos/inmunología , Ingeniería Celular/métodos , Sangre Fetal/citología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Neoplasias/inmunología , Receptores Quiméricos de Antígenos/genética , Resultado del Tratamiento
11.
Blood ; 112(2): 295-307, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18332228

RESUMEN

We compared the transcriptomes of marrow-derived mesenchymal stem cells (MSCs) with differentiated adipocytes, osteocytes, and chondrocytes derived from these MSCs. Using global gene-expression profiling arrays to detect RNA transcripts, we have identified markers that are specific for MSCs and their differentiated progeny. Further, we have also identified pathways that MSCs use to differentiate into adipogenic, chondrogenic, and osteogenic lineages. We identified activin-mediated transforming growth factor (TGF)-beta signaling, platelet-derived growth factor (PDGF) signaling and fibroblast growth factor (FGF) signaling as the key pathways involved in MSC differentiation. The differentiation of MSCs into these lineages is affected when these pathways are perturbed by inhibitors of cell surface receptor function. Since growth and differentiation are tightly linked processes, we also examined the importance of these 3 pathways in MSC growth. These 3 pathways were necessary and sufficient for MSC growth. Inhibiting any of these pathways slowed MSC growth, whereas a combination of TGF-beta, PDGF, and beta-FGF was sufficient to grow MSCs in a serum-free medium up to 5 passages. Thus, this study illustrates it is possible to predict signaling pathways active in cellular differentiation and growth using microarray data and experimentally verify these predictions.


Asunto(s)
Perfilación de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/fisiología , Células Madre Mesenquimatosas/citología , Transducción de Señal , Adipocitos/citología , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Condrocitos/citología , Factores de Crecimiento de Fibroblastos/fisiología , Humanos , Osteoblastos/citología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Factor de Crecimiento Transformador beta/fisiología
12.
Methods Mol Biol ; 549: 3-16, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19378192

RESUMEN

Recent results have raised important questions on our ability to amplify stem cell populations in sufficient numbers as to be useful for therapy. Several reports have indicated that human stem cell populations harvested from the adult have low or undetectable telomerase levels, age in culture, and may not be propagated indefinitely. Other groups have shown that stem cells age and as such, their properties will have changed depending on the age of the individual from which they are harvested, and the time for which they are propagated in culture. Other groups have shown that cells maintained in culture may undergo alterations as they are propagated, and that these alterations may alter the predicted behavior of stem cells. Yet others have shown that human cells differ from their counterparts in other species in significant ways and have identified important difficulties in assessing cells in a xeno environment. Clinical colleagues have identified issues of variability and difficulties in the long-term follow-up that is being requested. Researchers in the stem cell field focused on translational work need to develop a practical plan that takes into account such difficulties while developing manufacturing protocols, designing animal studies, or developing trial protocols. Such proactive planning will be critical in ensuring a successful transition from the bench to the clinic.


Asunto(s)
Neuronas , Trasplante de Células Madre , Adulto , Animales , Diferenciación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Ensayos Clínicos Fase I como Asunto , Células Madre Embrionarias/fisiología , Humanos , Neuronas/citología , Neuronas/fisiología , Selección de Paciente , Roedores , Trasplante de Células Madre/métodos
13.
Cell Reprogram ; 21(5): 270-284, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31596624

RESUMEN

The embryonic stem cell line derivation from nonpermissive mouse strains is a challenging and highly inefficient process. The cellular reprogramming strategy provides an alternative route for generating pluripotent stem cell (PSC) lines from such strains. In this study, we successfully derived an enhanced green fluorescent protein (EGFP)-transgenic "N9" induced pluripotent stem cell (iPS cell, iPSC) line from the FVB/N strain-derived mouse embryonic fibroblasts (MEFs). The exposure of MEFs to human OCT4, SOX2, KLF4, and c-MYC (OSKM) transgenes via lentiviral transduction resulted in complete reprogramming. The N9 iPS cell line demonstrated all the criteria of a typical mouse PSC line, including normal colony morphology and karyotype (40,XY), high replication and propagation efficiencies, expression of the pluripotency-associated genes, spontaneous differentiation to three germ lineage-derived cell types, and robust potential of chimeric blastocyst formation. Taken together, using human OSKM genes for transduction, we report, for the first time, the successful derivation of an EGFP-expressing iPS cell line from a genetically nonpermissive transgenic FVB/N mouse. This cell line could provide opportunities for designing protocols for efficient derivation of PSC lines from other nonpermissive strains and developing mouse models of human diseases.


Asunto(s)
Embrión de Mamíferos/citología , Fibroblastos/citología , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Pluripotentes Inducidas/citología , Teratoma/patología , Animales , Linaje de la Célula , Células Cultivadas , Embrión de Mamíferos/metabolismo , Femenino , Fibroblastos/metabolismo , Proteínas Fluorescentes Verdes/genética , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Ratones , Ratones Transgénicos , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Teratoma/genética , Teratoma/metabolismo
14.
Methods Mol Biol ; 407: 149-63, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18453255

RESUMEN

Umbilical cord blood (UCB) is gaining more prominence in recent times as a source of non-embryonic multipotent stem cells. Global annual human birth rate (100 million) presents UCB as the largest non-controversial stem cell source, with an added advantage of naive immune status. Cord blood stem cells are routinely utilized in stem cell transplantation in leukemia patients and carry huge potential to treat other human diseases with less concern of rejection. Because UCB contains low number of stem cells, their use is associated with significant delays in engraftment of neutrophils and platelets. Development of reliable methods for isolation and expansion of cord blood stem cells is critical for consequent clinical application. The focus of this chapter is to review the methods currently used by different research groups and to recommend an isolation protocol that yields optimal number of UCB stem cells.


Asunto(s)
Separación Celular/métodos , Sangre Fetal/citología , Citometría de Flujo/métodos , Inmunofenotipificación/métodos , Células Madre/citología , Femenino , Sangre Fetal/fisiología , Humanos , Células Madre/fisiología
15.
Neurotoxicology ; 28(2): 396-401, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16678263

RESUMEN

The developing nervous system has long been recognized as a primary target site for lead (Pb)-induced toxicity. Pb-exposure causes cognitive dysfunction, growth retardation, hyperactivity and neurochemical deficits in animals and humans. In the present study the effects of 17-beta-estradiol on human SH-SY5Y neuroblastoma cells in culture exposed to low-levels of Pb were assessed. The cells were exposed to Pb (0.01-10 microM) for 48 h and cell proliferation was determined by the MTT reduction assay. Pb significantly inhibited the proliferation and growth of neuroblastoma cells in a concentration-dependent manner. A 50% inhibition (IC50) in the proliferation of cells was observed with 5 microM Pb. Exposure of cells to Pb (5 microM) for 48 h resulted in a significant increase (+732% of control) in caspase-3 activity, an indicator of apoptosis and total cellular prostaglandin E2 level (+1180% of control), marker of programmed cell death/neuronal cell loss. Pretreatment with 17-beta-estradiol (10 nM) effectively blocked the effects of Pb on caspase-3 activity but not prostaglandin E2 level. Further, Pb but not 17-beta-estradiol in a concentration (0.1-10 microM)-dependent manner effectively decreased (38-84%) the cellular concentration of glutathione (GSH), an important intracellular antioxidant. However, the effect of Pb on GSH level was effectively blocked when pretreated with 17-beta-estradiol. The data indicate that even low concentrations of Pb can be detrimental and potentially toxic to the developing brain. In conclusion, these results suggest that at least some of the neurotoxic effects of Pb may be mediated by apoptosis, which by pretreatment with 17-beta-estradiol can be prevented. This study further confirms previous reports of 17-beta-estradiol acting as a neuroprotective and antiapoptotic agent during induced toxic stress conditions.


Asunto(s)
Antioxidantes/metabolismo , Citoprotección , Estradiol/metabolismo , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/metabolismo , Compuestos Organometálicos/toxicidad , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dinoprostona/metabolismo , Relación Dosis-Respuesta a Droga , Estradiol/farmacología , Glutatión/metabolismo , Humanos , Concentración 50 Inhibidora , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Factores de Tiempo
16.
Methods Mol Biol ; 407: 1-10, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18453244

RESUMEN

Human embryonic stem cells (hESC) have the potential to treat a wide range of diseases. Currently, the use of existing hESC lines in human clinical applications is limited, as they are derived from blastocysts subjected to immunosurgery with animal derived antibodies, and are maintained on mouse embryonic feeder (MEF) cells, in the presence of either fetal calf serum (FCS) or on Matrigel or with conditioned media from MEFs. Successful derivation of hESCs in xeno-free conditions is crucial in advancing stem cell therapy applications. Two hESC lines, one from chromosomally abnormal embryos and another cell line from normal embryos from the inner cell mass of human blastocysts are derived using a culture media that had 20% serum replacement (SR) and human FGF2 on human foreskin fibroblasts as feeder cells. Derivation and characterization of such xenofree hESCs suitable for clinical studies is described in this chapter.


Asunto(s)
Células Madre Embrionarias/citología , Inmunohistoquímica/métodos , Hibridación Fluorescente in Situ/métodos , Blastocisto/citología , Blastocisto/metabolismo , Células Cultivadas , Células Madre Embrionarias/metabolismo , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Int J Mol Med ; 39(3): 587-594, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28204820

RESUMEN

Preclinical studies have suggested that paracrine factors from adipose-derived stem cells (ASCs) promote the healing of chronic wounds, and that the exposure of ASCs to hypoxia enhances their wound healing effect. To aid the translation of these findings into clinical use, robust wound models are necessary to explore each aspect of wound healing. The aspect of re-epithelization is often studied in a scratch assay based on transformed keratinocytes. However, there are concerns regarding the validity of this model, since these cell lines differ from normal keratinocytes, both in terms of proliferative capacity and differentiation, and sensitivity to environmental cues. In this study, the main challenge of using primary keratinocytes to examine the effects of ASCs was identified to be their different requirements for calcium in the culture media. We confirmed that a high calcium content led to morphological and cytoskeletal changes in primary keratinocytes, and demonstrated that a low calcium content compromised the growth of ASCs. We found that it is possible to perform the wound healing assay with primary keratinocytes, if the conditioned media from the ASCs is dialyzed to reduce the calcium concentration. Additionally, using this model of re-epithelization, conditioned media from normoxic ASCs was shown to markedly increase the rate of wound closure by primary keratinocytes, and this effect was significantly enhanced with media from the hypoxia-exposed ASCs. These findings, which are in line with the observations from previous in vivo studies, highlight the validity of this modified assay to investigate the wound healing properties of ASCs in vitro.


Asunto(s)
Tejido Adiposo/citología , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Diferenciación Celular , Hipoxia/metabolismo , Queratinocitos/metabolismo , Cicatrización de Heridas , Calcio , Técnicas de Cultivo de Célula , Células Cultivadas , Medios de Cultivo/química , Humanos
18.
ALTEX ; 34(1): 95-132, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27554434

RESUMEN

The first guidance on Good Cell Culture Practice (GCCP) dates back to 2005. This document expands this to include aspects of quality assurance for in vitro cell culture focusing on the increasingly diverse cell types and culture formats used in research, product development, testing and manufacture of biotechnology products and cell-based medicines. It provides a set of basic principles of best practice that can be used in training new personnel, reviewing and improving local procedures, and helping to assure standard practices and conditions for the comparison of data between laboratories and experimentation performed at different times. This includes recommendations for the documentation and reporting of culture conditions. It is intended as guidance to facilitate the generation of reliable data from cell culture systems, and is not intended to conflict with local or higher level legislation or regulatory requirements. It may not be possible to meet all recommendations in this guidance for practical, legal or other reasons. However, when it is necessary to divert from the principles of GCCP, the risk of decreasing the quality of work and the safety of laboratory staff should be addressed and any conclusions or alternative approaches justified. This workshop report is considered a first step toward a revised GCCP 2.0.


Asunto(s)
Alternativas a las Pruebas en Animales/normas , Técnicas de Cultivo de Célula/normas , Guías como Asunto/normas , Control de Calidad , Alternativas a las Pruebas en Animales/métodos , Animales , Técnicas de Cultivo de Célula/métodos , Congresos como Asunto , Humanos , Laboratorios/normas , Células Madre
19.
Cancer Lett ; 231(2): 262-9, 2006 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-16399227

RESUMEN

N-(4-hydroxyphenyl) retinamide (4-HPR, fenretinide) a synthetic retinoid is in clinical trials for the treatment of several malignancies. However, its biological effects and therapeutic value in childhood brain tumor medulloblastoma (MB) has not been investigated. In this study, we report for the first time that fenretinide (2.5-10 microM) induces apoptotic cell death in human MB cells. We observed significant inhibition of cell survival in four MB cell lines (D425MED, D458MED, D283MED and D341MED) as determined by MTT assays. These results were further supported by inhibition of anchorage-independent colony formation in soft agar. Fenretinide-induced decrease in cell viability was in part due to activation of caspase-3 dependent cell death, which was further supported by the cleavage of poly(ADP-ribose) polymerase-1 (PARP-1), a caspase-3 substrate. Cell death was partially prevented by the antioxidant, l-ascorbic acid suggesting that free radical intermediates might be involved in fenretinide effects. These results suggest that pharmacologically achievable concentrations of fenretinide are effective in killing MB cells and thus show its therapeutic potential to treat human MB.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Cerebelosas/tratamiento farmacológico , Fenretinida/uso terapéutico , Meduloblastoma/tratamiento farmacológico , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Ácido Ascórbico/farmacología , Caspasa 3 , Caspasas/metabolismo , Proliferación Celular/efectos de los fármacos , Neoplasias Cerebelosas/metabolismo , Neoplasias Cerebelosas/patología , Activación Enzimática/efectos de los fármacos , Humanos , Meduloblastoma/metabolismo , Meduloblastoma/patología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Células Tumorales Cultivadas
20.
J Immunol Res ; 2016: 5474602, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27298832

RESUMEN

The development of novel targeted therapies with acceptable safety profiles is critical to successful cancer outcomes with better survival rates. Immunotherapy offers promising opportunities with the potential to induce sustained remissions in patients with refractory disease. Recent dramatic clinical responses in trials with gene modified T cells expressing chimeric antigen receptors (CARs) in B-cell malignancies have generated great enthusiasm. This therapy might pave the way for a potential paradigm shift in the way we treat refractory or relapsed cancers. CARs are genetically engineered receptors that combine the specific binding domains from a tumor targeting antibody with T cell signaling domains to allow specifically targeted antibody redirected T cell activation. Despite current successes in hematological cancers, we are only in the beginning of exploring the powerful potential of CAR redirected T cells in the control and elimination of resistant, metastatic, or recurrent nonhematological cancers. This review discusses the application of the CAR T cell therapy, its challenges, and strategies for successful clinical and commercial translation.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Inmunoterapia/métodos , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T , Linfocitos T/inmunología , Linfocitos B/inmunología , Ensayos Clínicos como Asunto , Ingeniería Genética , Neoplasias Hematológicas/terapia , Humanos , Activación de Linfocitos , Recurrencia Local de Neoplasia/terapia , Receptores de Antígenos de Linfocitos T/genética , Proteínas Recombinantes de Fusión/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA