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











Base de datos
Intervalo de año de publicación
1.
Cell Biol Int ; 47(6): 1033-1048, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36994843

RESUMEN

Recently, mesenchymal stem/stromal cells (MSCs) transplantation has been introduced as a promising option to support cartilage structure and improve its function in preclinical models and patients suffering from osteoarthritis (OA). MSCs strongly provoke their preferred influence in vivo by inhibiting the inflammatory responses and applying immunomodulation by releasing anti-inflammatory mediators such as transforming growth factor-ß and interleukin-10. Such mediators downregulate fibroblast-like synoviocytes growth and migration, leading to chondroprotection. Furthermore, improving the chondrocyte proliferation and extracellular matrix hemostasis in addition to the suppression of the matrix metalloproteinases activities can support cartilage tissue organization. In this light, various published results have demonstrated that MSCs therapy can considerably decrease pain and restore knee function in OA patients. In the current review, we have concentrated on recent advances in MSCs-based therapeutics to elicit both chondrogenic and chondroprotective impacts in OA patients, focusing on the last decade in vivo results.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Osteoartritis , Humanos , Cartílago , Matriz Extracelular , Trasplante de Células Madre Mesenquimatosas/métodos , Condrocitos
2.
J Biomed Mater Res B Appl Biomater ; 106(4): 1578-1586, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28792664

RESUMEN

Human endometrial stem cells (hEnSCs) are known as an attractive source of stem cells for regenerative medicine. hEnSCs are easily isolated and are capable of repairing uterine through their strong ability of creating new capillaries. In this study, a three-dimensional (3D) nanofibrous polycaprolactone (PCL)/collagen scaffold was fabricated and characterized in order to be applied as a new approach for skin reconstruction. Furthermore, the behavior of hEnSCs on this scaffold was investigated. First, a PCL 3D scaffold was constructed using electrospinning technique. Plasma treated and PCL was grafted by collagen. The constructs were characterized for mechanical and structural properties. Cell attachment, proliferation, viability, and differentiation of hEnSCs were assessed after being seeded on PCL and PCL/collagen scaffolds using scanning electron microscopy, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, and real-time polymerase chain reaction tests. The results showed higher wettability for the PCL/collagen scaffold with desirable mechanical and structural characteristics compared to PCL and collagen alone. The attachment and proliferation rates of hEnSCs on the PCL/collagen scaffold were higher compared to those on the bare PCL. Hence, hEnSCs are newly discovered stem cell source for skin tissue engineering in vitro, particularly when developed on PCL/collagen nanofiber scaffolds. Therefore, application of hEnSCs for skin regeneration is a novel therapeutic approach for temporary skin substitute. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 1578-1586, 2018.


Asunto(s)
Endometrio , Poliésteres/química , Piel , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Adulto , Proliferación Celular , Células Cultivadas , Endometrio/citología , Endometrio/metabolismo , Femenino , Humanos , Masculino , Piel/citología , Piel/metabolismo
3.
J Tissue Eng Regen Med ; 9(11): 1268-76, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23319462

RESUMEN

Reconstruction of the bladder wall via in vitro differentiated stem cells on an appropriate scaffold could be used in such conditions as cancer and neurogenic urinary bladder. This study aimed to examine the potential of human endometrial stem cells (EnSCs) to form urinary bladder epithelial cells (urothelium) on nanofibrous silk-collagen scaffolds, for construction of the urinary bladder wall. After passage 4, EnSCs were induced by keratinocyte growth factor (KGF) and epidermal growth factor (EGF) and seeded on electrospun collagen-V, silk and silk-collagen nanofibres. Later we tested urothelium-specific genes and proteins (uroplakin-Ia, uroplakin-Ib, uroplakin-II, uroplakin-III and cytokeratin 20) by immunocytochemistry, RT-PCR and western blot analyses. Scanning electron microscopy (SEM) and histology were used to detect cell-matrix interactions. DMEM/F12 supplemented by KGF and EGF induced EnSCs to express urothelial cell-specific genes and proteins. Either collagen, silk or silk-collagen scaffolds promoted cell proliferation. The nanofibrous silk-collagen scaffolds provided a three-dimensional (3D) structure to maximize cell-matrix penetration and increase differentiation of the EnSCs. Human EnSCs seeded on 3D nanofibrous silk-collagen scaffolds and differentiated to urothelial cells provide a suitable source for potential use in bladder wall reconstruction in women.


Asunto(s)
Endometrio/citología , Células Madre/citología , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Vejiga Urinaria/patología , Urotelio/patología , Adulto , Materiales Biocompatibles/farmacología , Técnicas de Cultivo de Célula , Diferenciación Celular , Células Cultivadas , Colágeno/química , Ensayo de Inmunoadsorción Enzimática , Factor de Crecimiento Epidérmico/metabolismo , Femenino , Factor 7 de Crecimiento de Fibroblastos/metabolismo , Humanos , Inmunohistoquímica , Queratinocitos/citología , Microscopía Electrónica de Rastreo , Nanofibras/química , Fenotipo , Seda/química , Urotelio/citología
4.
Neural Regen Res ; 9(8): 845-50, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25206899

RESUMEN

Several studies have demonstrated that selective serotonin reuptake inhibitor antidepressants can promote neuronal cell proliferation and enhance neuroplasticity both in vitro and in vivo. It is hypothesized that citalopram, a selective serotonin reuptake inhibitor, can promote the neuronal differentiation of adult bone marrow mesenchymal stem cells. Citalopram strongly enhanced neuronal characteristics of the cells derived from bone marrow mesenchymal stem cells. The rate of cell death was decreased in citalopram-treated bone marrow mesenchymal stem cells than in control cells in neurobasal medium. In addition, the cumulative population doubling level of the citalopram-treated cells was significantly increased compared to that of control cells. Also BrdU incorporation was elevated in citalopram-treated cells. These findings suggest that citalopram can improve the neuronal-like cell differentiation of bone marrow mesenchymal stem cells by increasing cell proliferation and survival while maintaining their neuronal characteristics.

5.
Cell Biol Int ; 38(8): 901-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24715678

RESUMEN

An increase in the number of viable in vitro differentiated neuronal cells is important for their use in clinics. A proportion of differentiated cells lose their viability before being used, and therefore we decided to use a pharmacological agent, sertraline, to increase neural cell differentiation and their survival. Purified endometrial stem cells (EnSCs) were examined for neuronal and glial cell specific markers after retinoic acid (RA) and sertraline treatment via RT-PCR, immunocytochemistry and Western blot analysis. The survival of differentiated cells was measured by MTT assay and the frequency of apoptosis, demonstrated by caspase-3-like activity. EnSCs were differentiated into neuronal cells after RA induction. Sertraline increased neuronal cell differentiation by 1.2-fold and their survival by 1.4-fold, and decreased from glial cell differentiation significantly. The findings indicate that sertraline could be used to improve the in vitro differentiation process of stem cells into neuronal cells, and may be involved in regenerative pharmacology in future.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Células Madre Mesenquimatosas/fisiología , Neuroglía/fisiología , Neuronas/fisiología , Sertralina/farmacología , Tretinoina/farmacología , Antígenos de Diferenciación/metabolismo , Apoptosis/efectos de los fármacos , Diferenciación Celular , Células Cultivadas , Endometrio/citología , Femenino , Humanos , Neuroglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología
6.
BJU Int ; 112(6): 854-63, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24028767

RESUMEN

OBJECTIVE: To investigate manufacturing smooth muscle cells (SMCs) for regenerative bladder reconstruction from differentiation of endometrial stem cells (EnSCs), as the recent discovery of EnSCs from the lining of women's uteri, opens up the possibility of using these cells for tissue engineering applications, such as building up natural tissue to repair prolapsed pelvic floors as well as building urinary bladder wall. MATERIALS AND METHODS: Human EnSCs that were positive for cluster of differentiation 146 (CD146), CD105 and CD90 were isolated and cultured in Dulbecco's modified Eagle/F12 medium supplemented with myogenic growth factors. The myogenic factors included: transforming growth factor ß, platelet-derived growth factor, hepatocyte growth factor and vascular endothelial growth factor. Differentiated SMCs on bioabsorbable polyethylene-glycol and collagen hydrogels were checked for SMC markers by real-time reverse-transcriptase polymerase chain reaction (RT-PCR), western blot (WB) and immunocytochemistry (ICC) analyses. RESULTS: Histology confirmed the growth of SMCs in the hydrogel matrices. The myogenic growth factors decreased the proliferation rate of EnSCs, but they differentiated the human EnSCs into SMCs more efficiently on hydrogel matrices and expressed specific SMC markers including α-smooth muscle actin, desmin, vinculin and calponin in RT-PCR, WB and ICC experiments. The survival rate of cultures on the hydrogel-coated matrices was significantly higher than uncoated cultures. CONCLUSIONS: Human EnSCs were successfully differentiated into SMCs, using hydrogels as scaffold. EnSCs may be used for autologous bladder wall regeneration without any immunological complications in women. Currently work is in progress using bioabsorbable nanocomposite materials as EnSC scaffolds for developing urinary bladder wall tissue.


Asunto(s)
Endometrio/ultraestructura , Miocitos del Músculo Liso/ultraestructura , Células Madre/citología , Ingeniería de Tejidos/métodos , Actinas/biosíntesis , Actinas/genética , Adulto , Biopsia , Western Blotting , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Endometrio/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Microscopía Electrónica de Rastreo , Miocitos del Músculo Liso/metabolismo , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Factor de Crecimiento Derivado de Plaquetas/genética , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo , Factor de Crecimiento Transformador beta/biosíntesis , Factor de Crecimiento Transformador beta/genética , Vejiga Urinaria/citología
7.
Neurol Sci ; 34(1): 19-23, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22258359

RESUMEN

The enzyme 5alpha-reductase 1 (5α-R(1)) that converts testosterone (T) to dihydrotestosterone (DHT) is present in many mammalian tissues including the spinal cord. It is established that morphine administration decreases spinal cord T levels, but the mechanism is still undetermined. Here, we investigated the link between T and the enzyme 5α-R(1) in the spinal cord after morphine administration. For spinal cord steroid extraction, all the animals were killed 30 min, 2 h (acute) and 14 days (chronic) after first drug injection by decapitation. The whole spinal cord was removed and kept frozen at -20°C until T and DHT extraction. The effects of acute and chronic morphine administration on 5α-R(1) expression in the adult male rat spinal cord were evaluated using RT-PCR. Spinal cord T and DHT levels were measured using radioimmunoassay before and after the morphine exposure. Morphine significantly reduced the T concentration after acute and chronic exposure in the spinal cord. In contrast, the 5α-R(1) expression and of course DHT levels increased the following chronic morphine administration. One important reason for the decreasing effect of morphine exposure on the spinal cord T level is due to an increase in the 5α-R(1) levels. We suggest that morphine plays a regulatory role in metabolism of neurosteroids, especially T in the spinal cord via 5α-R(1).


Asunto(s)
3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/fisiología , Morfina/farmacología , Narcóticos/farmacología , Médula Espinal/metabolismo , Testosterona/metabolismo , Animales , Dihidrotestosterona/metabolismo , Masculino , Neurotransmisores/metabolismo , Radioinmunoensayo , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Médula Espinal/efectos de los fármacos , Médula Espinal/enzimología
8.
Arch Immunol Ther Exp (Warsz) ; 61(1): 75-83, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23224340

RESUMEN

Phage display of many nanobodies via filamentous phage in combination with helper phage has been reported by many scientists. The aim of this study was to produce lambda (λ) bacteriophage displaying high-affinity nanobody against HER-2 expressing breast carcinoma cells. Bacteriophage λ is a temperate phage with inherent biological safety in mammalian cells. Here we report the construction of a recombinant λ phage that efficiently expresses specific nanobody towards third domain of HER-2 target on SKBR-3 and MCF-7 cell lines in vitro. We constructed recombinant λ phage particles containing a mammalian expression cassette, C-Myc tagged, encoding VHH gene of camelid anti HER-2 third domain epitope using λ ZAP-cytomegalic virus (CMV) vector. The SKBR-3, MCF-7 and human endometrial stem cells were treated by the nanobody displayed recombinant λ phage. The cell growth inhibition assay was performed by MTT Cell Viability Assay Kit. After the fourth round of biopanning there was a significant enrichment in the phage specifically binding to the antigen. The ratio of targeted phage increased approximately 1,000-fold in the fifth round. The nanobody expressed by λ ZAP-CMV-VHH phagemid cloned in λ bioparticles significantly inhibited the proliferation of HER-2 positive SKBR-3 and MCF-7 cells. Recombinant bacteriophage λ ZAP-CMV-VHH-cDNA could be used efficiently for construction of nanobodies to mortify HER-2 positive breast carcinoma cells as a nanomedical therapeutic.


Asunto(s)
Neoplasias de la Mama/inmunología , Carcinoma/inmunología , Técnicas de Visualización de Superficie Celular/métodos , Anticuerpos de Dominio Único/farmacología , Células Madre Adultas/efectos de los fármacos , Animales , Afinidad de Anticuerpos , Bacteriófago lambda , Neoplasias de la Mama/terapia , Camélidos del Nuevo Mundo , Carcinoma/terapia , Proliferación Celular/efectos de los fármacos , Citomegalovirus/genética , Epítopos de Linfocito B/inmunología , Genes myc/genética , Ingeniería Genética , Humanos , Células MCF-7 , Estructura Terciaria de Proteína/genética , Receptor ErbB-2/genética , Receptor ErbB-2/inmunología , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/aislamiento & purificación
9.
Cell Biol Int ; 36(10): 961-6, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22804708

RESUMEN

The potential of cell therapy is promising in nerve regeneration, but is limited by ethical considerations about the proper and technically safe source of stem cells. We report the successful differentiation of human EnSCs (endometrial stem cells) as a rich source of renewable and safe progenitors into high-efficiency cholinergic neurons. The extracellular signals of NGF (nerve growth factor) and bFGF (basic fibroblast growth factor) could induce cholinergic neuron differentiation. ChAT (choline acetyltransferase), MAP2 (microtubule associated protein 2) and NF-l (neurofilament L) increased after administration of bFGF and NGF to the EnSC cultures. trkC and FGFR2 (fibroblast growth factor receptor 2), which belong to the NGF and bFGF receptors respectively, were determined in populations of EnSCs. NGF, bFGF and their combination differentially influenced human EnSCs high efficiency differentiation. By inducing cholinergic neurons from EnSCs in a chemically defined medium, we could produce human neural cells without resorting to primary culture of neurons. This in vitro method provides an unlimited source of human neural cells and facilitates clinical applications of EnSCs for neurological diseases.


Asunto(s)
Endometrio/citología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Neurogénesis , Neuronas/citología , Células Madre/citología , Adulto , Antígeno CD146/análisis , Separación Celular , Supervivencia Celular , Células Cultivadas , Colina O-Acetiltransferasa/análisis , Femenino , Humanos , Proteínas Asociadas a Microtúbulos/análisis , Proteínas de Neurofilamentos/análisis , Neuronas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/análisis , Antígenos Thy-1/análisis , Molécula 1 de Adhesión Celular Vascular/análisis
10.
Cell Biol Int ; 35(10): 1037-41, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21355850

RESUMEN

Expression of four major reprogramming transgenes, including Oct4, Sox2, Klf4 and c-myc, in somatic cells enables them to have pluripotency. These cells are iPSC (induced pluripotent stem cell) that currently show the greatest potential for differentiation into cells of the three germ lineages. One of the issues facing the successful reprogramming and clinical translation of iPSC technology is the high rate of apoptosis after the reprogramming process. Reprogramming is a stressful process, and the p53 apoptotic pathway plays a negative role in cell growth and self-renewal. Apoptosis via the p53 pathway serves as a major barrier in nuclear somatic cell reprogramming during iPSC generation. DHEA (dehydroepiandrosterone) is an abundant steroid that is produced at high levels in the adrenal cells, and withdrawal of DHEA increases the levels of p53 in the epithelial and stromal cells, resulting in increased levels of apoptotic cells; meanwhile, DHEA decreases cellular apoptosis. DHEA could improve the efficacy of reprogramming yield due to a decrease in apoptosis via the p53 pathway and an increase in cell viability.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Reprogramación Celular/efectos de los fármacos , Deshidroepiandrosterona/farmacología , Células Madre Pluripotentes/citología , Apoptosis , Humanos , Factor 4 Similar a Kruppel , Modelos Teóricos , Células Madre Pluripotentes/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
11.
Med Hypotheses ; 76(6): 843-6, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21402451

RESUMEN

Recently, transplantation of adult stem cells over embryonic stem cells increased in regenerative medicine. Among the adult stem cells, human endometrium stromal (hEnS) cells are under the strict control of the steroid hormones and have the potential to differentiate into other cell lineages including neural cells. Unfortunately these cells may lose their neurogenic differentiation ability upon extended expansion in cultures. To avoid the back-differentiation, it is important to establish growth conditions that support the rapid proliferation and stable differentiation of hEnS cells over extended periods of time without compromising their neuronal phenotype. Differentiation of transplanted cells is strongly influenced by environmental signals. The steroidal microenvironment of the stem cells plays a major role in controlling neurogenesis in the cultures. Dehydroepiandrosterone (DHEA) administration to the cultures could support this propose. DHEA enhance survival rates of dissociated neurons in cultures. It can activate AKT protein kinase pathway as well as nerve growth factor (NGF) that enhances neurogenesis efficiently. On the other hand it seems that DHEA increase survival rate of neural cells via production of brain derived neurotrophic factor (BDNF), indirectly. BDNF is a mediator product of the DHEA that promotes the differentiation and survival of neurons. Here, we offer that DHEA is a suitable candidate that could provide a microenvironment to stimulate neurogenesis and enhanced survival of newly formed neurons derived from hEnS cells. From the point that DHEA is the most abundant steroid in the body, marketed as a supplement and is increasingly self-prescription we hypothesized that it could be the safe and high available choice. This provides a better insight into the maintenance of neural cells for treatment of a wide variety of neurological diseases such as Alzheimer's and Parkinson's by non-invasively autologous cell therapy by hEnS cells especially in women.


Asunto(s)
Deshidroepiandrosterona/administración & dosificación , Endometrio/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Células Madre/efectos de los fármacos , Deshidroepiandrosterona/farmacología , Endometrio/citología , Femenino , Humanos , Células Madre/citología
12.
Med Hypotheses ; 76(1): 11-3, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20851532

RESUMEN

Recent advances in stem cell biology have resulted in identifying new agents to differentiate stem cell-derived-neural cells. Different stem cell types have been shown to differentiate into neural cells. It has been shown that P19 line of embryonal carcinoma cells develops into neurons and astroglia after exposure to some hormones such as dehydroepiandrosterone (DHEA). Steroid 5α-reductase is a key enzyme in the conversion of several Δ4-3 keto steroids, such as testosterone into their respective 5α-reductase derivatives. Finasteride is a 5α-reductase inhibitor that inhibits conversion of testosterone to the more potent androgen dihydrotestosterone (DHT). Reduction in DHT and sustaining testosterone levels has an important impact on differentiation and proliferation of embryonal carcinoma cells to neural cells. We hypothesize that finasteride, a 5α-reductase inhibitor, will be differentiate embryonal carcinoma cell to the neural cell and increase their proliferation due to the elevation levels of testosterone, a neuroprotective neurosteroid.


Asunto(s)
Inhibidores de 5-alfa-Reductasa/farmacología , Carcinoma Embrionario/patología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Finasterida/farmacología , Neuronas/efectos de los fármacos , Humanos , Neuronas/patología
13.
Med Hypotheses ; 76(2): 225-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21051157

RESUMEN

In spite of widespread use of morphine to treat pain in patients, little is known about the effects of this opioid on many cells including stem cells. Moreover the studies have been shown controversial results about morphine effects on several kinds of cells. It is well-known that morphine exposure could decrease testosterone levels in brain and spinal cord. Morphine could increase the activity of 5α-redutase, the enzyme that converts testosterone into its respective 5α-redutase derivative dihydrotestosterone (DHT). Also it could increase aromatase activity that converts testosterone to estradiol. Proliferation of neural stem cells was observed in human stem cells after exposure to certain combinations of steroids especially testosterone. On the other hand DHT has negative effect in neural stem cell reproduction. Morphine induces over-expression of p53 gene that could mediate stem cell apoptosis. Therefore we hypothesized that due to reduction in the testosterone levels, elevation in the DHT levels, and over-expression of p53 gene, morphine could prevent neural stem cell proliferation.


Asunto(s)
Morfina/farmacología , Células-Madre Neurales/efectos de los fármacos , Enfermedades Neurodegenerativas/prevención & control , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/metabolismo , Inhibidores de 5-alfa-Reductasa/farmacología , Animales , Apoptosis , Proliferación Celular , Dihidrotestosterona/metabolismo , Genes p53 , Humanos , Modelos Biológicos , Enfermedades Neurodegenerativas/terapia , Ratas , Regeneración , Testosterona/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA