Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Stem Cells Transl Med ; 3(8): 923-35, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24972599

RESUMEN

Given their intrinsic ability to home to tumor sites, endothelial progenitor cells (EPCs) are attractive as cellular vehicles for targeted cancer gene therapy. However, collecting sufficient EPCs is one of the challenging issues critical for effective clinical translation of this new approach. In this study, we sought to explore whether human induced pluripotent stem (iPS) cells could be used as a reliable and accessible cell source to generate human EPCs suitable for cancer treatment. We used an embryoid body formation method to derive CD133(+)CD34(+) EPCs from human iPS cells. The generated EPCs expressed endothelial markers such as CD31, Flk1, and vascular endothelial-cadherin without expression of the CD45 hematopoietic marker. After intravenous injection, the iPS cell-derived EPCs migrated toward orthotopic and lung metastatic tumors in the mouse 4T1 breast cancer model but did not promote tumor growth and metastasis. To investigate their therapeutic potential, the EPCs were transduced with baculovirus encoding the potent T cell costimulatory molecule CD40 ligand. The systemic injection of the CD40 ligand-expressing EPCs stimulated the secretion of both tumor necrosis factor-α and interferon-γ and increased the caspase 3/7 activity in the lungs with metastatic tumors, leading to prolonged survival of the tumor bearing mice. Therefore, our findings suggest that human iPS cell-derived EPCs have the potential to serve as tumor-targeted cellular vehicles for anticancer gene therapy.


Asunto(s)
Neoplasias de la Mama/terapia , Ligando de CD40/biosíntesis , Células Endoteliales/trasplante , Terapia Genética/métodos , Células Madre Pluripotentes Inducidas/trasplante , Neoplasias Pulmonares/terapia , Antígeno AC133 , Animales , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Baculoviridae/genética , Baculoviridae/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ligando de CD40/genética , Línea Celular , Línea Celular Tumoral , Movimiento Celular , Células Endoteliales/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Glicoproteínas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Ratones Endogámicos BALB C , Ratones Desnudos , Péptidos/metabolismo , Factores de Tiempo , Transducción Genética , Transfección , Carga Tumoral
2.
Hum Gene Ther ; 25(8): 747-58, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24773154

RESUMEN

The interaction between CD40 ligand (CD40L) and CD40 can directly inhibit growth of CD40-positive carcinoma cells and may indirectly inhibit tumor growth through coordination of immune responses. Many efforts in CD40L cancer gene therapy have been focused on direct CD40L gene transfer into malignant target cells. This in vivo gene therapy approach relies on high-efficiency gene transfer and could be technically challenging for the treatment of certain cancers, especially multisite metastases. We report herein an alternative means of using the tumor-homing property of neural stem cells (NSCs) to deliver CD40L molecules into tumor tissues. NSCs were derived from human induced pluripotent stem cells, transduced in vitro with a baculoviral vector encoding CD40L, and intravenously injected into immunocompetent mice with orthotopic and metastatic breast cancers. Through a bystander mechanism of intercellular transfer of CD40L from the donor NSCs to tumor target cells, the treatment impeded tumor growth, leading to prolonged survival of the tumor-bearing mice. We further showed that compared with the stem cell-based gene therapy that employed a suicide gene, the CD40L immunogene therapy did not cause liver and kidney injury in the treated mice. This new approach may be particularly valuable for metastatic cancer treatments after systemic stem cell administration.


Asunto(s)
Baculoviridae/genética , Neoplasias de la Mama/terapia , Ligando de CD40/genética , Células Madre Pluripotentes Inducidas/fisiología , Neoplasias Pulmonares/terapia , Células-Madre Neurales/metabolismo , Animales , Apoptosis , Neoplasias de la Mama/patología , Ligando de CD40/biosíntesis , Línea Celular Tumoral , Citocinas/metabolismo , Femenino , Expresión Génica , Terapia Genética , Vectores Genéticos , Humanos , Neoplasias Pulmonares/secundario , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Transducción Genética
3.
Mol Ther ; 21(8): 1621-30, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23752308

RESUMEN

Intravenously injected neural stem cells (NSCs) can infiltrate both primary and metastatic tumor sites; thus, they are attractive tumor-targeting vehicles for delivering anticancer agents. However, because the systemic distribution of the injected NSCs involves normal organs and might induce off-target actions leading to unintended side effects, clinical applications of this approach is impeded. Given that the vesicular stomatitis virus glycoprotein (VSV-G) can promote the formation of multinucleated syncytia to kill cells in a pH-dependent manner, we engineered a pH sensor of VSV-G and generated a novel VSV-G mutant that efficiently promotes syncytium formation at the tumor extracellular pH (pHe) but not at pH 7.4. Using transduced NSCs derived from induced pluripotent stem cells (iPSCs), the VSV-G mutant was delivered into mice with metastatic breast cancers in the lung through tail vein injection. Compared with the conventional stem cell-based gene therapy that uses the herpes simplex virus thymidine kinase (HSVtk) suicide gene, this treatment did not display toxicity to normal non-targeted organs while retaining therapeutic effects in tumor-bearing organs. Our findings demonstrate the effectiveness of a new approach for achieving tumor-selective killing effects following systemic stem cell administration. Its potential in stem cell-based gene therapy for metastatic cancer is worthy of further exploration.


Asunto(s)
Glicoproteínas de Membrana/genética , Neoplasias/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/trasplante , Proteínas del Envoltorio Viral/genética , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Muerte Celular , Línea Celular Tumoral , Tratamiento Basado en Trasplante de Células y Tejidos , Modelos Animales de Enfermedad , Femenino , Genes Transgénicos Suicidas , Terapia Genética , Células Gigantes/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Células Madre Pluripotentes Inducidas/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Metástasis de la Neoplasia , Neoplasias/mortalidad , Neoplasias/patología , Neoplasias/terapia , Proteínas del Envoltorio Viral/metabolismo
4.
J Neurol Sci ; 330(1-2): 85-93, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23664653

RESUMEN

Neural stem cells (NSCs) possess immunosuppressive characteristics, but effects of NSCs on human dendritic cells (DCs), the most important antigen presenting cells, are less well studied. We used an in vitro approach to evaluate the effects of human NSCs on differentiation of human blood CD14(+) monocytes into DCs. NSCs derived from H1 human embryonic stem cells (hESC-NSCs) and human ReNcell NSC line, as well as human bone marrow derived mesenchymal stem cells (MSCs), were tested. We observed that in response to treatment with interleukin-4 and granulocyte macrophage colony-stimulating factor CD14(+) monocytes co-cultured with NSCs were able to down-regulate CD14 and up-regulate the differentiation marker CD1a, whereas MSC co-culture strongly inhibited CD1a expression and supported prolonged expression of CD14. A similar difference between NSCs and MSCs was noted when lipopolysaccharides were included to induce maturation of monocyte-derived DCs. However, when effects on the function of derived DCs were investigated, NSCs suppressed the elevation of the DC maturation marker CD83, although not the up-regulation of costimulatory molecules CD80, CD86 and CD40, and impaired the functional capacity of the derived DCs to stimulate alloreactive T cells. We did not observe any obvious difference between hESC-NSCs and ReNcell NSCs in inhibiting DC maturation and function. Our data suggest that although human NSCs are less effective than human MSCs in suppressing monocyte differentiation into DCs, these stem cells can still affect the function of DCs, ultimately regulating specific immune responses.


Asunto(s)
Diferenciación Celular/fisiología , Células Dendríticas/fisiología , Células Madre Embrionarias/fisiología , Monocitos/fisiología , Células-Madre Neurales/fisiología , Antígenos CD/biosíntesis , Antígenos CD1/biosíntesis , Antígeno B7-1/biosíntesis , Linfocitos T CD4-Positivos/fisiología , Antígenos CD40/biosíntesis , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/biosíntesis , Citometría de Flujo , Humanos , Inmunoglobulinas/biosíntesis , Inmunohistoquímica , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Glicoproteínas de Membrana/biosíntesis , Antígeno CD83
5.
J Neurochem ; 126(3): 318-30, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23406356

RESUMEN

The breakthrough in derivation of human-induced pluripotent stem cells (hiPSCs) provides an approach that may help overcome ethical and allergenic challenges posed in numerous medical applications involving human cells, including neural stem/progenitor cells (NSCs). Considering the great potential of NSCs in targeted cancer gene therapy, we investigated in this study the tumor tropism of hiPSC-derived NSCs and attempted to enhance the tropism by manipulation of biological activities of proteins that are involved in regulating the migration of NSCs toward cancer cells. We first demonstrated that hiPSC-NSCs displayed tropism for both glioblastoma cells and breast cancer cells in vitro and in vivo. We then compared gene expression profiles between migratory and non-migratory hiPSC-NSCs toward these cancer cells and observed that the gene encoding neuronal nitric oxide synthase (nNOS) was down-regulated in migratory hiPSC-NSCs. Using nNOS inhibitors and nNOS siRNAs, we demonstrated that this protein is a relevant regulator in controlling migration of hiPSC-NSCs toward cancer cells, and that inhibition of its activity or down-regulation of its expression can sensitize poorly migratory NSCs and be used to improve their tumor tropism. These findings suggest a novel application of nNOS inhibitors in neural stem cell-mediated cancer therapy.


Asunto(s)
Movimiento Celular/fisiología , Neoplasias , Células-Madre Neurales/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Animales , Línea Celular Tumoral , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Ratones Desnudos , Células-Madre Neurales/citología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcriptoma
6.
Biochimie ; 94(3): 759-67, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22133614

RESUMEN

Germ cells express a unique subset of genes called germ genes mostly encoding RNA-binding proteins such as Dazl, Dnd and Vasa. How germ gene expression is controlled remains illusive, because in no organism has a transcription factor been identified that regulate expression of these genes. Microphthalmia-associated transcription factor (Mitf) has been reported to show expression in male mouse germ cells of the adult testis. Here we report in the fish medaka (Oryzias latipes) that Mitf is a transcription activator of germ gene expression. Mitf is a master regulator of melanocyte development, which activates melanogenic genes through binding to the E-box containing consensus CANNTG. The E-box was found to be present in 23-26 copies in the promoters of medaka germ genes dazl, dnd and vasa. Importantly, forced Mitf expression enhanced the transcriptional activity of the three gene promoters by up to more than 10 fold and remarkably increased the level of endogenous dazl, dnd and vasa transcripts in cell culture. Transfection of Mitf expression vectors was sufficient to induce directed differentiation of medaka embryonic stem cells into melanocytes. Fluorescence in situ hybridization revealed the expression of both medaka mitf genes in adult germ cells of male and female gonads. Mitf is well-known as the melanocyte master regulator. Our results offer first evidence that Mitf may act as a transcriptional activator of germ gene expression in medaka.


Asunto(s)
Proteínas de Peces/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo , Oryzias/metabolismo , Animales , Proteínas de Peces/genética , Masculino , Factor de Transcripción Asociado a Microftalmía/genética , Oryzias/genética , Regiones Promotoras Genéticas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Testículo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...