Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Cell Biol Int ; 38(9): 1050-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24797366

RESUMEN

The beneficial effects of mesenchymal stem cells (MSCs) are mediated partly by the paracrine production of cytoprotective and trophic factors. Vascular endothelial growth factor (VEGF) is released from MSCs as a paracrine trophic factor and contributes to the therapeutic effects of the stem cell by regulating angiogenesis and promoting revascularization in injured tissues. Interleukin-8 (IL-8), an inflammatory chemokine with potent proangiogenic properties, is upregulated in the ischemic brain and has been shown to promote homing of bone marrow-derived cells to injured sites. However, the effect of IL-8 on MSCs paracrine function remains unknown. We found that IL-8 induced VEGF production and phosphorylation of Akt and ERK. Both effects could be blocked by inhibitors (LY294002, PD098059) or siRNA-mediated silencing of Akt and ERK in human bone marrow MSCs (hBM-MSCs). IL-8-induced VEGF production in hBM-MSCs significantly increased tube formation on Matrigel compared with basal secreted VEGF. In a rat stroke model, administration of IL-8-treated hBM-MSCs decreased the infarction volume and increased angiogenesis in the ischemic boundary zone compared with hBM-MSC treatment alone. In conclusion, IL-8 stimulates VEGF production in hBM-MSCs in part via the PI3K/Akt and MAPK/ERK signal transduction pathways and that administration of IL-8-treated hBM-MSCs increases angiogenesis after stroke. This approach may be used to optimize MSC-based therapies for numerous diseases including stroke, myocardial ischemia, and spinal cord injury.


Asunto(s)
Células de la Médula Ósea/citología , Interleucina-8/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Encéfalo/metabolismo , Células Cultivadas , Cromonas/farmacología , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Flavonoides/farmacología , Humanos , Isquemia/metabolismo , Isquemia/patología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Morfolinas/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos
2.
Biochem Biophys Res Commun ; 431(2): 354-9, 2013 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-23261452

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) triggers specific apoptosis in tumor cells and is one of the most promising candidates for cancer gene therapy. However, resistance to TRAIL is one of the main impediments to use of TRAIL in cancer treatment. We showed previously that the lipoxygenase inhibitor MK886 in combination with TRAIL exhibits enhanced antitumor activities compared with each agent alone in human glioma cells. In this study, we elucidated the molecular mechanisms responsible for MK886-mediated sensitization to TRAIL-induced apoptosis. We found that MK886 sensitized glioma cells to TRAIL-induced apoptosis by upregulating the death receptor 5 (DR5) and that specific knockdown of DR5 attenuated cell death. The mechanisms underlying this sensitization involved activation of the MK886-induced p38 mitogen-activated protein kinase (MAPK) pathway and subsequent DR5 overexpression. However, treatment with a specific inhibitor or gene silencing of p38 MAPK abolished both the DR5 induction and the increase in apoptosis caused by TRAIL. Taken together, our findings indicate that the increased expression of DR5 in a p38 MAPK-dependent manner plays an important role in the sensitization of MK886 to TRAIL-induced apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Glioma/metabolismo , Indoles/farmacología , Inhibidores de la Lipooxigenasa/farmacología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Sinergismo Farmacológico , Técnicas de Silenciamiento del Gen , Humanos , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
3.
Biochem Biophys Res Commun ; 421(3): 585-90, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22525671

RESUMEN

Suicide gene therapy of glioma based on herpes simplex virus type I thymidine kinase (HSV-TK) and prodrug ganciclovir (GCV) suffers from the lack of efficacy in clinical trials, which is mostly due to low transduction efficacy and absence of bystander effect in tumor cells. Recently, stem cells as cellular delivery vehicles of prodrug converting gene has emerged as a new treatment strategy for malignant glioma. In this study, we evaluated the anti-glioma effect of suicide gene therapy using human bone marrow mesenchymal stem cells expressing HSV-TK (MSCs-TK) combined with valproic acid (VPA), which can upregulate the gap junction proteins and may enhance the bystander effect of suicide gene therapy. Expression of HSV-TK in MSCs was confirmed by RT-PCR analysis and the sensitivity of MSCs-TK to GCV was assessed. A bystander effect was observed in co-cultures of MSCs-TK and U87 glioma cells by GCV in a dose-dependent manner. VPA induced the expression of the gap junction proteins connexin (Cx) 43 and 26 in glioma cell and thereby enhanced the bystander effect in co-culture experiment. The enhanced bystander effect was inhibited by the gap junction inhibitor 18-ß-glycyrrhetinic acid (18-GA). Moreover, the combined treatment with VPA and MSCs-TK synergistically enhanced apoptosis in glioma cells by caspase activation. In vivo efficacy experiments showed that combination treatment of MSCs-TK and VPA significantly inhibited tumor growth and prolonged the survival of glioma-bearing mice compared with single-treatment groups. In addition, TUNEL staining also demonstrated a significant increase in the number of apoptotic cells in the combination treated group compared with single-treatment groups. Taken together, these results provide the rational for designing novel experimental protocols to increase bystander killing effect against intracranial gliomas using MSCs-TK and VPA.


Asunto(s)
Neoplasias Encefálicas/terapia , Efecto Espectador , Terapia Genética/métodos , Glioma/terapia , Células Madre Mesenquimatosas/enzimología , Timidina Quinasa/genética , Ácido Valproico/administración & dosificación , Animales , Apoptosis , Línea Celular Tumoral , Conexinas/agonistas , Conexinas/biosíntesis , Herpesvirus Humano 1/enzimología , Humanos , Trasplante de Células Madre Mesenquimatosas , Ratones , Ratones Desnudos , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Reprod Biol Endocrinol ; 10: 31, 2012 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-22531046

RESUMEN

BACKGROUND: Interleukin (IL)-18 is an important regulator of innate and acquired immune responses and has multiple roles in chronic inflammation and autoimmune disorders. Obesity is characterized by low- grade chronic inflammation. IL-18 has been suggested as an adipogenic cytokine that is associated with excess adiposity. The purpose of this study is to evaluate the relationship between IL-18 gene polymorphisms (-137 G/C and -607 C/A) and obesity. METHODS: All 680 subjects were genotyped for the polymorphisms of IL-18 gene promoters (at positions -137 G/C and -607 C/A) using a polymerase chain reaction (271 cases with BMI ≥25 kg/m² and 409 controls with BMI <25 kg/m²). A chi-square test was used to compare the genotype and allele frequencies between the cases and control populations. RESULTS: Analyses of the genotype distributions revealed that IL-18 -607 C/A polymorphism was associated with an increase in body mass index in obese women in the Korean population (chi(2) = 12.301, df = 2, p = 0.015). CONCLUSION: Carriage of the A allele at position -607 in the promoter of the IL-18 gene may have a role in the development of obesity.


Asunto(s)
Índice de Masa Corporal , Interleucina-18/genética , Obesidad/genética , Adulto , Pueblo Asiatico/genética , Femenino , Frecuencia de los Genes , Haplotipos , Humanos , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple
5.
J Biomed Biotechnol ; 2012: 362473, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22500090

RESUMEN

Numerous studies have shown the benefits of mesenchymal stem cells (MSCs) on the repair of spinal cord injury (SCI) model and on behavioral improvement, but the underlying mechanisms remain unclear. In this study, to investigate possible mechanisms by which MSCs contribute to the alleviation of neurologic deficits, we examined the potential effect of human umbilical cord blood-derived MSCs (hUCB-MSCs) on the endogenous cell proliferation and oligogenesis after SCI. SCI was injured by contusion using a weight-drop impactor and hUCB-MSCs were transplanted into the boundary zone of the injured site. Animals received a daily injection of bromodeoxyuridine (BrdU) for 7 days after treatment to identity newly synthesized cells of ependymal and periependymal cells that immunohistochemically resembled stem/progenitor cells was evident. Behavior analysis revealed that locomotor functions of hUCB-MSCs group were restored significantly and the cavity volume was smaller in the MSCs-transplanted rats compared to the control group. In MSCs-transplanted group, TUNEL-positive cells were decreased and BrdU-positive cells were significantly increased rats compared with control group. In addition, more of BrdU-positive cells expressed neural stem/progenitor cell nestin and oligo-lineage cell such as NG2, CNPase, MBP and glial fibrillary acidic protein typical of astrocytes in the MSC-transplanted rats. Thus, endogenous cell proliferation and oligogenesis contribute to MSC-promoted functional recovery following SCI.


Asunto(s)
Sangre Fetal/citología , Trasplante de Células Madre Mesenquimatosas , Neurogénesis/fisiología , Traumatismos de la Médula Espinal/cirugía , Cicatrización de Heridas/fisiología , Análisis de Varianza , Animales , Apoptosis/fisiología , Conducta Animal/fisiología , Procesos de Crecimiento Celular/fisiología , Histocitoquímica , Humanos , Masculino , Células Madre Mesenquimatosas/fisiología , Ratas , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/metabolismo
6.
J Biomed Biotechnol ; 2012: 987495, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22701311

RESUMEN

Temozolomide (TMZ) has become a key therapeutic agent in patients with malignant gliomas; however, its survival benefit remains unsatisfactory. Valproic acid (VPA) has emerged as an anticancer drug via inhibition of histone deacetylases (HDACs), but the therapeutic advantages of a combination with VPA and TMZ remain poorly understood. The main aim of the present study was to determine whether an antitumor effect could be potentiated by a combination of VPA and TMZ, especially in TMZ-resistant cell lines. A combination of VPA and TMZ had a significantly enhanced antitumor effect in TMZ-resistant malignant glioma cells (T98 and U138). This enhanced antitumor effect correlated with VPA-mediated reduced O6-methylguanine-DNA methyltransferase (MGMT) expression, which plays an important role in cellular resistance to alkylating agents. In vitro, the combination of these drugs enhanced the apoptotic and autophagic cell death, as well as suppressed the migratory activities in TMZ-resistant cell lines. Furthermore, in vivo efficacy experiment showed that treatment of combination of VPA and TMZ significantly inhibited tumor growth compared with the monotherapy groups of mice. These results suggest that the clinical efficacy of TMZ chemotherapy in TMZ-resistant malignant glioma may be improved by combination with VPA.


Asunto(s)
Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Dacarbazina/análogos & derivados , Glioma/tratamiento farmacológico , Proteínas Supresoras de Tumor/genética , Ácido Valproico/administración & dosificación , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Metilasas de Modificación del ADN/biosíntesis , Enzimas Reparadoras del ADN/biosíntesis , Dacarbazina/administración & dosificación , Regulación hacia Abajo , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Humanos , Ratones , Temozolomida , Proteínas Supresoras de Tumor/biosíntesis , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Biochem Biophys Res Commun ; 407(4): 741-6, 2011 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-21439934

RESUMEN

In this study, we showed that knocking-down interleukin-8 (IL-8) in glioma cells, or its receptor, CXC chemokine receptor 1 (CXCR1) in hUCB-MSCs reduced hUCB-MSC migration toward glioma cells in a Transwell chamber. In contrast, CXCR1-transfected hUCB-MSCs (CXCR1-MSCs) showed a superior capacity to migrate toward glioma cells in a Transwell chamber compared to primary hUCB-MSCs. Furthermore, these transfected cells also demonstrated the same ability to migrate toward tumors in mice bearing intracranial human gliomas as shown by histological and in vivo imaging analysis. Our findings indicate that overexpression of CXCR1 could be a useful tool for MSC-based gene therapy to achieve a sufficient quantity of therapeutic MSCs that are localized within tumors.


Asunto(s)
Neoplasias Encefálicas/terapia , Movimiento Celular , Terapia Genética/métodos , Glioma/terapia , Células Madre Mesenquimatosas/fisiología , Receptores de Interleucina-8A/genética , Animales , Línea Celular Tumoral , Sangre Fetal/citología , Técnicas de Silenciamiento del Gen , Humanos , Interleucina-8/genética , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Desnudos , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Biochem Biophys Res Commun ; 398(1): 105-10, 2010 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-20558135

RESUMEN

Human mesenchymal stem cells (hMSCs) have been used for cell-based therapies in degenerative disease and as vehicles for delivering therapeutic genes to sites of injury and tumors. Recently, umbilical cord blood (UCB) was identified as a source for MSCs, and human UCB-derived MSCs (hUCB-MSCs) can serve as an alternative source of bone marrow-derived mesenchymal stem cells (BM-MSCs). However, migration signaling pathways required for homing and recruitment of hUCB-MSCs are not fully understood. Stromal cell-derived factor-1 (SDF-1), a ligand for the CXCR4 chemokine receptor, plays a pivotal role in mobilization and homing of stem cells and modulates different biological responses in various stem cells. In this study, expression of CXCR4 in hUCB-MSCs was studied by western blot analysis and the functional role of SDF-1 was assessed. SDF-1 induced the migration of hUCB-MSCs in a dose-dependent manner. The induced migration was inhibited by the CXCR4-specific peptide antagonist (AMD3100) and by inhibitors of phosphoinositide 3-kinase (LY294002), mitogen-activated protein kinase/extracellular signal related kinase (PD98059) and p38MAPK inhibitor (SB203580). hUCB-MSCs treated with SDF-1 displayed increased phosphorylation of Akt, ERK and p38, which were inhibited by AMD3100. Small-interfering RNA-mediated knock-down of Akt, ERK and p38 blocked SDF-1 induced hUCB-MSC migration. In addition, SDF-1-induced actin polymerization was also blocked by these inhibitors. Taken together, these results demonstrate that Akt, ERK and p38 signal transduction pathways may be involved in SDF-1-mediated migration of hUCB-MSCs.


Asunto(s)
Movimiento Celular , Quimiocina CXCL12/fisiología , Células Madre Mesenquimatosas/fisiología , Receptores CXCR4/fisiología , Cordón Umbilical/citología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Células del Estroma/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
BMC Biotechnol ; 10: 38, 2010 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-20462460

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSCs) are an attractive source of adult stem cells for therapeutic application in clinical study. Genetic modification of MSCs with beneficial genes makes them more effective for therapeutic use. However, it is difficult to transduce genes into MSCs by common transfection methods, especially nonviral methods. In this study, we applied microporation technology as a novel electroporation technique to introduce enhanced green fluorescent protein (EGFP) and brain-derived neurotropfic factor (BDNF) plasmid DNA into human umbilical cord blood-derived MSCs (hUCB-MSCs) with significant efficiency, and investigated the stem cell potentiality of engineered MSCs through their phenotypes, proliferative capacity, ability to differentiate into multiple lineages, and migration ability towards malignant glioma cells. RESULTS: Using microporation with EGFP as a reporter gene, hUCB-MSCs were transfected with higher efficiency (83%) and only minimal cell damage than when conventional liposome-based reagent (<20%) or established electroporation methods were used (30-40%). More importantly, microporation did not affect the immunophenotype of hUCB-MSCs, their proliferation activity, ability to differentiate into mesodermal and ectodermal lineages, or migration ability towards cancer cells. In addition, the BDNF gene could be successfully transfected into hUCB-MSCs, and BDNF expression remained fairly constant for the first 2 weeks in vitro and in vivo. Moreover, microporation of BDNF gene into hUCB-MSCs promoted their in vitro differentiation into neural cells. CONCLUSION: Taken together, the present data demonstrates the value of microporation as an efficient means of transfection of MSCs without changing their multiple properties. Gene delivery by microporation may enhance the feasibility of transgenic stem cell therapy.


Asunto(s)
Electroporación/métodos , Células Madre Mesenquimatosas/metabolismo , Transfección/métodos , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Sangre Fetal/citología , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Humanos , Masculino , Plásmidos , Ratas , Ratas Sprague-Dawley
10.
J Neurosci Res ; 86(10): 2168-78, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18438930

RESUMEN

Brain-derived neurotrophic factor (BDNF) plays an important role in the differentiation, development, and survival of neural stem cells. In this study, we analyzed its effects on the stimulation of human umbilical cord blood-derived mesenchymal stem cells in terms of their potential to differentiate into neuron-like cells, their survival characteristics, and the molecular mechanisms involved. The treatment of cells with neural induction medium (NIM) and BDNF generated more cells that were neuron-like and produced stronger expression of neural-lineage markers than cells treated with NIM and without BDNF. Raf-1 and ERK phosphorylation and p35 expression levels increased significantly in cells treated with both NIM and BDNF. This treatment also effectively blocked cell death following neural induction and increased Akt phosphorylation and Bcl2 expression compared with cells treated with NIM without BDNF. Inhibition of ERKs inhibited the BDNF-stimulated up-regulation of p35 and Bcl2. In addition, the inhibition of PI3K abrogated Akt phosphorylation and Bcl2 expression, but not p35 expression. Thus, MAPK/ERK-dependent p35 up-regulation and MAPK/ERK-dependent and PI3K/Akt-dependent Bcl2 up-regulation contribute to BDNF-stimulated neural differentiation and to the survival of differentiated cells.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Diferenciación Celular/fisiología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Neuronas/citología , Transducción de Señal/fisiología , Apoptosis/fisiología , Western Blotting , Supervivencia Celular/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sangre Fetal/citología , Técnica del Anticuerpo Fluorescente , Expresión Génica/fisiología , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Regulación hacia Arriba
11.
J Histochem Cytochem ; 55(8): 821-30, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17409380

RESUMEN

To investigate the possible role of vascular endothelial growth factor (VEGF) in the injured spinal cord, we analyzed the distribution and time course of the two tyrosine kinase receptors for VEGF, Flt-1 and Flk-1, in the rat spinal cord following contusion injury using a weight-drop impactor. The semi-quantitative RT-PCR analysis of Flt-1 and Flk-1 in the spinal cord showed slight upregulation of these receptors following spinal cord injury. Although mRNAs for Flt-1 and Flk-1 were constitutively expressed in neurons, vascular endothelial cells, and some astrocytes in laminectomy control rats, their upregulation was induced in association with microglia/macrophages and reactive astrocytes in the vicinity of the lesion within 1 day in rats with a contusion injury and persisted for at least 14 days. The spatiotemporal expression of Flt-1 in the contused spinal cord mirrored that of Flk-1 expression. In the early phase of spinal cord injury, upregulation of Flt-1 and Flk-1 mRNA occurred in microglia/macrophages that infiltrated the lesion. In addition, the expression of both receptors increased progressively in reactive astrocytes within the vicinity of the lesion, predominately in the white matter, and almost all reactive astrocytes coexpressed Flt-1 or Flk-1 and nestin. These results suggest that VEGF may be involved in the inflammatory response and the astroglial reaction to contusion injuries of the spinal cord via specific VEGF receptors.


Asunto(s)
Traumatismos de la Médula Espinal/metabolismo , Médula Espinal/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Enfermedad Aguda , Animales , Hibridación in Situ , Masculino , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
12.
Opt Express ; 15(20): 12866-71, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19550554

RESUMEN

We discuss bending properties of a long-period fiber grating (LPFG) inscribed into a holey fiber (HF) depending on an axial rotation angle. High quality of the HF-based LPFG with a high extinction ratio of more than 20 dB is achieved. The proposed HF-based LPFG has bending insensitivity under a certain range of the bending curvature. As the bending curvature is higher than 4 m(-1), the center wavelength of the grating is shifted into the shorter wavelength. Bending sensitivity of the HF-based LPFG is changed by an axial rotation angle, which shows its dependence on the rotational orientation. We measure the transmission characteristics of the HF-based LPFG with the ambient index change. The HF-based LPFG has ambient index insensitivity because of the air holes in the inner cladding.

13.
Int J Nanomedicine ; 11: 13-23, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26719691

RESUMEN

Mesenchymal stem cell (MSC)-based gene therapy is a promising tool for the treatment of various neurological diseases, including brain tumors. However, the tracking of in vivo stem cell migration, distribution, and survival need to be defined for their clinical application. The systemic routes of stem cell delivery must be determined because direct intracerebral injection as a cure for brain tumors is an invasive method. In this study, we show for the first time that near-infrared (NIR) imaging can reveal the distribution and tumor tropism of intravenously injected MSCs in an intracranial xenograft glioma model. MSCs were labeled with NIR fluorescent nanoparticles, and the effects of the NIR dye on cell proliferation and migratory capacity were evaluated in vitro. We investigated the tumor-targeting properties and tissue distribution of labeled MSCs introduced by intravenous injection and followed by in vivo imaging analysis, histological analysis, and real-time quantitative polymerase chain reaction. We observed no cytotoxicity or change in the overall growth rate and characteristics of labeled MSCs compared with control MSCs. NIR fluorescent imaging showed the organ distribution and targeted tumor tropism of systemically injected human MSCs. A significant number of MSCs accumulated specifically at the tumor site in the mouse brain. These results suggest that NIR-based cell tracking is a potentially useful imaging technique to visualize cell survival, migration, and distribution for the application of MSC-mediated therapies in the treatment of malignant gliomas.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , Mediciones Luminiscentes/métodos , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas , Adulto , Animales , Neoplasias Encefálicas/terapia , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Colorantes Fluorescentes/química , Colorantes Fluorescentes/farmacocinética , Glioma/terapia , Humanos , Masculino , Células Madre Mesenquimatosas/química , Ratones Desnudos , Persona de Mediana Edad , Nanopartículas/análisis , Distribución Tisular , Tropismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Neuroimmunol ; 274(1-2): 20-7, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25005115

RESUMEN

We previously demonstrated that interferon ß (IFN-ß)-secreting mesenchymal stem cells (MSCs-IFN-ß) strongly reduced the clinical severity of experimental autoimmune encephalomyelitis (EAE), compared with MSCs alone. Recently, minocycline ameliorates the clinical severity of multiple sclerosis (MS). Herein, we evaluated the effects of a combined treatment of MSCs-IFN-ß and minocycline on EAE mice. The combined treatment significantly alleviated the clinical severity mainly by maintaining the integrity of blood-spinal cord barrier, in a manner likely involving inhibition of microvascular disruption, matrix metalloproteinases, neuroinflammation, and enhancement of immunomodulatory effects. Therefore, this combined treatment has the potential to improve the functional recovery of patients with MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Interferón beta/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Minociclina/farmacología , Animales , Antibacterianos/farmacología , Células Cultivadas , Terapia Combinada , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Humanos , Interferón beta/inmunología , Metaloproteinasa 2 de la Matriz/inmunología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/inmunología , Metaloproteinasa 9 de la Matriz/metabolismo , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C57BL , Médula Espinal/inmunología , Médula Espinal/metabolismo , Células TH1/inmunología , Células Th2/inmunología
15.
Biomed Res Int ; 2014: 129145, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24672780

RESUMEN

Numerous studies have reported that mesenchymal stem cells (MSCs) can ameliorate neurological deficits in ischemic stroke models. Among the various hypotheses that have been suggested to explain the therapeutic mechanism underlying these observations, neurogenesis is thought to be critical. To enhance the therapeutic benefits of human bone marrow-derived MSCs (hBM-MSCs), we efficiently modified hBM-MSCs by introduction of the brain-derived neurotrophic factor (BDNF) gene via adenoviral transduction mediated by cell-permeable peptides and investigated whether BDNF-modified hBM-MSCs (MSCs-BDNF) contributed to functional recovery and endogenous neurogenesis in a rat model of middle cerebral artery occlusion (MCAO). Transplantation of MSCs induced the proliferation of 5-bromo-2'-deoxyuridine (BrdU-) positive cells in the subventricular zone. Transplantation of MSCs-BDNF enhanced the proliferation of endogenous neural stem cells more significantly, while suppressing cell death. Newborn cells differentiated into doublecortin (DCX-) positive neuroblasts and Neuronal Nuclei (NeuN-) positive mature neurons in the subventricular zone and ischemic boundary at higher rates in animals with MSCs-BDNF compared with treatment using solely phosphate buffered saline (PBS) or MSCs. Triphenyltetrazolium chloride staining and behavioral analysis revealed greater functional recovery in animals with MSCs-BDNF compared with the other groups. MSCs-BDNF exhibited effective therapeutic potential by protecting cell from apoptotic death and enhancing endogenous neurogenesis.


Asunto(s)
Isquemia Encefálica/terapia , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Neurogénesis , Accidente Cerebrovascular/terapia , Adulto , Animales , Apoptosis , Isquemia Encefálica/complicaciones , Isquemia Encefálica/fisiopatología , Células Cultivadas , Modelos Animales de Enfermedad , Proteína Doblecortina , Ensayo de Inmunoadsorción Enzimática , Humanos , Etiquetado Corte-Fin in Situ , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/fisiopatología , Infarto de la Arteria Cerebral Media/terapia , Masculino , Células Madre Mesenquimatosas/citología , Persona de Mediana Edad , Ratas Sprague-Dawley , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/fisiopatología , Adulto Joven
16.
Stem Cells Transl Med ; 3(2): 172-82, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24436439

RESUMEN

Because the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) selectively kills tumor cells, it is one of the most promising candidates for cancer treatment. TRAIL-secreting human mesenchymal stem cells (MSC-TRAIL) provide targeted and prolonged delivery of TRAIL in glioma therapy. However, acquired resistance to TRAIL of glioma cells is a major problem to be overcome. We showed a potential therapy that used MSC-TRAIL combined with the chemotherapeutic agent temozolomide (TMZ). The antitumor effects of the combination with MSC-TRAIL and TMZ on human glioma cells were determined by using an in vitro coculture system and an in vivo experimental xenografted mouse model. Intracellular signaling events that are responsible for the TMZ-mediated sensitization to TRAIL-induced apoptosis were also evaluated. Treatment of either TRAIL-sensitive or -resistant human glioma cells with TMZ and MSC-TRAIL resulted in a significant enhancement of apoptosis compared with the administration of each agent alone. We demonstrated that TMZ effectively increased the sensitivity to TRAIL-induced apoptosis via extracellular signal-regulated kinase-mediated upregulation of the death receptor 5 and downregulation of antiapoptotic proteins, such as X-linked inhibitor of apoptosis protein and cellular FLICE-inhibitory protein. Subsequently, this combined treatment resulted in a substantial increase in caspase activation. Furthermore, in vivo survival experiments and bioluminescence imaging analyses showed that treatment using MSC-TRAIL combined with TMZ had greater therapeutic efficacy than did single-agent treatments. These results suggest that the combination of clinically relevant TMZ and MSC-TRAIL is a potential therapeutic strategy for improving the treatment of malignant gliomas.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Dacarbazina/análogos & derivados , Glioma/tratamiento farmacológico , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Adulto , Animales , Antineoplásicos Alquilantes/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Dacarbazina/farmacología , Terapia Genética/métodos , Glioma/patología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Desnudos , Persona de Mediana Edad , Temozolomida , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven
17.
Biomed Res Int ; 2013: 696738, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23710456

RESUMEN

Interferon-beta (IFN- ß ), a well-established standard treatment for multiple sclerosis (MS), has proved to exhibit clinical efficacy. In this study, we first evaluated the therapeutic effects for MS using human bone marrow-derived mesenchymal stem cells (hBM-MSCs) as delivery vehicles with lesion-targeting capability and IFN- ß as therapeutic gene. We also engineered hBM-MSCs to secret IFN- ß (MSCs-IFN ß ) via adenoviral transduction and confirmed the secretory capacity of MSCs-IFN ß by an ELISA assay. MSCs-IFN ß -treated mice showed inhibition of experimental autoimmune encephalomyelitis (EAE) onset, and the maximum and average score for all animals in each group was significantly lower in the MSCs-IFN ß -treated EAE mice when compared with the MSCs-GFP-treated EAE mice. Inflammatory infiltration and demyelination in the lumbar spinal cord also significantly decreased in the MSCs-IFN ß -treated EAE mice compared to PBS- or MSCs-GFP-treated EAE mice. Moreover, MSCs-IFN ß treatment enhanced the immunomodulatory effects, which suppressed proinflammatory cytokines (IFN-γ and TNF-α) and conversely increased anti-inflammatory cytokines (IL-4 and IL-10). Importantly, injected MSCs-IFN ß migrated into inflamed CNS and significantly reduced further injury of blood-brain barrier (BBB) permeability in EAE mice. Thus, our results provide the rationale for designing novel experimental protocols to enhance the therapeutic effects for MS using hBM-MSCs as an effective gene vehicle to deliver the therapeutic cytokines.


Asunto(s)
Terapia Genética , Interferón beta/metabolismo , Células Madre Mesenquimatosas/metabolismo , Esclerosis Múltiple/tratamiento farmacológico , Animales , Células de la Médula Ósea/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Interferón beta/administración & dosificación , Interferón beta/genética , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Ratones , Esclerosis Múltiple/genética , Esclerosis Múltiple/patología , Factor de Necrosis Tumoral alfa/metabolismo
18.
Stem Cell Res Ther ; 4(4): 77, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23826999

RESUMEN

INTRODUCTION: Multiple sclerosis (MS) is the most common inflammatory demyelinating disorder of the central nervous system (CNS). Minocycline ameliorates the clinical severity of MS and exhibits antiinflammatory, neuroprotective activities, and good tolerance for long-term use, whereas it is toxic to the CNS. Recently, the immunomodulation and neuroprotection capabilities of human bone marrow mesenchymal stem cells (hBM-MSCs) were shown in experimental autoimmune encephalomyelitis (EAE). In this study, we evaluated whether the combination of hBM-MSCs and a low-dose minocycline could produce beneficial effects in EAE mice. METHODS: The sensitivity of hBM-MSCs to minocycline was determined by an established cell-viability assay. Minocycline-treated hBM-MSCs were also characterized with flow cytometry by using MSC surface markers and analyzed for their multiple differentiation capacities. EAE was induced in C57BL/6 mice by using immunization with MOG35-55. Immunopathology assays were used to detect the inflammatory cells, demyelination, and neuroprotection. Interferon gamma (IFN-γ)/tumor necrosis factor alpha (TNF-α) and interleukin-4 (IL-4)/interleukin-10 (IL-10), the hallmark cytokines that direct Th1 and Th2 development, were detected with enzyme-linked immunosorbent assay (ELISA). terminal dUTP nick-end labeling (TUNEL) staining was performed to elucidate the cell apoptosis in the spinal cords of EAE mice. RESULTS: Minocycline did not affect the viability, surface phenotypes, or differentiation capacity of hBM-MSCs, while minocycline affected the viability of astrocytes at a high dose. In vivo efficacy experiments showed that combined treatment, compared to the use of minocycline or hBM-MSCs alone, resulted in a significant reduction in clinical scores, along with attenuation of inflammation, demyelination, and neurodegeneration. Moreover, the combined treatment with hBM-MSCs and minocycline enhanced the immunomodulatory effects, which suppressed proinflammatory cytokines (IFN-γ, TNF-α) and conversely increased anti-inflammatory cytokines (IL-4, IL-10). In addition, TUNEL staining also demonstrated a significant decrease of the number of apoptotic cells in the combined treatment compared with either treatment alone. CONCLUSIONS: The combination of hBM-MSCs and minocycline provides a novel experimental protocol to enhance the therapeutic effects in MS.


Asunto(s)
Antibacterianos/uso terapéutico , Células de la Médula Ósea/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Minociclina/uso terapéutico , Animales , Apoptosis , Diferenciación Celular , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Humanos , Ratones , Ratones Endogámicos C57BL
19.
Cancer Res ; 72(18): 4807-17, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22962275

RESUMEN

The apoptotic ligand TRAIL is believed to have promise as a cancer gene therapy, yet many types of cancer, including gliomas, have exhibited resistance to TRAIL-induced apoptosis. Here, we show that therapeutic combination of the lipoxygenase inhibitor MK886 and TRAIL-secreting human mesenchymal stem cells (MSC-TRAIL) provide targeted and prolonged delivery of TRAIL both in vitro and in orthotopic mouse models of glioma. Treatment of either TRAIL-sensitive or TRAIL-resistant human glioma cells with MK886 and MSC-TRAIL resulted in significantly enhanced apoptosis compared with each agent alone. MK886 effectively increased the sensitivity to TRAIL-induced apoptosis via upregulation of the death receptor 5 and downregulation of the antiapoptotic protein survivin in human glioma cell lines and in primary glioma cells. This regulation was accompanied by a substantial increase in caspase activation after combined treatment. Furthermore, in vivo survival experiments and imaging analysis in orthotopic xenografted mice showed that MSC-based TRAIL gene delivery combined with MK886 into the tumors had greater therapeutic efficacy than single-agent treatment. Together, our findings indicate that MK886 combined with MSC-based TRAIL gene delivery may represent a novel strategy for improving the treatment of malignant gliomas.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Terapia Genética/métodos , Glioma/tratamiento farmacológico , Indoles/administración & dosificación , Trasplante de Células Madre Mesenquimatosas/métodos , Animales , Western Blotting , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Terapia Combinada , Citometría de Flujo , Glioma/metabolismo , Humanos , Inmunohistoquímica , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Desnudos , ARN Interferente Pequeño , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Food Chem Toxicol ; 50(9): 2987-95, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22705330

RESUMEN

Angelicae Gigantis (AG) Radix, commonly used medicinal food, has been reported as a promising candidate for inflammatory diseases. However, the anti-allergic effects of AG and its molecular mechanisms have yet to be clarified. The present study investigated the anti-allergy effects of ethanol extracts of AG on mast cell-mediated allergic inflammation in vivo and in vitro. The finding of this study demonstrated that AG reduced anti-dinitrophenyl IgE antibody-induced passive cutaneous anaphylaxis, compound 48/80-induced histamine release, 2,4-dinitrofluoro benzene-induced contact hypersensitivity. In addition, AG inhibited the production of interleukin (IL)-6, IL-8, and TNF-α, as well as the activation of Jun N-terminal kinase and nuclear factor-κB in phorbol 12-myristate 13-acetate plus calcium ionophore A23187-stimulated human mast cells. In conclusion, our results provide a novel insight into the pharmacological actions of AG as a potential candidate for use in allergic inflammatory diseases.


Asunto(s)
Angelica/química , Hipersensibilidad/prevención & control , Inflamación/prevención & control , Mastocitos/efectos de los fármacos , Extractos Vegetales/farmacología , Animales , Cromatografía Líquida de Alta Presión , Femenino , Liberación de Histamina/efectos de los fármacos , Hipersensibilidad/inmunología , Técnicas In Vitro , Inflamación/inmunología , Mastocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA