Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ScientificWorldJournal ; 2021: 7594092, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33628141

RESUMO

Red Ganoderma lucidum (G. lucidum) is a popular medicinal herb commonly used in Vietnamese traditional remedies due to its potential value for health. In this study, polysaccharides were extracted from G. lucidum using ultrasound-assisted enzymatic extraction method. The response surface methodology and Box-Behnken design were employed to investigate the effects of pH, extraction temperature, extraction time, and ultrasonic power on the content of polysaccharides. Based on ultraviolet-visible spectroscopy analysis, the highest content of polysaccharides in the extract was 32.08 mg/g under optimum experimental parameters including enzyme concentration of 3%, pH of 5.5, extraction temperature of 45°C, extraction time of 30 min, and ultrasonic power of 480 W. The Fourier-transform infrared spectroscopy was also used to identify the functional groups in the extracts. The molecular weights of polysaccharides were determined by gel permeation chromatography. The obtained extract was then evaluated for anticancer activities by using (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, showing the anticancer activities with the half-maximal inhibitory concentration value of more than 512 µg/mL. This result suggested that UAEE could be considered as an appropriate and effective extraction method for bioactive crude polysaccharides from G. lucidum.


Assuntos
Antioxidantes/farmacologia , Extratos Vegetais/farmacologia , Polissacarídeos/isolamento & purificação , Reishi/química , Antioxidantes/química , Antioxidantes/isolamento & purificação , Povo Asiático , Humanos , Neoplasias/tratamento farmacológico , Extratos Vegetais/química , Polissacarídeos/química , Polissacarídeos/farmacologia , Espectroscopia de Infravermelho com Transformada de Fourier , Propriedades de Superfície/efeitos dos fármacos , Temperatura , Ondas Ultrassônicas
3.
Stem Cells Int ; 2015: 649080, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26074975

RESUMO

Recent progress in neural stem cell- (NSC-) based tumor-targeted gene therapy showed that NSC vectors expressing an artificially engineered viral fusogenic protein, VSV-G H162R, could cause tumor cell death specifically under acidic tumor microenvironment by syncytia formation; however, the killing efficiency still had much room to improve. In the view that coexpression of another antitumoral gene with VSV-G can augment the bystander effect, a synthetic regulatory system that triggers transgene expression in a cell fusion-inducible manner has been proposed. Here we have developed a double-switch cell fusion-inducible transgene expression system (DoFIT) to drive transgene expression upon VSV-G-mediated NSC-glioma cell fusion. In this binary system, transgene expression is coregulated by a glioma-specific promoter and targeting sequences of a microRNA (miR) that is highly expressed in NSCs but lowly expressed in glioma cells. Thus, transgene expression is "switched off" by the miR in NSC vectors, but after cell fusion with glioma cells, the miR is diluted and loses its suppressive effect. Meanwhile, in the syncytia, transgene expression is "switched on" by the glioma-specific promoter. Our in vitro and in vivo experimental data show that DoFIT successfully abolishes luciferase reporter gene expression in NSC vectors but activates it specifically after VSV-G-mediated NSC-glioma cell fusion.

4.
Stem Cells Transl Med ; 3(8): 923-35, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24972599

RESUMO

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.


Assuntos
Neoplasias da Mama/terapia , Ligante de CD40/biossíntese , Células Endoteliais/transplante , Terapia Genética/métodos , Células-Tronco Pluripotentes Induzidas/transplante , Neoplasias Pulmonares/terapia , Antígeno AC133 , Animais , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Baculoviridae/genética , Baculoviridae/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ligante de CD40/genética , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular , Células Endoteliais/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Glicoproteínas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Camundongos Endogâmicos BALB C , Camundongos Nus , Peptídeos/metabolismo , Fatores de Tempo , Transdução Genética , Transfecção , Carga Tumoral
5.
Hum Gene Ther ; 25(8): 747-58, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24773154

RESUMO

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.


Assuntos
Baculoviridae/genética , Neoplasias da Mama/terapia , Ligante de CD40/genética , Células-Tronco Pluripotentes Induzidas/fisiologia , Neoplasias Pulmonares/terapia , Células-Tronco Neurais/metabolismo , Animais , Apoptose , Neoplasias da Mama/patologia , Ligante de CD40/biossíntese , Linhagem Celular Tumoral , Citocinas/metabolismo , Feminino , Expressão Gênica , Terapia Genética , Vetores Genéticos , Humanos , Neoplasias Pulmonares/secundário , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Transdução Genética
6.
Mol Ther ; 21(8): 1621-30, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23752308

RESUMO

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.


Assuntos
Glicoproteínas de Membrana/genética , Neoplasias/metabolismo , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/transplante , Proteínas do Envelope Viral/genética , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Morte Celular , Linhagem Celular Tumoral , Terapia Baseada em Transplante de Células e Tecidos , Modelos Animais de Doenças , Feminino , Genes Transgênicos Suicidas , Terapia Genética , Células Gigantes/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Células-Tronco Pluripotentes Induzidas/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Metástase Neoplásica , Neoplasias/mortalidade , Neoplasias/patologia , Neoplasias/terapia , Proteínas do Envelope Viral/metabolismo
7.
Biochem Biophys Res Commun ; 434(1): 110-6, 2013 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-23537655

RESUMO

Imaging of transduced cells and tissues is valuable in developing gene transfer vectors and evaluating gene therapy efficacy. We report here a simple method to use bright and photostable quantum dots to label baculovirus, an emerging gene therapy vector. The labeling was achieved through the non-covalent interaction of glutathione-capped CdTe quantum dots with the virus envelope, without the use of chemical conjugation. The quantum dot labeling was nondestructive to viral transduction function and enabled the identification of baculoviral vector-transduced, living cells based on red fluorescence. When the labeled baculoviral vectors were injected intravenously or intraventricularly for in vivo delivery of a transgene into mice, quantum dot fluorescence signals allow us monitor whether or not the injected tissues were transduced. More importantly, using a dual-color whole-body imaging technology, we demonstrated that in vivo viral transduction could be evaluated in a real-time manner in living mice. Thus, our method of labeling a read-to-use gene delivery vector with quantum dots could be useful towards the improvement of vector design and will have the potential to assess baculovirus-based gene therapy protocols in future.


Assuntos
Vetores Genéticos , Nucleopoliedrovírus/genética , Imagem Óptica , Pontos Quânticos , Transdução Genética , Animais , Linhagem Celular Tumoral , Feminino , Terapia Genética , Vetores Genéticos/ultraestrutura , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nucleopoliedrovírus/ultraestrutura , Ligação Proteica/genética , Transdução Genética/métodos
8.
J Neurochem ; 126(3): 318-30, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23406356

RESUMO

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.


Assuntos
Movimento Celular/fisiologia , Neoplasias , Células-Tronco Neurais/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Técnicas de Silenciamento de Genes , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos Nus , Células-Tronco Neurais/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real , Transcriptoma
9.
J Clin Invest ; 122(11): 4059-76, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23093778

RESUMO

In the human brain, microRNAs (miRNAs) from the microRNA-376 (miR-376) cluster undergo programmed "seed" sequence modifications by adenosine-to-inosine (A-to-I) editing. Emerging evidence suggests a link between impaired A-to-I editing and cancer, particularly in high-grade gliomas. We hypothesized that disruption of A-to-I editing alters expression of genes regulating glioma tumor phenotypes. By sequencing the miR-376 cluster, we show that the overall miRNA editing frequencies were reduced in human gliomas. Specifically in high-grade gliomas, miR-376a* accumulated entirely in an unedited form. Clinically, a significant correlation was found between accumulation of unedited miR-376a* and the extent of invasive tumor spread as measured by magnetic resonance imaging of patient brains. Using both in vitro and orthotopic xenograft mouse models, we demonstrated that the unedited miR-376a* promoted glioma cell migration and invasion, while the edited miR-376a* suppressed these features. The effects of the unedited miR-376a* were mediated by its sequence-dependent ability to target RAP2A and concomitant inability to target AMFR. Thus, the tumor-dependent introduction of a single base difference in the miR-376a* sequence dramatically alters the selection of its target genes and redirects its function from inhibiting to promoting glioma cell invasion. These findings uncover a new mechanism of miRNA deregulation and identify unedited miR-376a* as a potential therapeutic target in glioblastoma cells.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , MicroRNAs/metabolismo , Edição de RNA , RNA Neoplásico/metabolismo , Adenosina/genética , Adenosina/metabolismo , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Feminino , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Inosina/genética , Inosina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , Invasividade Neoplásica , Transplante de Neoplasias , RNA Neoplásico/genética , Análise de Sequência de RNA , Transplante Heterólogo
10.
Stem Cells ; 30(5): 1021-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22311724

RESUMO

Human pluripotent stem cells can serve as an accessible and reliable source for the generation of functional human cells for medical therapies. In this study, we used a conventional lentiviral transduction method to derive human-induced pluripotent stem (iPS) cells from primary human fibroblasts and then generated neural stem cells (NSCs) from the iPS cells. Using a dual-color whole-body imaging technology, we demonstrated that after tail vein injection, these human NSCs displayed a robust migratory capacity outside the central nervous system in both immunodeficient and immunocompetent mice and homed in on established orthotopic 4T1 mouse mammary tumors. To investigate whether the iPS cell-derived NSCs can be used as a cellular delivery vehicle for cancer gene therapy, the cells were transduced with a baculoviral vector containing the herpes simplex virus thymidine kinase suicide gene and injected through tail vein into 4T1 tumor-bearing mice. The transduced NSCs were effective in inhibiting the growth of the orthotopic 4T1 breast tumor and the metastatic spread of the cancer cells in the presence of ganciclovir, leading to prolonged survival of the tumor-bearing mice. The use of iPS cell-derived NSCs for cancer gene therapy bypasses the sensitive ethical issue surrounding the use of cells derived from human fetal tissues or human embryonic stem cells. This approach may also help to overcome problems associated with allogeneic transplantation of other types of human NSCs.


Assuntos
Células-Tronco Pluripotentes Induzidas/transplante , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/terapia , Células-Tronco Neurais/transplante , Transplante de Células-Tronco , Animais , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Modelos Animais de Doenças , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Metástase Neoplásica , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Especificidade de Órgãos , Transplante Heterólogo
11.
Cell Tissue Bank ; 13(2): 341-51, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21656022

RESUMO

It is known that umbilical cord blood (UCB) is a rich source of stem cells with practical and ethical advantages. Three important types of stem cells which can be harvested from umbilical cord blood and used in disease treatment are hematopoietic stem cells (HSCs), mesenchymal stem cells (MSCs) and endothelial progenitor cells (EPCs). Since these stem cells have shown enormous potential in regenerative medicine, numerous umbilical cord blood banks have been established. In this study, we examined the ability of banked UCB collected to produce three types of stem cells from the same samples with characteristics of HSCs, MSCs and EPCs. We were able to obtain homogeneous plastic rapidly-adherent cells (with characteristics of MSCs), slowly-adherent (with characteristics of EPCs) and non-adherent cells (with characteristics of HSCs) from the mononuclear cell fractions of cryopreserved UCB. Using a protocol of 48 h supernatant transferring, we successfully isolated MSCs which expressed CD13, CD44 and CD90 while CD34, CD45 and CD133 negative, had typical fibroblast-like shape, and was able to differentiate into adipocytes; EPCs which were CD34, and CD90 positive, CD13, CD44, CD45 and CD133 negative, adherent with cobble-like shape; HSCs which formed colonies when cultured in MethoCult medium.


Assuntos
Separação Celular/métodos , Sangue Fetal/citologia , Células-Tronco/citologia , Bancos de Tecidos , Adipócitos/citologia , Biomarcadores/metabolismo , Diferenciação Celular , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Citometria de Fluxo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco/metabolismo
12.
Mol Pharm ; 8(5): 1515-24, 2011 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-21755959

RESUMO

Using neural stem cells (NSCs) with tumor tropic migratory capacity to deliver therapeutic genes is an attractive strategy in eliminating metastatic or disseminated tumors. While different methods have been developed to isolate or generate NSCs, it has not been assessed whether induced pluripotent stem (iPS) cells, a type of pluripotent stem cells that hold great potential for regenerative medicine, can be used as a source for derivation of NSCs with tumor tropism. In this study, we used a conventional lentivirus transduction method to derive iPS cells from primary mouse embryonic fibroblasts and then generated NSCs from the iPS cells. To investigate whether the iPS cell derived NSCs can be used in the treatment of disseminated brain tumors, the cells were transduced with a baculoviral vector containing the herpes simplex virus thymidine kinase suicide gene and injected into the cerebral hemisphere contralateral to a tumor inoculation site in a mouse intracranial human glioma xenograft model. We observed that NSCs expressing the suicide gene were, in the presence of ganciclovir, effective in inhibiting the growth of the glioma xenografts and prolonging survival of tumor-bearing mice. Our findings provide evidence for the feasibility of using iPS cell derived NSCs as cellular vehicles for targeted anticancer gene therapy.


Assuntos
Técnicas de Transferência de Genes , Glioma/terapia , Células-Tronco Pluripotentes Induzidas/fisiologia , Células-Tronco Neurais/transplante , Animais , Antineoplásicos/uso terapêutico , Células Cultivadas , Terapia Combinada , Embrião de Mamíferos/citologia , Estudos de Viabilidade , Feminino , Genes Transgênicos Suicidas , Glioma/tratamento farmacológico , Glioma/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/virologia , Camundongos , Camundongos Endogâmicos , Camundongos Nus , Células-Tronco Neurais/citologia , Células-Tronco Neurais/virologia , Prosencéfalo/citologia , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/patologia , Simplexvirus/enzimologia , Análise de Sobrevida , Timidina Quinase/genética , Timidina Quinase/metabolismo , Timidina Quinase/uso terapêutico , Proteínas Virais/genética , Proteínas Virais/metabolismo , Proteínas Virais/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
13.
In Vitro Cell Dev Biol Anim ; 47(5-6): 368-75, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21424240

RESUMO

Dendritic cells (DCs) are the most professional antigen-presenting cells of the mammalian immune system. They are able to phagocytize, process antigen materials, and then present them to the surface of other cells including T lymphocytes in the immune system. These capabilities make DC therapy become a novel and promising immune-therapeutic approach for cancer treatment as well as for cancer vaccination. Many trials of DC therapy to treat cancers have been performed and have shown their application value. They involve harvesting monocytes or hematopoietic stem cells from a patient and processing them in the laboratory to produce DCs and then reintroduced into a patient in order to activate the immune system. DCs were successfully produced from peripheral, umbilical cord blood-derived monocytes or hematopoietic stem cells. In this research, we produced DCs from human menstrual blood-derived monocytes. Briefly, monocytes were isolated by FACS based on FSC vs. SSC plot from lysed menstrual blood. Obtained monocytes were induced into DCs by a two-step protocol. In the first step, monocytes were incubated in RPMI medium supplemented with 2% FBS, GM-CSF, and IL-4, followed by incubation in RPMI medium supplemented with α-TNF in the second step. Our data showed that induced monocytes had typical morphology of DCs, expressed HLA-DR, HLA-ABC, CD80 and CD86 markers, exhibited uptake of dextran-FITC, stimulated allogenic T cell proliferation, and released IL-12. These results demonstrated that menstrual blood can not only be a source of stromal stem cell but also DCs, which are a potential candidate for immune therapy.


Assuntos
Células Dendríticas/citologia , Ciclo Menstrual/imunologia , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Células Dendríticas/metabolismo , Feminino , Citometria de Fluxo , Humanos , Interleucina-12/imunologia , Interleucina-12/metabolismo , Ciclo Menstrual/sangue , Monócitos/citologia , Monócitos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
14.
Hum Gene Ther ; 22(11): 1365-77, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21425958

RESUMO

Mesenchymal stem cells (MSCs) possess tumor-tropic properties and consequently have been used to deliver therapeutic agents for cancer treatment. Their potential in cancer therapy highlights the need for a consistent and renewable source for the production of uniform human MSCs suitable for clinical applications. In this study, we seek to investigate whether human embryonic stem cells can be used as a cell source to fulfill this goal. We generated MSC-like cells from two human embryonic stem cell lines, HuES9 and H1, and observed that MSC-like cells derived from human embryonic stem cells were able to migrate into human glioma intracranial xenografts after being injected into the cerebral hemisphere contralateral to the tumor inoculation site. We engineered these cells with baculoviral and lentiviral vectors, respectively, for transient and stable expression of the herpes simplex virus thymidine kinase gene. In tumor-bearing mice the engineered MSC-like cells were capable of inhibiting tumor growth and prolonging survival in the presence of ganciclovir after they were injected either directly into the xenografts or into the opposite hemisphere. Our findings suggest that human embryonic stem cell-derived MSCs may be a viable and attractive alternative for large-scale derivation of targeting vehicles for cancer therapy.


Assuntos
Células-Tronco Embrionárias/citologia , Glioblastoma/terapia , Células-Tronco Mesenquimais/citologia , Pró-Fármacos/administração & dosagem , Timidina Quinase/genética , Proteínas Virais/genética , Animais , Baculoviridae/genética , Linhagem Celular Tumoral , Movimento Celular , Estudos de Viabilidade , Ganciclovir/uso terapêutico , Vetores Genéticos , Glioblastoma/genética , Herpesviridae , Humanos , Lentivirus/genética , Masculino , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...