Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
Int J Radiat Biol ; 97(9): 1316-1323, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34047676

RESUMEN

PURPOSE: To assess the effects of 1800 MHz radiofrequency electromagnetic field (RF-EMF) exposure on the expression of signal transduction and antioxidant proteins in a human-derived A172 glioblastoma cell line. MATERIALS AND METHODS: Adherent human-derived A172 glioblastoma cells (1.0 × 105 cells per 35 mm culture dish, containing 2 mL DMEM media) were exposed to 1800 MHz continuous-wave (CW) or GSM-modulated RF fields, in the presence or absence of serum for 5, 30 or 240 min at a specific absorption rate (SAR) of 0 (sham) or 2.0 W/kg. Concurrent negative (vehicle) and positive controls (1 µg/mL anisomycin) were included in each experiment. Cell lysates were collected immediately after exposure, stabilized by protease and phosphatase inhibitors in lysis buffer, then frozen and maintained at -80 °C until analysis. The relative expression levels of phosphorylated- and total-signal transduction proteins (CREB, JNK, NF-κB, ERK1/2, Akt, p70S6K, STAT3 and STAT5) and antioxidant proteins (SOD1, SOD2, CAT, TRX1, PRX2) were assessed using Milliplex magnetic bead array panels and a MagPix Multiplex imaging system. RESULTS: In cells exposed to 1800 MHz continuous-wave RF-EMF with the presence of serum in the culture medium, CAT expression was statistically significantly decreased after a 30 min exposure, total JNK was decreased at both 30 and 240 min of exposure, STAT3 was decreased after 240 min of exposure and phosphorylated-CREB expression was decreased after 30 min of exposure. In cells exposed to 1800 MHz GSM-modulated RF-EMF in serum-free cultures, the expression level of total STAT5 was decreased after 30 and 240 min of exposure. These observed changes were detected sporadically across time-points, culture conditions and RF-EMF exposure conditions indicating the likelihood of false positive events. When cells were treated with anisomycin for 15 min as a positive control, dramatic increases in the expression of phosphorylated signaling proteins were observed in both serum-starved and serum-fed A172 cells, with larger fold change increases in the serum-free cultures. No statistically significant differences in the expression levels of SOD1, SOD2 or TRX1 were observed under any tested conditions after exposure to RF-EMF. CONCLUSIONS: The current study found no consistent evidence of changes in the expression of antioxidant proteins (SOD1, SOD2, CAT or TRX2) or a variety of signal transductions proteins (CREB, JNK, NF-κB, ERK1/2, Akt, p70S6K, STAT3, STAT5) in a human-derived glioblastoma A172 cell line in response to exposure to 1800 MHz continuous-wave or GSM-modulated RF-EMF for 5, 30 or 240 min in either serum-free or serum-containing cultures.


Asunto(s)
Antioxidantes/metabolismo , Glioblastoma/patología , Fosfoproteínas/metabolismo , Ondas de Radio , Transducción de Señal/efectos de la radiación , Línea Celular Tumoral , Humanos , Fosforilación/efectos de la radiación
2.
Sci Rep ; 10(1): 4650, 2020 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-32157159

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

3.
Basic Res Cardiol ; 115(3): 26, 2020 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-32146560

RESUMEN

Mesenchymal stromal cells (MSCs) exhibit antiapoptotic and proangiogenic functions in models of myocardial infarction which may be mediated by secreted small extracellular vesicles (sEVs). However, MSCs have frequently been harvested from aged or diseased patients, while the isolated sEVs often contain high levels of impurities. Here, we studied the cardioprotective and proangiogenic activities of size-exclusion chromatography-purified sEVs secreted from human foetal amniotic fluid stem cells (SS-hAFSCs), possessing superior functional potential to that of adult MSCs. We demonstrated for the first time that highly pure (up to 1.7 × 1010 particles/µg protein) and thoroughly characterised SS-hAFSC sEVs protect rat hearts from ischaemia-reperfusion injury in vivo when administered intravenously prior to reperfusion (38 ± 9% infarct size reduction, p < 0.05). SS-hAFSC sEVs did not protect isolated primary cardiomyocytes in models of simulated ischaemia-reperfusion injury in vitro, indicative of indirect cardioprotective effects. SS-hAFSC sEVs were not proangiogenic in vitro, although they markedly stimulated endothelial cell migration. Additionally, sEVs were entirely responsible for the promigratory effects of the medium conditioned by SS-hAFSC. Mechanistically, sEV-induced chemotaxis involved phosphatidylinositol 3-kinase (PI3K) signalling, as its pharmacological inhibition in treated endothelial cells reduced migration by 54 ± 7% (p < 0.001). Together, these data indicate that SS-hAFSC sEVs have multifactorial beneficial effects in a myocardial infarction setting.


Asunto(s)
Líquido Amniótico/citología , Cardiotónicos/metabolismo , Movimiento Celular , Vesículas Extracelulares/metabolismo , Células Madre Mesenquimatosas/metabolismo , Daño por Reperfusión/metabolismo , Animales , Quimiotaxis , Humanos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fosfatidilinositol 3-Quinasa/metabolismo , Ratas , Daño por Reperfusión/patología
4.
Sci Rep ; 9(1): 8126, 2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-31148575

RESUMEN

Expression of OCT4A is one of the hallmarks of pluripotency, defined as a stem cell's ability to differentiate into all the lineages of the three germ layers. Despite being defined as non-tumorigenic cells with high translational potential, human mid-trimester amniotic fluid stem cells (hAFSCs) are often described as sharing features with embryonic stem cells, including the expression of OCT4A, which could hinder their clinical potential. To clarify the OCT4A status of hAFSCs, we first undertook a systematic review of the literature. We then performed extensive gene and protein expression analyses to discover that neither frozen, nor fresh hAFSCs cultivated in multipotent stem cell culture conditions expressed OCT4A, and that the OCT4A positive results from the literature are likely to be attributed to the expression of pseudogenes or other OCT4 variants. To address this issue, we provide a robust protocol for the assessment of OCT4A in other stem cells.


Asunto(s)
Líquido Amniótico/citología , Regulación del Desarrollo de la Expresión Génica , Factor 3 de Transcripción de Unión a Octámeros/genética , Células Madre/citología , Linaje de la Célula , Exones , Femenino , Perfilación de la Expresión Génica , Variación Genética , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Microscopía Fluorescente , Células Madre Multipotentes/citología , Embarazo , Segundo Trimestre del Embarazo , Isoformas de Proteínas
5.
Sci Data ; 5: 180100, 2018 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-29809174

RESUMEN

Micro-computed tomography (micro-CT) is commonly used to assess bone quality and to evaluate the outcome of experimental therapies in animal models of bone diseases. Generating large datasets is however challenging and data are rarely made publicly available through shared repositories. Here we describe a dataset of micro-CT reconstructed scans of the proximal part of 21 tibiae from wild-type mice, osteogenesis imperfecta mice (homozygous oim/oim) and oim/oim mice transplanted with human amniotic fluid stem cells. The dataset contains, for each sample, 991 8-bit Bitmap reconstructed images and a 3D reconstruction of the bone in the PLY format, available at the online repository Figshare. In line with the increasing effort to make scientific datasets open-access, our data can be downloaded and used by other researchers to compare their observations with ours and to directly test scientific questions on osteogenesis imperfecta bones without the need to generate complete datasets.


Asunto(s)
Osteogénesis Imperfecta , Trasplante de Células Madre , Animales , Humanos , Ratones , Osteogénesis Imperfecta/diagnóstico por imagen , Osteogénesis Imperfecta/patología , Osteogénesis Imperfecta/terapia , Células Madre/patología , Tibia/diagnóstico por imagen , Microtomografía por Rayos X
6.
Stem Cell Res Ther ; 9(1): 113, 2018 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-29695308

RESUMEN

BACKGROUND: Established therapies for managing kidney dysfunction such as kidney dialysis and transplantation are limited due to the shortage of compatible donated organs and high costs. Stem cell-based therapies are currently under investigation as an alternative treatment option. As amniotic fluid is composed of fetal urine harboring mesenchymal stem cells (AF-MSCs), we hypothesized that third-trimester amniotic fluid could be a novel source of renal progenitor and differentiated cells. METHODS: Human third-trimester amniotic fluid cells (AFCs) were isolated and cultured in distinct media. These cells were characterized as renal progenitor cells with respect to cell morphology, cell surface marker expression, transcriptome and differentiation into chondrocytes, osteoblasts and adipocytes. To test for renal function, a comparative albumin endocytosis assay was performed using AF-MSCs and commercially available renal cells derived from kidney biopsies. Comparative transcriptome analyses of first, second and third trimester-derived AF-MSCs were conducted to monitor expression of renal-related genes. RESULTS: Regardless of the media used, AFCs showed expression of pluripotency-associated markers such as SSEA4, TRA-1-60, TRA-1-81 and C-Kit. They also express the mesenchymal marker Vimentin. Immunophenotyping confirmed that third-trimester AFCs are bona fide MSCs. AF-MSCs expressed the master renal progenitor markers SIX2 and CITED1, in addition to typical renal proteins such as PODXL, LHX1, BRN1 and PAX8. Albumin endocytosis assays demonstrated the functionality of AF-MSCs as renal cells. Additionally, upregulated expression of BMP7 and downregulation of WT1, CD133, SIX2 and C-Kit were observed upon activation of WNT signaling by treatment with the GSK-3 inhibitor CHIR99201. Transcriptome analysis and semiquantitative PCR revealed increasing expression levels of renal-specific genes (e.g., SALL1, HNF4B, SIX2) with gestational time. Moreover, AF-MSCs shared more genes with human kidney cells than with native MSCs and gene ontology terms revealed involvement of biological processes associated with kidney morphogenesis. CONCLUSIONS: Third-trimester amniotic fluid contains AF-MSCs of renal origin and this novel source of kidney progenitors may have enormous future potentials for disease modeling, renal repair and drug screening.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Embarazo/fisiología , Células Madre/metabolismo , Líquido Amniótico , Diferenciación Celular , Femenino , Humanos
7.
Stem Cells Transl Med ; 7(5): 439-449, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29489062

RESUMEN

Human mesenchymal stem cells (MSCs) have huge potential for regenerative medicine. In particular, the use of pluripotent stem cell-derived mesenchymal stem cells (PSC-MSCs) overcomes the hurdle of replicative senescence associated with the in vitro expansion of primary cells and has increased therapeutic benefits in comparison to the use of various adult sources of MSCs in a wide range of animal disease models. On the other hand, fetal MSCs exhibit faster growth kinetics and possess longer telomeres and a wider differentiation potential than adult MSCs. Here, for the first time, we compare the therapeutic potential of PSC-MSCs (ES-MSCs from embryonic stem cells) to fetal MSCs (AF-MSCs from the amniotic fluid), demonstrating that ES-MSCs have a superior neuroprotective potential over AF-MSCs in the mouse brain following hypoxia-ischemia. Further, we demonstrate that nuclear factor (NF)-κB-stimulated interleukin (IL)-13 production contributes to an increased in vitro anti-inflammatory potential of ES-MSC-conditioned medium (CM) over AF-MSC-CM, thus suggesting a potential mechanism for this observation. Moreover, we show that induced pluripotent stem cell-derived MSCs (iMSCs) exhibit many similarities to ES-MSCs, including enhanced NF-κB signaling and IL-13 production in comparison to AF-MSCs. Future studies should assess whether iMSCs also exhibit similar neuroprotective potential to ES-MSCs, thus presenting a potential strategy to overcome the ethical issues associated with the use of embryonic stem cells and providing a potential source of cells for autologous use against neonatal hypoxic-ischemic encephalopathy in humans. Stem Cells Translational Medicine 2018;7:439-449.


Asunto(s)
Encéfalo/patología , Células Madre Embrionarias/citología , Células Madre Fetales/citología , Hipoxia/patología , Células Madre Mesenquimatosas/citología , Neuroprotección/fisiología , Líquido Amniótico/citología , Animales , Encéfalo/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Células Madre Embrionarias/metabolismo , Femenino , Células Madre Fetales/metabolismo , Células HEK293 , Humanos , Hipoxia/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Isquemia/metabolismo , Isquemia/patología , Masculino , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Medicina Regenerativa/métodos , Transducción de Señal/fisiología
8.
Sci Rep ; 8(1): 2425, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29402914

RESUMEN

Human amniotic fluid contains two morphologically-distinct sub-populations of stem cells with regenerative potential, spindle-shaped (SS-hAFSCs) and round-shaped human amniotic fluid stem cells (RS-hAFSCs). However, it is unclear whether morphological differences correlate with functionality, and this lack of knowledge limits their translational applications. Here, we show that SS-hAFSCs and RS-hAFSCs differ in their neuro-protective ability, demonstrating that a single contralateral injection of SS-hAFSCs into hypoxic-ischemic P7 mice conferred a 47% reduction in hippocampal tissue loss and 43-45% reduction in TUNEL-positive cells in the hippocampus and striatum 48 hours after the insult, decreased microglial activation and TGFß1 levels, and prevented demyelination. On the other hand, RS-hAFSCs failed to show such neuro-protective effects. It is possible that SS-hAFSCs exert their neuroprotection via endoglin-dependent inhibition of TGFß1 signaling in target cells. These findings identify a sub-population of CD117+CD90+CD105+ stem cells as a promising source for the neuro-protection of the developing brain.


Asunto(s)
Líquido Amniótico/citología , Isquemia Encefálica/terapia , Enfermedades Desmielinizantes/prevención & control , Hipoxia/prevención & control , Neuroprotección/fisiología , Trasplante de Células Madre , Células Madre/citología , Líquido Amniótico/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Apoptosis , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Linaje de la Célula , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Endoglina/genética , Endoglina/metabolismo , Expresión Génica , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Hipoxia/genética , Hipoxia/metabolismo , Hipoxia/patología , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Microglía/patología , Células Madre/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
9.
Sci Rep ; 7(1): 6601, 2017 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-28747757

RESUMEN

The human amniotic fluid stem cell (hAFSC) population consists of two morphologically distinct subtypes, spindle-shaped and round-shaped cells (SS-hAFSCs and RS-hAFSCs). Whilst SS-hAFSCs are routinely expanded in mesenchymal-type (MT) conditions, we previously showed that they acquire broader differentiation potential when cultured under embryonic-type (ET) conditions. However, the effects of culture conditions on RS-hAFSCs have not been determined. Here, we show that culturing RS-hAFSCs under ET conditions confers faster proliferation and enhances the efficiency of osteogenic differentiation of the cells. We show that this occurs via TGFß-induced activation of CD73 and the associated increase in the generation of extracellular adenosine. Our data demonstrate that culture conditions are decisive for the expansion of hAFSCs and that TGFß present in ET conditions causes the phenotype of RS-hAFSCs to revert to an earlier state of stemness. Cultivating RS-hAFSCs in ET conditions with TGFß may therefore increase their therapeutic potential for clinical applications.


Asunto(s)
5'-Nucleotidasa/metabolismo , Adenosina/metabolismo , Líquido Amniótico/citología , Osteogénesis , Células Madre/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteínas Ligadas a GPI/metabolismo , Humanos
10.
Mol Ther ; 25(2): 427-442, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28153093

RESUMEN

Restoring pluripotency using chemical compounds alone would be a major step forward in developing clinical-grade pluripotent stem cells, but this has not yet been reported in human cells. We previously demonstrated that VPA_AFS cells, human amniocytes cultivated with valproic acid (VPA) acquired functional pluripotency while remaining distinct from human embryonic stem cells (hESCs), questioning the relationship between the modulation of cell fate and molecular regulation of the pluripotency network. Here, we used single-cell analysis and functional assays to reveal that VPA treatment resulted in a homogeneous population of self-renewing non-transformed cells that fulfill the hallmarks of pluripotency, i.e., a short G1 phase, a dependence on glycolytic metabolism, expression of epigenetic modifications on histones 3 and 4, and reactivation of endogenous OCT4 and downstream targets at a lower level than that observed in hESCs. Mechanistic insights into the process of VPA-induced reprogramming revealed that it was dependent on OCT4 promoter activation, which was achieved independently of the PI3K (phosphatidylinositol 3-kinase)/AKT/mTOR (mammalian target of rapamycin) pathway or GSK3ß inhibition but was concomitant with the presence of acetylated histones H3K9 and H3K56, which promote pluripotency. Our data identify, for the first time, the pluripotent transcriptional and molecular signature and metabolic status of human chemically induced pluripotent stem cells.


Asunto(s)
Amnios/citología , Transdiferenciación Celular/efectos de los fármacos , Reprogramación Celular/efectos de los fármacos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Biomarcadores , Ciclo Celular/genética , Transdiferenciación Celular/genética , Reprogramación Celular/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Metabolismo Energético , Epigénesis Genética , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Genes Reporteros , Glucólisis , Histonas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Proteína Homeótica Nanog/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Recombinantes de Fusión , Serina-Treonina Quinasas TOR/metabolismo , Activación Transcripcional
11.
Sci Rep ; 6: 39656, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-27995994

RESUMEN

The impaired maturation of bone-forming osteoblasts results in reduced bone formation and subsequent bone weakening, which leads to a number of conditions such as osteogenesis imperfecta (OI). Transplantation of human fetal mesenchymal stem cells has been proposed as skeletal anabolic therapy to enhance bone formation, but the mechanisms underlying the contribution of the donor cells to bone health are poorly understood and require further elucidation. Here, we show that intraperitoneal injection of human amniotic mesenchymal stem cells (AFSCs) into a mouse model of OI (oim mice) reduced fracture susceptibility, increased bone strength, improved bone quality and micro-architecture, normalised bone remodelling and reduced TNFα and TGFß sigalling. Donor cells engrafted into bones and differentiated into osteoblasts but importantly, also promoted endogenous osteogenesis and the maturation of resident osteoblasts. Together, these findings identify AFSC transplantation as a countermeasure to bone fragility. These data have wider implications for bone health and fracture reduction.


Asunto(s)
Amnios/citología , Fracturas Óseas/prevención & control , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Osteogénesis Imperfecta/prevención & control , Animales , Remodelación Ósea , Huesos/metabolismo , Diferenciación Celular , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Marcadores Genéticos , Humanos , Masculino , Ratones , Osteoblastos/metabolismo , Osteogénesis , Estrés Mecánico , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Microtomografía por Rayos X
12.
Int J Radiat Biol ; 92(6): 338-50, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27028625

RESUMEN

PURPOSE: To assess 1.9 GHz radiofrequency (RF) field exposure on gene expression within a variety of discrete mouse brain regions using whole genome microarray analysis. MATERIALS AND METHODS: Adult male C57BL/6 mice were exposed to 1.9 GHz pulse-modulated or continuous-wave RF fields for 4 h/day for 5 consecutive days at whole body average (WBA) specific absorption rates of 0 (sham), ∼0.2 W/kg and ∼1.4 W/kg. Total RNA was isolated from the auditory cortex, amygdala, caudate, cerebellum, hippocampus, hypothalamus, and medial prefrontal cortex and differential gene expression was assessed using Illumina MouseWG-6 (v2) BeadChip arrays. Validation of potentially responding genes was conducted by RT-PCR. RESULTS: When analysis of gene expression was conducted within individual brain regions when controlling the false discovery rate (FDR), no differentially expressed genes were identified relative to the sham control. However, it must be noted that most fold changes among groups were observed to be less than 1.5-fold and this study had limited ability to detect such small changes. While some genes were differentially expressed without correction for multiple-comparisons testing, no consistent pattern of response was observed among different RF-exposure levels or among different RF-modulations. CONCLUSIONS: The current study provides the most comprehensive analysis of potential gene expression changes in the rodent brain in response to RF field exposure conducted to date. Within the exposure conditions and limitations of this study, no convincing evidence of consistent changes in gene expression was found in response to 1.9 GHz RF field exposure.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/efectos de la radiación , Regulación de la Expresión Génica/efectos de la radiación , Microondas , Proteínas del Tejido Nervioso/metabolismo , Irradiación Corporal Total/métodos , Absorción de Radiación/fisiología , Animales , Relación Dosis-Respuesta en la Radiación , Regulación de la Expresión Génica/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Dosis de Radiación , Distribución Tisular
13.
Stem Cells Dev ; 25(5): 395-404, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26728561

RESUMEN

Alport syndrome (AS) is a hereditary glomerulopathy caused by a mutation in type IV collagen genes, which disrupts glomerular basement membrane, leading to progressive glomerulosclerosis and end-stage renal failure. There is at present no cure for AS, and cell-based therapies offer promise to improve renal function. In this study, we found that human first trimester fetal chorionic stem cells (CSC) are able to migrate to glomeruli and differentiate down the podocyte lineage in vitro and in vivo. When transplanted into 7-week-old Alport 129Sv-Col4α3(tm1Dec)/J (-/-) mice, a single intraperitoneal injection of CSC significantly lowered blood urea and urine proteinuria levels over the ensuing 2 weeks. In addition, nearly two-thirds of transplanted -/- mice maintained their weight above the 80% welfare threshold, with both males and females weighing more than age-matched nontransplanted -/- mice. This was associated with less renal cortical fibrosis and interstitial inflammation compared to nontransplanted mice as shown by reduction in murine CD4, CD68, and CD45.2 cells. Transplanted CSC homed to glomeruli, where they expressed CR1, VEGFA, SYNAPTOPODIN, CD2AP, and PODOCIN at the RNA level and produced PODOCIN, CD2AP, and COLIVα3 proteins in nontransplanted -/- mice, indicating that CSC have adopted a podocyte phenotype. Together, these data indicate that CSC may be used to delay progression of renal pathology by a combination of anti-inflammatory effects and replacement of the defective resident podocytes.


Asunto(s)
Diferenciación Celular , Corion/citología , Nefritis Hereditaria/terapia , Podocitos/citología , Células Madre/citología , Animales , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Corion/trasplante , Técnicas de Cocultivo , Colágeno Tipo IV/farmacología , Regulación hacia Abajo/efectos de los fármacos , Femenino , Fibrosis , Humanos , Inflamación/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Corteza Renal/patología , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Mutación/genética , Nefritis Hereditaria/patología , Fenotipo , Podocitos/efectos de los fármacos , Podocitos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo
14.
Artículo en Inglés | MEDLINE | ID: mdl-26427551

RESUMEN

Pluripotency defines the ability of stem cells to differentiate into all the lineages of the three germ layers and self-renew indefinitely. Somatic cells can regain the developmental potential of embryonic stem cells following ectopic expression of a set of transcription factors or, in certain circumstances, via modulation of culture conditions and supplementation with small molecule, that is, induced pluripotent stem (iPS) cells. Here, we discuss the use of fetal tissues for reprogramming, focusing in particular on stem cells derived from human amniotic fluid, and the development of chemical reprogramming. We next address the advantages and disadvantages of deriving pluripotent cells from fetal tissues and the potential clinical applications.


Asunto(s)
Células Madre Fetales/citología , Células Madre Pluripotentes Inducidas/citología , Líquido Amniótico/citología , Diferenciación Celular , Técnicas de Reprogramación Celular , Feto/citología , Humanos , Medicina Regenerativa
16.
Stem Cells Dev ; 24(1): 132-43, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25072397

RESUMEN

Amniotic Fluid Stem (AFS) cells are broadly multipotent fetal stem cells derived from the positive selection and ex vivo expansion of amniotic fluid CD117/c-kit(pos) cells. Considering the differentiation potential in vitro toward cell lineages belonging to the three germ layers, AFS cells have raised great interest as a new therapeutic tool, but their immune properties still need to be assessed. We analyzed the in vitro immunological properties of AFS cells from different gestational age in coculture with T, B, and natural killer (NK) cells. Nonactivated (resting) first trimester-AFS cells showed lower expression of HLA class-I molecules and NK-activating ligands than second and third trimester-AFS cells, whose features were associated with lower sensitivity to NK cell-mediated lysis. Nevertheless, inflammatory priming with interferon gamma (IFN-γ) and tumor necrosis factor alpha (TNF-α) enhanced resistance of all AFS cell types to NK cytotoxicity. AFS cells modulated lymphocyte proliferation in a different manner according to gestational age: first trimester-AFS cells significantly inhibited T and NK cell proliferation, while second and third trimester-AFS cells were less efficient. In addition, only inflammatory-primed second trimester-AFS cells could suppress B cell proliferation, which was not affected by the first and third trimester-AFS cells. Indolamine 2,3 dioxygenase pathway was significantly involved only in T cell suppression mediated by second and third trimester-AFS cells. Overall, this study shows a number of significant quantitative differences among AFS cells of different gestational age that have to be considered in view of their clinical application.


Asunto(s)
Líquido Amniótico/inmunología , Proliferación Celular , Edad Gestacional , Tolerancia Inmunológica , Linfocitos/inmunología , Células Madre Multipotentes/inmunología , Proteínas Proto-Oncogénicas c-kit/inmunología , Líquido Amniótico/citología , Técnicas de Cocultivo , Femenino , Humanos , Linfocitos/citología , Células Madre Multipotentes/citología , Embarazo , Tercer Trimestre del Embarazo/inmunología
17.
Cell Reprogram ; 16(5): 331-44, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25162836

RESUMEN

Trisomy 21 is the most common chromosomal abnormality and is associated primarily with cardiovascular, hematological, and neurological complications. A robust patient-derived cellular model is necessary to investigate the pathophysiology of the syndrome because current animal models are limited and access to tissues from affected individuals is ethically challenging. We aimed to derive induced pluripotent stem cells (iPSCs) from trisomy 21 human mid-trimester amniotic fluid stem cells (AFSCs) and describe their hematopoietic and neurological characteristics. Human AFSCs collected from women undergoing prenatal diagnosis were selected for c-KIT(+) and transduced with a Cre-lox-inducible polycistronic lentiviral vector encoding SOX2, OCT4, KLF-4, and c-MYC (50,000 cells at a multiplicity of infection (MOI) 1-5 for 72 h). The embryonic stem cell (ESC)-like properties of the AFSC-derived iPSCs were established in vitro by embryoid body formation and in vivo by teratoma formation in RAG2(-/-), γ-chain(-/-), C2(-/-) immunodeficient mice. Reprogrammed cells retained their cytogenetic signatures and differentiated into specialized hematopoietic and neural precursors detected by morphological assessment, immunostaining, and RT-PCR. Additionally, the iPSCs expressed all pluripotency markers upon multiple rounds of freeze-thawing. These findings are important in establishing a patient-specific cellular platform of trisomy 21 to study the pathophysiology of the aneuploidy and for future drug discovery.


Asunto(s)
Líquido Amniótico/citología , Criopreservación , Síndrome de Down , Células Madre Pluripotentes Inducidas/citología , Modelos Biológicos , Animales , Femenino , Humanos , Ratones , Embarazo , Diagnóstico Prenatal
18.
Nucleic Acids Res ; 42(14): 9424-35, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25056318

RESUMEN

We have sequenced miRNA libraries from human embryonic, neural and foetal mesenchymal stem cells. We report that the majority of miRNA genes encode mature isomers that vary in size by one or more bases at the 3' and/or 5' end of the miRNA. Northern blotting for individual miRNAs showed that the proportions of isomiRs expressed by a single miRNA gene often differ between cell and tissue types. IsomiRs were readily co-immunoprecipitated with Argonaute proteins in vivo and were active in luciferase assays, indicating that they are functional. Bioinformatics analysis predicts substantial differences in targeting between miRNAs with minor 5' differences and in support of this we report that a 5' isomiR-9-1 gained the ability to inhibit the expression of DNMT3B and NCAM2 but lost the ability to inhibit CDH1 in vitro. This result was confirmed by the use of isomiR-specific sponges. Our analysis of the miRGator database indicates that a small percentage of human miRNA genes express isomiRs as the dominant transcript in certain cell types and analysis of miRBase shows that 5' isomiRs have replaced canonical miRNAs many times during evolution. This strongly indicates that isomiRs are of functional importance and have contributed to the evolution of miRNA genes.


Asunto(s)
MicroARNs/metabolismo , Animales , Proteínas Argonautas/metabolismo , Línea Celular , Evolución Molecular , Humanos , Ratones , MicroARNs/química , MicroARNs/genética , Precursores del ARN/química , ARN Mensajero/metabolismo , Células Madre/metabolismo
19.
Clin Perinatol ; 41(1): 133-48, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24524451

RESUMEN

This article introduces the basic concepts of modeling neonatal brain injury and provides background information regarding each of the commonly used types of stem cells. It summarizes the findings of preclinical research testing the therapeutic potential of stem cells in animal models of neonatal brain injury, reports briefly on the status of clinical trials, and discusses the important ongoing issues that need to be addressed before stem cell therapy is used to repair the injured brain.


Asunto(s)
Lesiones Encefálicas/terapia , Células Madre Embrionarias/trasplante , Hipoxia-Isquemia Encefálica/terapia , Células-Madre Neurales/trasplante , Trasplante de Células Madre/métodos , Amnios/citología , Animales , Terapia Combinada , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Modelos Animales de Enfermedad , Femenino , Humanos , Hipotermia Inducida , Recién Nacido , Trasplante de Células Madre Mesenquimatosas/métodos , Placenta/citología , Embarazo
20.
Stem Cells Dev ; 23(5): 541-54, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24172175

RESUMEN

Cartilage injuries and osteoarthritis are leading causes of disability in developed countries. The regeneration of damaged articular cartilage using cell transplantation or tissue engineering holds much promise but requires the identification of an appropriate cell source with a high proliferative propensity and consistent chondrogenic capacity. Human fetal mesenchymal stem cells (MSCs) have been isolated from a range of perinatal tissues, including first-trimester bone marrow, and have demonstrated enhanced expansion and differentiation potential. However, their ability to form mature chondrocytes for use in cartilage tissue engineering has not been clearly established. Here, we compare the chondrogenic potential of human MSCs isolated from fetal and adult bone marrow and show distinct differences in their responsiveness to specific growth factors. Transforming growth factor beta 3 (TGFß3) induced chondrogenesis in adult but not fetal MSCs. In contrast, bone morphogenetic protein 2 (BMP2) induced chondrogenesis in fetal but not adult MSCs. When fetal MSCs co-stimulated with BMP2 and TGFß3 were used for cartilage tissue engineering, they generated tissue with type II collagen and proteoglycan content comparable to adult MSCs treated with TGFß3 alone. Investigation of the TGFß/BMP signaling pathway showed that TGFß3 induced phosphorylation of SMAD3 in adult but not fetal MSCs. These findings demonstrate that the initiation of chondrogenesis is modulated by distinct signaling mechanisms in fetal and adult MSCs. This study establishes the feasibility of using fetal MSCs in cartilage repair applications and proposes their potential as an in vitro system for modeling chondrogenic differentiation and skeletal development studies.


Asunto(s)
Diferenciación Celular/genética , Condrogénesis/genética , Células Madre Mesenquimatosas/citología , Transducción de Señal/genética , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Células de la Médula Ósea/citología , Cartílago/patología , Cartílago/trasplante , Células Cultivadas , Condrocitos/citología , Condrocitos/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Ingeniería de Tejidos , Factor de Crecimiento Transformador beta3
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...