Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Antioxidants (Basel) ; 8(6)2019 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-31234608

RESUMEN

Green tea and its catechins have been shown to ameliorate high fat diet-induced oxidative stress and hyperlipidemia. However, low bioavailability of catechins limits their therapeutic potential. Lemon juice (LJ) has been suggested to enhance the bioavailability of catechins in vitro. This study investigated the antioxidative and hypolipidemic efficacy of a single dose of green tea extract (GTE) or GTE plus LJ (GTE + LJ) in high-fat diet fed pigs. Sixteen pigs ingested a single dose of GTE (190 mg/kg/day) or GTE + LJ (0.75 mL/kg/day) mixed with low-fat (LF; 5% fat) or high-fat (HF; 22% fat) diets and blood samples were collected for 24 h. Plasma catechin level peaked at two hours, and gradually returned to baseline after six hours following the intake. The addition of LJ significantly increased plasma catechin level. The diet containing GTE did not lower plasma cholesterol and triacylglycerol (TG) concentrations, superoxide dismutase (SOD) and catalase activity, or malondialdehyde concentration in 24 h in HF-fed pigs. Addition of a single dose of LJ, however, significantly decreased plasma TG level in LF groups but did not cause further changes on any other markers compared to the GTE alone. Our findings indicate limited effect of a single meal containing GTE on plasma antioxidant enzymes, lipid profile, and lipid peroxidation in pigs and no significant synergistic/additive action of adding LJ to GTE within 24 h in pigs. A study with a longer treatment period is warranted to further understand the potential role of GTE in reducing HF diet-induced oxidative stress and the possible synergistic role of LJ.

3.
Lab Anim ; 52(5): 490-496, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29558859

RESUMEN

The purpose of this study is to evaluate the accuracy of an oscillometric blood pressure monitor in anesthetized pigs. Invasive blood pressure (IBP) and noninvasive blood pressure (NIBP) measurements were taken using a DRE Waveline Pro multiparameter monitor at four different time points in 17 pigs undergoing injectable anesthesia. NIBP measurements were taken on both the thoracic and pelvic limbs. Bland Altman analysis was used to assess agreement between methods and a linear mixed-effects model was used to evaluate the effect of cuff position and blood pressure on bias. Invasive systolic arterial pressure (SAP) ranged between 112 and 161 mmHg (mean ± SD: 138.8 ± 13.3; median: 139.5). Invasive diastolic arterial pressure (DAP) ranged between 60 and 104 mmHg (mean ± SD: 86.0 ± 9.1; median: 87.0). Invasive mean arterial pressure (MAP) ranged between 79 and 121 mmHg (mean ± SD: 103.2 ± 9.3; median 103.0). Only the diastolic and mean measurements obtained from the pelvic limb met criteria outlined by the American College of Internal Medicine for required accuracy of NIBP monitors. Bias was significantly higher in the thoracic limb in comparison to the pelvic limb and was significantly higher at blood pressures above median. In general, NIBP measurements underestimated IBP measurements. In conclusion, the use of the DRE Waveline Pro to assess NIBP in anesthetized pigs may be useful in monitoring trends in mean and diastolic blood pressure and is most accurate when used on the pelvic limb.


Asunto(s)
Determinación de la Presión Sanguínea/métodos , Monitores de Presión Sanguínea , Cateterismo/métodos , Oscilometría/métodos , Sus scrofa/fisiología , Anestesia/estadística & datos numéricos , Animales , Determinación de la Presión Sanguínea/instrumentación , Femenino , Masculino , Porcinos
4.
Stem Cells Int ; 2016: 8610964, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27746821

RESUMEN

Ultrasound-guided intralesional injection of mesenchymal stem cells (MSCs) is held as the benchmark for cell delivery in tendonitis. The primary objective of this study was to investigate the immediate cell distribution following intralesional injection of MSCs. Unilateral superficial digital flexor tendon (SDFT) lesions were created in the forelimb of six horses and injected with 10 × 106 MSCs labeled with superparamagnetic iron oxide nanoparticles (SPIOs) under ultrasound guidance. Assays were performed to confirm that there were no significant changes in cell viability, proliferation, migration, or trilineage differentiation due to the presence of SPIOs. Limbs were imaged on a 1.5-tesla clinical MRI scanner postmortem before and after injection to determine the extent of tendonitis and detect SPIO MSCs. Clusters of labeled cells were visible as signal voids in 6/6 subjects. Coalescing regions of signal void were diffusely present in the peritendinous tissues. Although previous reports have determined that local injury retains cells within a small radius of the site of injection, our study shows greater than expected delocalization and relatively few cells retained within collagenous tendon compared to surrounding fascia. Further work is needed if this is a reality in vivo and to determine if directed intralesional delivery of MSCs is as critical as presently thought.

5.
Res Vet Sci ; 108: 18-24, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27663365

RESUMEN

OBJECTIVE: To evaluate the effects of allogeneic mesenchymal stem cells (MSCs) in a model of ischemic acute kidney injury (AKI). STUDY DESIGN: Randomized controlled trial. ANIMALS: Adult, purpose-bred research cats (n=15) and a historical reference group (n=3). METHODS: Cats underwent unilateral, in vivo, warm renal ischemia, then intravenous administration of 4 million adipose-derived MSCs, bone marrow-derived MSCs, or fibroblasts (n=5/treatment) 1h after reperfusion. Serum creatinine and blood urea nitrogen concentrations were measured at baseline and days 1 and 6. Urine specific gravity, urine protein to urine creatinine ratio, and glomerular filtration rate were measured at baseline and day 6. Both kidneys were harvested on day 6; histopathology was described and scored and smooth muscle actin was quantified with histomorphometry. A 2-way ANOVA was used to compare time and treatment. Chi square analysis was used to determine the % of cats with at least International Renal Interest Society (IRIS) Grade 1 AKI. RESULTS: Time, but not treatment, had a significant effect on renal function. No difference was noted in % of cats with IRIS AKI. Significantly fewer mitotic figures were observed in ischemic kidneys that received bone-marrow derived MSCs vs. fibroblasts. No differences in smooth muscle actin staining were noted. CONCLUSIONS: This study did not support the use of allogeneic MSCs in AKI in the regimen described here. Type of renal injury, MSC dose, allogenicity, duration, and route or timing of administration could influence the efficacy MSCs.


Asunto(s)
Lesión Renal Aguda/cirugía , Trasplante de Células Madre Mesenquimatosas , Tejido Adiposo/citología , Administración Intravenosa , Animales , Células de la Médula Ósea/citología , Gatos , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Trasplante Homólogo
6.
Res Vet Sci ; 103: 60-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26679797

RESUMEN

Feline bone marrow-derived MSCs (BMMSCs), adipose-derived MSCs (AMSCs) and fibroblasts (FBs) were isolated and cultured. Tri-lineage differentiation assays and flow cytometry were used to characterize MSCs. Neutrophils (NPs) were isolated from whole blood and the NPs production of reactive oxygen reactive oxygen species (ROS) was measured. NPs were cultured alone, with MSC culture supernatant (SN), BMMSCs or AMSCs. NPs incubated with BMMSCs had significantly lower ROS production than NPs incubated with AMSCs (p=0.0006) or FB (p<0.0001); NPs ROS production significantly decreased with increasing BMMSC cell number (p=0.0023) and significantly increased with NPs were incubated with FB compared to BMMSC (p=0.0003). Both BMMSC SN and AMSC SN had statistically significantly lower ROS production than FB SN when incubated with NPs (both p<0.0001). ROS production was significantly reduced with increased fractions of SN from BMMSCs (p=0.0467) and AMSCs (p=0.0017).


Asunto(s)
Gatos , Fibroblastos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Neutrófilos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Células de la Médula Ósea , Línea Celular
7.
Contrast Media Mol Imaging ; 10(5): 388-97, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26033748

RESUMEN

The goal of this study was to establish an SPIO-based cell-tracking method in an ovine model of tendonitis and to determine if this method may be useful for further study of cellular therapies in tendonitis in vivo. Functional assays were performed on labeled and unlabeled cells to ensure that no significant changes were induced by intracellular SPIOs. Following biosafety validation, tendon lesions were mechanically (n = 4) or chemically (n = 4) induced in four sheep and scanned ex vivo at 7 and 14 days to determine the presence and distribution of intralesional cells. Ovine MSCs labeled with 50 µg SPIOs/mL remained viable, proliferate, and undergo tri-lineage differentiation (p < 0.05). Labeled ovine MSCs remained detectable in vitro in concentrated cell numbers as low as 10 000 and in volumetric distributions as low as 100 000 cells/mL. Cells remained detectable by MRI at 7 days, as confirmed by correlative histology for dually labeled SPIO+/GFP+ cells. Histological evidence at 14 days suggested that SPIO particles remained embedded in tissue, providing MRI signal, although cells were no longer present. SPIO labeling has proven to be an effective method for cell tracking for a large animal model of tendon injury for up to 7 days post-injection. The data obtained in this study justify further investigation into the effects of MSC survival and migration on overall tendon healing and tissue regeneration.


Asunto(s)
Compuestos Férricos/química , Células Madre Mesenquimatosas/citología , Nanopartículas del Metal/química , Traumatismos de los Tendones/patología , Animales , Medios de Contraste/química , Modelos Animales de Enfermedad , Ovinos
8.
Cell Reprogram ; 16(5): 366-78, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25084370

RESUMEN

Chickens are vitally important in numerous countries as a primary food source and a major component of economic development. Efforts have been made to produce transgenic birds through pluripotent stem cell [primordial germ cells and embryonic stem cells (ESCs)] approaches to create animals with improved traits, such as meat and egg production or even disease resistance. However, these cell types have significant limitations because they are hard to culture long term while maintaining developmental plasticity. Induced pluripotent stem cells (iPSCs) are a novel class of stem cells that have proven to be robust, leading to the successful development of transgenic mice, rats, quail, and pigs and may potentially overcome the limitations of previous pluripotent stem cell systems in chickens. In this study we generated chicken (c) iPSCs from fibroblast cells for the first time using a nonviral minicircle reprogramming approach. ciPSCs demonstrated stem cell morphology and expressed key stem cell markers, including alkaline phosphatase, POU5F1, SOX2, NANOG, and SSEA-1. These cells were capable of rapid growth and expressed high levels of telomerase. Late-passage ciPSCs transplanted into stage X embryos were successfully incorporated into tissues of all three germ layers, and the gonads demonstrated significant cellular plasticity. These cells provide an exciting new tool to create transgenic chickens with broad implications for agricultural and transgenic animal fields at large.


Asunto(s)
Quimera , Células Madre Pluripotentes Inducidas/citología , Animales , Diferenciación Celular , Proliferación Celular , Embrión de Pollo , Pollos , Cartilla de ADN , Citometría de Flujo , Células Madre Pluripotentes Inducidas/enzimología , Reacción en Cadena de la Polimerasa , Telomerasa/metabolismo
9.
Cell Reprogram ; 15(2): 107-16, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23402576

RESUMEN

The shortage of human organs and tissues for transplant has led to significant interest in xenotransplantation of pig tissues for human patients. However, transplantation of pig organs results in an acute immune rejection, leading to death of the organ within minutes. The α-1,3-galactosyltransferase (GALT) gene has been knocked out in pigs to reduce rejection, yet additional genes need to be modified to ultimately make pig tissue immunocompatible with humans. The development of pig induced pluripotent stem cells (piPSCs) from GALT knockout (GALT-KO) tissue would provide an excellent cell source for complex genetic manipulations (e.g., gene targeting) that often require highly robust and proliferative cells. In this report, we generated GALT-KO piPSCs by the overexpression of POU5F1, SOX2, NANOG, LIN28, KLF-4, and C-MYC reprogramming genes. piPSCs showed classical stem cell morphology and characteristics, expressing integrated reprogramming genes in addition to the pluripotent markers AP, SSEA1, and SSEA4. GALT-KO piPSCs were highly proliferative and possessed doubling times and telomerase activity similar to human embryonic stem cells. These results demonstrated successful reprogramming of GALT-KO fibroblasts into GALT-KO piPSCs. GALT-KO piPSCs are potentially an excellent immortal cell source for the generation of pigs with complex genetic modifications for xenotransplantation, somatic cell nuclear transfer, or chimera formation.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Galactosiltransferasas , Expresión Génica , Técnicas de Silenciamiento del Gen , Células Madre Pluripotentes Inducidas/metabolismo , Factores de Transcripción/biosíntesis , Animales , Antígenos de Diferenciación/genética , Células Cultivadas , Humanos , Células Madre Pluripotentes Inducidas/citología , Trasplante de Células Madre , Porcinos , Factores de Transcripción/genética , Trasplante Heterólogo
10.
Cell Transplant ; 22(6): 945-59, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23043799

RESUMEN

Neural cells derived from induced pluripotent stem cells (iPSCs) have the potential for autologous cell therapies in treating patients with severe neurological disorders or injury. However, further study of efficacy and safety are needed in large animal preclinical models that have similar neural anatomy and physiology to humans such as the pig. The pig model for pluripotent stem cell therapy has been made possible for the first time with the development of pig iPSCs (piPSCs) capable of in vitro and in vivo differentiation into tissues of all three germ layers. Still, the question remains if piPSCs are capable of undergoing robust neural differentiation using a system similar to those being used with human iPSCs. In this study, we generated a new line of piPSCs from fibroblast cells that expressed pluripotency markers and were capable of embryoid body differentiation into all three germ layers. piPSCs demonstrated robust neural differentiation forming ßIII-TUB/MAP2+ neurons, GFAP+ astrocytes, and O4+ oligodendrocytes and demonstrated strong upregulation of neural cell genes representative of all three major neural lineages of the central nervous system. In the presence of motor neuron signaling factors, piPSC-derived neurons showed expression of transcription factors associated with motor neuron differentiation (HB9 and ISLET1). Our findings demonstrate that SSEA4 expression is required for piPSCs to differentiate into neurons, astrocytes, and oligodendrocytes and furthermore develop specific neuronal subtypes. This indicates that the pigs can fill the need for a powerful model to study autologous neural iPSC therapies in a system similar to humans.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/citología , Antígenos Embrionarios Específico de Estadio/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Biomarcadores/metabolismo , Forma de la Célula , Reprogramación Celular/genética , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Humanos , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Neuronas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Oligodendroglía/citología , Oligodendroglía/metabolismo , Sus scrofa , Transducción Genética , Tubulina (Proteína)/metabolismo
11.
Int J Biomater ; 2012: 861794, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22500171

RESUMEN

Autologous bone grafting is the most effective treatment for long-bone nonunions, but it poses considerable risks to donors, necessitating the development of alternative therapeutics. Poly(ethylene glycol) (PEG) microencapsulation and BMP2 transgene delivery are being developed together to induce rapid bone formation. However, methods to make these treatments available for clinical applications are presently lacking. In this study we used mesenchymal stem cells (MSCs) due to their ease of harvest, replication potential, and immunomodulatory capabilities. MSCs were from sheep and pig due to their appeal as large animal models for bone nonunion. We demonstrated that cryopreservation of these microencapsulated MSCs did not affect their cell viability, adenoviral BMP2 production, or ability to initiate bone formation. Additionally, microspheres showed no appreciable damage from cryopreservation when examined with light and electron microscopy. These results validate the use of cryopreservation in preserving the viability and functionality of PEG-encapsulated BMP2-transduced MSCs.

12.
Stem Cells Dev ; 21(3): 394-403, 2012 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-21970437

RESUMEN

Avian species are important model animals for developmental biology and disease research. However, unlike in mice, where clonal lines of pluripotent stem cells have enabled researchers to study mammalian gene function, clonal and highly proliferative pluripotent avian cell lines have been an elusive goal. Here we demonstrate the generation of avian induced pluripotent stem cells (iPSCs), the first nonmammalian iPSCs, which were clonally isolated and propagated, important attributes not attained in embryo-sourced avian cells. This was accomplished using human pluripotency genes rather than avian genes, indicating that the process in which mammalian and nonmammalian cells are reprogrammed is a conserved process. Quail iPSCs (qiPSCs) were capable of forming all 3 germ layers in vitro and were directly differentiated in culture into astrocytes, oligodendrocytes, and neurons. Ultimately, qiPSCs were capable of generating live chimeric birds and incorporated into tissues from all 3 germ layers, extraembryonic tissues, and potentially the germline. These chimera competent qiPSCs and in vitro differentiated cells offer insight into the conserved nature of reprogramming and genetic tools that were only previously available in mammals.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Reprogramación Celular , Células Madre Pluripotentes Inducidas/citología , Codorniz/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Diferenciación Celular , Proliferación Celular , Embrión de Pollo , Quimera , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Desarrollo Embrionario , Fibroblastos/citología , Fibroblastos/metabolismo , Genoma Humano , Estratos Germinativos/citología , Estratos Germinativos/metabolismo , Humanos , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Neuronas/citología , Neuronas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Oligodendroglía/citología , Oligodendroglía/metabolismo , Codorniz/genética , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Telomerasa/metabolismo , Transducción Genética
13.
Pharmacol Ther ; 129(3): 290-306, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21073897

RESUMEN

Stem cell-based therapeutics have the potential to effectively treat many terminal and debilitating human diseases, but the mechanisms by which their growth and differentiation are regulated are incompletely defined. Recent data from multiple systems suggest major roles for G protein coupled receptor (GPCR) pathways in regulating stem cell function in vivo and in vitro. The goal of this review is to illustrate common ground between the growing field of stem cell therapeutics and the long-established field of G protein coupled receptor signaling. Herein, we briefly introduce basic stem cell biology and discuss how several conserved pathways regulate pluripotency and differentiation in mouse and human stem cells. We further discuss general mechanisms by which GPCR signaling may impact these pluripotency and differentiation pathways, and summarize specific examples of receptors from each of the major GPCR subfamilies that have been shown to regulate stem cell function. Finally, we discuss possible therapeutic implications of GPCR regulation of stem cell function.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Pluripotentes/fisiología , Receptores Acoplados a Proteínas G/fisiología , Animales , Humanos , Células Madre Pluripotentes/citología , Receptores Acoplados a Proteínas G/metabolismo
14.
Stem Cells Dev ; 20(6): 1079-88, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20929355

RESUMEN

Early germ-like cells (GLCs) derived from human embryonic stem cells (hESCs) have presented new opportunities to study germ cell differentiation in vitro. However, differentiation conditions that facilitate the formation of haploid cells from the derived GLCs have eluded the field. The inability to propagate GLCs in culture is a further limitation, resulting in inconsistent rederivations of GLCs from hESCs with relatively few GLCs in these heterogeneous populations. Here we found in vitro conditions that enrich for DDX4/POU5F1+ GLCs (∼60%) and that has enabled continual propagation for >50 passages without loss of phenotype. Clonal isolation of single GLCs from these mixed cultures generated 3 GLC (>90% DDX4/POU5F1+) and 2 hESC (<0.1% DDX4+) lines that could be continually expanded without loss of phenotype. Differentiation of clonal GLC lines in serum resulted in expression of postmeiotic markers and >11% were haploid, ∼5-fold higher than previous studies. The robust clonal meiotic competent and incompetent GLC lines will be used to understand the factors controlling human germ cell meiosis and postmeiotic maturation.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Células Germinativas/citología , Haploidia , Meiosis , Animales , Biomarcadores/metabolismo , Diferenciación Celular/genética , Movimiento Celular/genética , Proliferación Celular , Separación Celular , Células Cultivadas , Células Clonales , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica , Humanos , Meiosis/genética , Ratones , Fenotipo
15.
Stem Cells Dev ; 19(8): 1211-20, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20380514

RESUMEN

Ethical and moral issues rule out the use of human induced pluripotent stem cells (iPSCs) in chimera studies that would determine the full extent of their reprogrammed state, instead relying on less rigorous assays such as teratoma formation and differentiated cell types. To date, only mouse iPSC lines are known to be truly pluripotent. However, initial mouse iPSC lines failed to form chimeric offspring, but did generate teratomas and differentiated embryoid bodies, and thus these specific iPSC lines were not completely reprogrammed or truly pluripotent. Therefore, there is a need to address whether the reprogramming factors and process used eventually to generate chimeric mice are universal and sufficient to generate reprogrammed iPSC that contribute to chimeric offspring in additional species. Here we show that porcine mesenchymal stem cells transduced with 6 human reprogramming factors (POU5F1, SOX2, NANOG, KLF4, LIN28, and C-MYC) injected into preimplantation-stage embryos contributed to multiple tissue types spanning all 3 germ layers in 8 of 10 fetuses. The chimerism rate was high, 85.3% or 29 of 34 live offspring were chimeras based on skin and tail biopsies harvested from 2- to 5-day-old pigs. The creation of pluripotent porcine iPSCs capable of generating chimeric offspring introduces numerous opportunities to study the facets significantly affecting cell therapies, genetic engineering, and other aspects of stem cell and developmental biology.


Asunto(s)
Quimera/embriología , Células Madre Pluripotentes Inducidas/citología , Sus scrofa , Estructuras Animales/citología , Estructuras Animales/metabolismo , Animales , Animales Recién Nacidos/anomalías , Animales Recién Nacidos/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Blastocisto/citología , Diferenciación Celular/genética , Quimera/anomalías , Quimera/metabolismo , Cuerpos Embrioides/citología , Proteínas Fetales/genética , Feto/citología , Feto/metabolismo , Expresión Génica/genética , Proteínas de Homeodominio/genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Células Madre Mesenquimatosas/citología , Proteína Homeótica Nanog , Proteínas del Tejido Nervioso/genética , 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 de Unión al ARN/genética , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Proteínas de Dominio T Box/genética , Transducción Genética , alfa-Fetoproteínas/genética
16.
Curr Stem Cell Res Ther ; 5(3): 251-60, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20214556

RESUMEN

Availability of human embryonic stem cells (hESCs) and its neural derivatives has opened up wide possibilities of using these cells as tools for developmental studies, drug screening and cell therapies for treating neurodegenerative diseases. However, for hESC-derived neurons to fulfill their potential they need to form functional synapses and spontaneously active neural networks. Until recently very few studies have reported hESC-derived neurons capable of forming such networks, suggesting lack of certain components in culture media to promote mature synaptogenesis. In this review we discuss the various factors that enhance functional synapse formation in primary and stem cell-derived neuronal cultures. These factors include astrocytes, astrocyte-derived factors, cell adhesion molecules and neurotrophins. We discuss the current literature on studies that have used these factors for functional differentiation of primary neural cultures, and discuss its implications for stem cell -derived neural cultures.


Asunto(s)
Astrocitos/fisiología , Moléculas de Adhesión Celular/metabolismo , Células Madre Embrionarias/fisiología , Factores de Crecimiento Nervioso/metabolismo , Neurogénesis/fisiología , Neuronas , Sinapsis/fisiología , Astrocitos/citología , Diferenciación Celular/fisiología , Células Cultivadas , Células Madre Embrionarias/citología , Ácido Glutámico/metabolismo , Humanos , Red Nerviosa/fisiología , Neuronas/fisiología , Neuronas/ultraestructura , Terminales Presinápticos/fisiología , Terminales Presinápticos/ultraestructura , Sinapsis/ultraestructura , Trombospondinas/metabolismo
17.
Microsc Microanal ; 16(1): 80-90, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20082731

RESUMEN

Neurodegerative disorders affect millions of people worldwide. Neural cells derived from human embryonic stem cells (hESC) have the potential for cell therapies and/or compound screening for treating affected individuals. While both protein and gene expression indicative of a neural phenotype has been exhibited in these differentiated cells, ultrastuctural studies thus far have been lacking. The objective of this study was to correlate hESC to neural differentiation culture conditions with ultrastructural changes observed in the treated cells. We demonstrate here that in basic culture conditions without growth factors or serum we obtain neural morphology. The addition of brain-derived neurotrophic factor (BDNF) and serum to cultures resulted in more robust neural differentiation. In addition to providing cues such as cell survival or lineage specification, additional factors also altered the intracellular structures and cell morphologies. Even though the addition of BDNF and serum did not increase synaptic formation, altered cellular structures such as abundant polyribosomes and more developed endoplasmic reticulum indicate a potential increase in protein production.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/fisiología , Células Madre Embrionarias/ultraestructura , Neuronas/fisiología , Neuronas/ultraestructura , Factor Neurotrófico Derivado del Encéfalo/farmacología , Células Cultivadas , Medios de Cultivo/química , Sustancias de Crecimiento/farmacología , Humanos
18.
BMC Neurosci ; 9: 118, 2008 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-19077254

RESUMEN

BACKGROUND: Lysophospholipids regulate the morphology and growth of neurons, neural cell lines, and neural progenitors. A stable human neural progenitor cell line is not currently available in which to study the role of lysophospholipids in human neural development. We recently established a stable, adherent human embryonic stem cell-derived neuroepithelial (hES-NEP) cell line which recapitulates morphological and phenotypic features of neural progenitor cells isolated from fetal tissue. The goal of this study was to determine if hES-NEP cells express functional lysophospholipid receptors, and if activation of these receptors mediates cellular responses critical for neural development. RESULTS: Our results demonstrate that Lysophosphatidic Acid (LPA) and Sphingosine-1-phosphate (S1P) receptors are functionally expressed in hES-NEP cells and are coupled to multiple cellular signaling pathways. We have shown that transcript levels for S1P1 receptor increased significantly in the transition from embryonic stem cell to hES-NEP. hES-NEP cells express LPA and S1P receptors coupled to G i/o G-proteins that inhibit adenylyl cyclase and to G q-like phospholipase C activity. LPA and S1P also induce p44/42 ERK MAP kinase phosphorylation in these cells and stimulate cell proliferation via G i/o coupled receptors in an Epidermal Growth Factor Receptor (EGFR)- and ERK-dependent pathway. In contrast, LPA and S1P stimulate transient cell rounding and aggregation that is independent of EGFR and ERK, but dependent on the Rho effector p160 ROCK. CONCLUSION: Thus, lysophospholipids regulate neural progenitor growth and morphology through distinct mechanisms. These findings establish human ES cell-derived NEP cells as a model system for studying the role of lysophospholipids in neural progenitors.


Asunto(s)
Línea Celular , Células Madre Embrionarias/metabolismo , Células Neuroepiteliales/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Proliferación Celular , Células Madre Embrionarias/citología , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas de Unión al GTP/metabolismo , Humanos , Microscopía por Video , Células Neuroepiteliales/citología , Células Neuroepiteliales/fisiología , ARN Mensajero/metabolismo , Receptores del Ácido Lisofosfatídico/genética , Receptores del Ácido Lisofosfatídico/fisiología , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología , Quinasas Asociadas a rho/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA