Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Neoplasma ; 71(2): 117-122, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38766855

RESUMEN

The incidence of distant metastases is associated with most cancer-related mortalities. Extracellular vesicles (EVs), secreted from tumors and cancer-associated fibroblasts, are involved in the metastatic process mediating their organotropism through their involvement in the pre-metastatic niche formation. We have been developing suicide gene therapy mediated by EVs secreted from mesenchymal stem/ stromal cells, tumor cells, and cancer-associated fibroblasts. Suicide gene EVs conjugated with prodrug are tumor tropic, penetrate tumor cells, and kill them by intracellular conversion of nontoxic prodrug to an efficient anti-cancer drug. Here, we discuss findings regarding the possibility of using suicide gene EVs as a novel therapeutic approach for metastases, via pre-metastatic niche modification. The suicide gene EVs provide a future perspective for metastasis prevention.


Asunto(s)
Vesículas Extracelulares , Genes Transgénicos Suicidas , Metástasis de la Neoplasia , Humanos , Terapia Genética , Neoplasias/patología , Neoplasias/genética , Neoplasias/prevención & control , Profármacos/uso terapéutico , Animales , Células Madre Mesenquimatosas
2.
Mol Cell Probes ; 67: 101894, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36706931

RESUMEN

Extracellular vesicles (EVs) are nowadays a target of interest in cancer therapy as a successful drug delivering tool. Based on their many beneficial biocompatible properties are designed to transport nucleic acids, proteins, various nanomaterials or chemotherapeutics. Extracellular vesicles derived from mesenchymal stem/stromal cells (MSCs) possess their tumor-homing abilities. This inspired us to engineer the MSC's EVs to be packed with chemotherapeutic agents and deliver it as a Trojan horse directly into tumor cells. In our study, human dental pulp MSCs (DP-MSCs) were cultivated with gemcitabine (GCB), which led to its absorption by the cells and subsequent secretion of the drug out into conditioned media in EVs. Concentrated conditioned media containing small EVs (potentially exosomes) significantly inhibited the cell growth of pancreatic carcinoma cell lines in vitro. DP-MSCs were simultaneously engineered to express a suicide gene fused yeast cytosinedeaminase:uracilphosphoribosyltransferase (yCD::UPRT). The product of the suicide gene converts non-toxic prodrug 5-fluorocytosine (5-FC) to highly cytotoxic chemotherapeutic drug 5-fluorouracil (5-FU) in the recipient cancer cells. Conversion of 5-FC to 5-FU had an additional effect on cancer cell's growth inhibition. Our results showed a therapeutic potential for DP-MSC-EVs to be designed for successful delivering of chemotherapeutic drugs, together with prodrug suicide gene therapy system.


Asunto(s)
Vesículas Extracelulares , Neoplasias Pancreáticas , Profármacos , Humanos , Gemcitabina , Profármacos/metabolismo , Medios de Cultivo Condicionados , Vesículas Extracelulares/metabolismo , Línea Celular , Fluorouracilo/metabolismo , Células del Estroma , Neoplasias Pancreáticas
3.
Neoplasma ; 70(3): 333-339, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37498070

RESUMEN

In this article, we describe the gene-directed enzyme prodrug therapy, also known as the "Trojan Horse" therapy mediated by exosomes - small extracellular vesicles (sEVs) secreted from mesenchymal stem/stromal cells (MSCs) and cancer cells. MSC-EVs possess strong migrating tropism toward tumor sites. EVs derived from tumor cells mimic the parental cells in an invasive metastatic growth trait and the capability to reprogram the recipient cells. The behavior of these EVs when modified with the suicide gene predestinates them to be a drug with guided intracellular action. EVs with therapeutic suicide gene are prepared from cells with integrated retrovirus vector containing its genetic message. These EVs are internalized by tumor cells and the product of the gene converts the non-toxic prodrug into a cytotoxic drug inside the cell causing its suicide. The action of two suicide gene systems are described: the yCD::UPRT-MSC/5-FC system and the HSVTK-MSC-GCV system. Suicide gene EVs either MSCs or tumor cell origin due to their intrinsic targeting capabilities, high modification flexibility, as well as biological barrier permeability represent potential drugs for tumors untreatable with present standard cancer therapies.


Asunto(s)
Antineoplásicos , Vesículas Extracelulares , Neoplasias , Profármacos , Humanos , Profármacos/farmacología , Profármacos/uso terapéutico , Neoplasias/genética , Neoplasias/terapia , Terapia Genética
4.
Int J Mol Sci ; 24(16)2023 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-37629139

RESUMEN

Extracellular vesicles secreted from uveal melanoma (UM) cells are involved in the establishment of the premetastatic niche and display transforming potential for the formation of metastases, preferentially in the liver. In this study, we cultivated human primary UM cells and uveal melanoma-associated fibroblasts in vitro to be transduced by infection with a retrovirus containing the suicide gene-fused yeast cytosine deaminase::uracil phospho-ribosyl transferase (yCD::UPRT). A homogenous population of yCD::UPRT-UM cells with the integrated provirus expressed the gene, and we found it to continuously secrete small extracellular vesicles (sEVs) possessing mRNA of the suicide gene. The yCD::UPRT-UM-sEVs were internalized by tumor cells to the intracellular conversion of the prodrug 5-fluorocytosine (5-FC) to the cytotoxic drug 5-fluorouracil (5-FU). The host range of the yCD::UPRT-UM-sEVs was not limited to UMs only. The yCD::UPRT-UM-sEVs inhibited the growth of the human cutaneous melanoma cell line A375 and uveal melanoma cell line MP38, as well as other primary UMs, to various extents in vitro. The yCD::UPRT-UM-sEVs hold the therapeutic and prophylactic potential to become a therapeutic drug for UM. However, the use of yCD::UPRT-UM-sEVs must first be tested in animal preclinical studies.


Asunto(s)
Vesículas Extracelulares , Melanoma , Neoplasias Cutáneas , Neoplasias de la Úvea , Animales , Humanos , Melanoma/genética , Melanoma/terapia , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/terapia , Flucitosina/farmacología , Fluorouracilo
5.
Int J Mol Sci ; 23(15)2022 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-35897723

RESUMEN

Hyper-radiosensitivity (HRS) is the increased sensitivity to low doses of ionizing radiation observed in most cell lines. We previously demonstrated that HRS is permanently abolished in cells irradiated at a low dose rate (LDR), in a mechanism dependent on transforming growth factor ß3 (TGF-ß3). In this study, we aimed to elucidate the activation and receptor binding of TGF-ß3 in this mechanism. T-47D cells were pretreated with inhibitors of potential receptors and activators of TGF-ß3, along with addition of small extracellular vesicles (sEVs) from LDR primed cells, before their radiosensitivity was assessed by the clonogenic assay. The protein content of sEVs from LDR primed cells was analyzed with mass spectrometry. Our results show that sEVs contain TGF-ß3 regardless of priming status, but only sEVs from LDR primed cells remove HRS in reporter cells. Inhibition of the matrix metalloproteinase (MMP) family prevents removal of HRS, suggesting an MMP-dependent activation of TGF-ß3 in the LDR primed cells. We demonstrate a functional interaction between TGF-ß3 and activin receptor like kinase 1 (ALK1) by showing that TGF-ß3 removes HRS through ALK1 binding, independent of ALK5 and TGF-ßRII. These results are an important contribution to a more comprehensive understanding of the mechanism behind TGF-ß3 mediated removal of HRS.


Asunto(s)
Vesículas Extracelulares , Factor de Crecimiento Transformador beta3 , Línea Celular , Vesículas Extracelulares/metabolismo , Dosis de Radiación , Tolerancia a Radiación/fisiología , Factor de Crecimiento Transformador beta/metabolismo
6.
Int J Cancer ; 148(1): 128-139, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-32621791

RESUMEN

Recently, we reported about exosomes possessing messenger RNA (mRNA) of suicide gene secreted from mesenchymal stem/stromal cells (MSCs) engineered to express the suicide gene-fused yeast cytosine deaminase::uracil phosphoribosyltransferase (yCD::UPRT). The yCD::UPRT-MSC exosomes are internalized by tumor cells and intracellularly convert prodrug 5-fluorocytosine (5-FC) to cytotoxic drug 5-fluorouracil (5-FU). Human tumor cells with the potential to metastasize release exosomes involved in the creation of a premetastatic niche at the predicted organs. We found that cancer cells stably transduced with yCD::UPRT gene by retrovirus infection released exosomes acting similarly like yCD::UPRT-MSC exosomes. Different types of tumor cells were transduced with the yCD::UPRT gene. The homogenous cell population of yCD::UPRT-transduced tumor cells expressed the yCD::UPRT suicide gene and secreted continuously exosomes with suicide gene mRNA in their cargo. All tumor cell suicide gene exosomes upon internalization into the recipient tumor cells induced the cell death by intracellular conversion of 5-FC to 5-FU and to 5-FUMP in a dose-dependent manner. Most of tumor cell-derived suicide gene exosomes were tumor tropic, in 5-FC presence they killed tumor cells but did not inhibit the growth of human skin fibroblast as well as DP-MSCs. Tumor cell-derived suicide gene exosomes home to their cells of origin and hold an exciting potential to become innovative specific therapy for tumors and potentially for metastases.


Asunto(s)
Antineoplásicos/uso terapéutico , Genes Transgénicos Suicidas , Terapia Genética/métodos , Neoplasias/terapia , Profármacos/administración & dosificación , Animales , Antineoplásicos/farmacología , Ingeniería Celular/métodos , Línea Celular Tumoral , Medios de Cultivo Condicionados , Citosina Desaminasa/genética , Exosomas/genética , Flucitosina/administración & dosificación , Flucitosina/metabolismo , Fluorouracilo/metabolismo , Proteínas Fúngicas/genética , Vectores Genéticos/genética , Humanos , Ratones , Pentosiltransferasa/genética , Profármacos/metabolismo , Proteínas Recombinantes de Fusión/genética , Retroviridae/genética , Transducción Genética , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Int J Cancer ; 144(4): 897-908, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30098225

RESUMEN

The natural behavior of mesenchymal stem cells (MSCs) and their exosomes in targeting tumors is a promising approach for curative therapy. Human tumor tropic mesenchymal stem cells (MSCs) isolated from various tissues and MSCs engineered to express the yeast cytosine deaminase::uracil phosphoribosyl transferase suicide fusion gene (yCD::UPRT-MSCs) released exosomes in conditional medium (CM). Exosomes from all tissue specific yCD::UPRT-MSCs contained mRNA of the suicide gene in the exosome's cargo. When the CM was applied to tumor cells, the exosomes were internalized by recipient tumor cells and in the presence of the prodrug 5-fluorocytosine (5-FC) effectively triggered dose-dependent tumor cell death by endocytosed exosomes via an intracellular conversion of the prodrug 5-FC to 5-fluorouracil. Exosomes were found to be responsible for the tumor inhibitory activity. The presence of microRNAs in exosomes produced from naive MSCs and from suicide gene transduced MSCs did not differ significantly. MicroRNAs from yCD::UPRT-MSCs were not associated with therapeutic effect. MSC suicide gene exosomes represent a new class of tumor cell targeting drug acting intracellular with curative potential.


Asunto(s)
Exosomas/metabolismo , Genes Transgénicos Suicidas/genética , Terapia Genética/métodos , Células Madre Mesenquimatosas/metabolismo , Antimetabolitos Antineoplásicos/metabolismo , Antimetabolitos Antineoplásicos/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Citosina Desaminasa/genética , Citosina Desaminasa/metabolismo , Exosomas/genética , Flucitosina/metabolismo , Fluorouracilo/metabolismo , Fluorouracilo/farmacología , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Humanos , Pentosiltransferasa/genética , Pentosiltransferasa/metabolismo , Profármacos/metabolismo , Levaduras/genética , Levaduras/metabolismo
8.
Int J Cancer ; 134(6): 1458-65, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24038033

RESUMEN

Suicide gene therapy mediated by mesenchymal stem cells with their ability to engraft into tumors makes these therapeutic stem cells an attractive tool to activate prodrugs directly within the tumor mass. In this study, we evaluated the therapeutic efficacy of human mesenchymal stem cells derived from bone marrow and from adipose tissue, engineered to express the suicide gene cytosine deaminase::uracil phosphoribosyltransferase to treat intracerebral rat C6 glioblastoma in a simulated clinical therapeutic scenario. Intracerebrally grown glioblastoma was treated by resection and subsequently with single or repeated intracerebral inoculations of therapeutic stem cells followed by a continuous intracerebroventricular delivery of 5-fluorocytosine using an osmotic pump. Kaplan-Meier survival curves revealed that surgical resection of the tumor increased the survival time of the resected animals depending on the extent of surgical intervention. However, direct injections of therapeutic stem cells into the brain tissue surrounding the postoperative resection cavity led to a curative outcome in a significant number of treated animals. Moreover, the continuous supply of therapeutic stem cells into the brain with growing glioblastoma by osmotic pumps together with continuous prodrug delivery also proved to be therapeutically efficient. We assume that observed curative therapy of glioblastoma by stem cell-mediated prodrug gene therapy might be caused by the destruction of both tumor cells and the niche where glioblastoma initiating cells reside.


Asunto(s)
Neoplasias Encefálicas/prevención & control , Citosina Desaminasa/genética , Terapia Genética , Glioblastoma/prevención & control , Trasplante de Células Madre Mesenquimatosas , Pentosiltransferasa/genética , Profármacos/uso terapéutico , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Animales , Antimetabolitos/uso terapéutico , Médula Ósea/metabolismo , Médula Ósea/patología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Proliferación Celular , Células Cultivadas , Terapia Combinada , Flucitosina/uso terapéutico , Vectores Genéticos , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Humanos , Imagen por Resonancia Magnética , Masculino , Ratas , Ratas Sprague-Dawley
9.
Int J Cancer ; 130(10): 2455-63, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21732344

RESUMEN

Prodrug cancer gene therapy by mesenchymal stem cells (MSCs) targeted to tumors represents an attractive tool to activate prodrugs directly within the tumor mass, thus avoiding systemic toxicity. In this study, we tested the feasibility and efficacy of human adipose tissue-derived MSCs, engineered to express the suicide gene cytosine deaminase::uracil phosphoribosyltransferase to treat intracranial rat C6 glioblastoma. Experiments were designed to simulate conditions of future clinical application for high-grade glioblastoma therapy by direct injections of therapeutic stem cells into tumor. We demonstrated that genetically modified therapeutic stem cells still have the tumor tropism when injected to a distant intracranial site and effectively inhibited glioblastoma growth after 5-fluorocytosine (5-FC) therapy. Coadministration of C6 cells and therapeutic stem cells with delayed 5-FC therapy improved the survival in a therapeutic stem cell dose-dependent manner and induced complete tumor regression in a significant number of animals. Continuous intracerebroventricular delivery of 5-FC using osmotic pump reduced the dose of prodrug required for the same therapeutic effect, and along with repeated administration of therapeutic stem cells increased the survival time. Intracerebral injection of therapeutic stem cells and treatment with 5-FC did not show any detectable adverse effects. Results support the arguments to begin clinical studies for treatment of high-grade brain tumors.


Asunto(s)
Neoplasias Encefálicas/terapia , Citosina Desaminasa/genética , Terapia Genética/métodos , Glioblastoma/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Pentosiltransferasa/genética , Tejido Adiposo/citología , Animales , Línea Celular Tumoral , Terapia Combinada , Estudios de Factibilidad , Flucitosina/administración & dosificación , Genes Transgénicos Suicidas , Humanos , Masculino , Profármacos , Ratas , Ratas Sprague-Dawley , Levaduras/genética
10.
Cancers (Basel) ; 14(3)2022 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-35159002

RESUMEN

MSC-driven, gene-directed enzyme prodrug therapy (GDEPT) mediated by extracellular vesicles (EV) represents a new paradigm-cell-free GDEPT tumor therapy. In this study, we tested the efficacy of yeast cytosine deaminase::uracilphosphoribosyl transferase (yCD::UPRT-MSC)-exosomes, in the form of conditioned medium (CM) to inhibit the growth of C6 glioblastoma cells both in vitro and in vivo. MSCs isolated from human adipose tissue, umbilical cord, or dental pulp engineered to express the yCD::UPRT gene secreted yCD::UPRT-MSC-exosomes that in the presence of the prodrug 5-fluorocytosine (5-FC), inhibited the growth of rat C6 glioblastoma cells and human primary glioblastoma cells in vitro in a dose-dependent manner. CM from these cells injected repeatedly either intraperitoneally (i.p.) or subcutaneously (s.c.), applied intranasally (i.n.), or infused continuously by an ALZET osmotic pump, inhibited the growth of cerebral C6 glioblastomas in rats. A significant number of rats were cured when CM containing yCD::UPRT-MSC-exosomes conjugated with 5-FC was repeatedly injected i.p. or applied i.n. Cured rats were subsequently resistant to challenges with higher doses of C6 cells. Our data have shown that cell-free GDEPT tumor therapy mediated by the yCD::UPRT-MSC suicide gene EVs for high-grade glioblastomas represents a safer and more practical approach that is worthy of further investigation.

11.
Mol Pharm ; 8(5): 1480-7, 2011 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-21755953

RESUMEN

The attractiveness of prodrug cancer gene therapy by stem cells targeted to tumors lies in activating the prodrug directly within the tumor mass, thus avoiding systemic toxicity. Suicide gene therapy using genetically engineered mesenchymal stem cells has the advantage of being safe, because prodrug administration not only eliminates tumor cells but consequently kills the more resistant therapeutic stem cells as well. This review provides an explanation of the stem cell-targeted prodrug cancer gene therapy principle, with focus on the choice of prodrug, properties of bone marrow and adipose tissue-derived mesenchymal stem and neural stem cells as well as the mechanisms of their tumor homing ability. Therapeutic achievements of the cytosine deaminase/5-fluorocytosine prodrug system and Herpes simplex virus thymidine kinase/ganciclovir are discussed. In addition, delivery of immunostimulatory cytokines, apoptosis inducing genes, nanoparticles and antiangiogenic proteins by stem cells to tumors and metastases is discussed as a promising approach for antitumor therapy. Combinations of traditional, targeted and stem cell-directed gene therapy could significantly advance the treatment of cancer.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética , Neoplasias/terapia , Trasplante de Células Madre , Células Madre Adultas/enzimología , Células Madre Adultas/trasplante , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Movimiento Celular , Células Cultivadas/enzimología , Células Cultivadas/trasplante , Terapia Combinada , Citosina Desaminasa/genética , Citosina Desaminasa/metabolismo , Citosina Desaminasa/uso terapéutico , Genes Transgénicos Suicidas , Humanos , Trasplante de Células Madre Mesenquimatosas , Neoplasias/enzimología , Neoplasias/metabolismo , Células-Madre Neurales/enzimología , Células-Madre Neurales/trasplante , Profármacos/farmacocinética , Profármacos/uso terapéutico , Simplexvirus/enzimología , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Timidina Quinasa/uso terapéutico , Proteínas Virales/genética , Proteínas Virales/metabolismo , Proteínas Virales/uso terapéutico
12.
Mol Ther ; 18(1): 223-31, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19844197

RESUMEN

The ability of human adipose tissue-derived mesenchymal stem cells (AT-MSCs), engineered to express the suicide gene cytosine deaminase::uracil phosphoribosyltransferase (CD::UPRT), to convert the relatively nontoxic 5-fluorocytosine (5-FC) into the highly toxic antitumor 5-fluorouracil (5-FU) together with their ability to track and engraft into tumors and micrometastases makes these cells an attractive tool to activate prodrugs directly within the tumor mass. In this study, we tested the feasibility and efficacy of these therapeutic cells to function as cellular vehicles of prodrug-activating enzymes in prostate cancer (PC) therapy. In in vitro migration experiments we have shown that therapeutic AT-MSCs migrated to all the prostate cell lines tested. In a pilot preclinical study, we observed that coinjections of human bone metastatic PC cells along with the transduced AT-MSCs into nude mice treated with 5-FC induced a complete tumor regression in a dose dependent manner or did not even allow the establishment of the tumor. More importantly, we also demonstrated that the therapeutic cells were effective in significantly inhibiting PC tumor growth after intravenous administration that is a key requisite for any clinical application of gene-directed enzyme prodrug therapies.


Asunto(s)
Citosina Desaminasa/fisiología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Pentosiltransferasa/fisiología , Neoplasias de la Próstata/terapia , Animales , Línea Celular Tumoral , Citosina Desaminasa/genética , Flucitosina/farmacología , Fluorouracilo/farmacología , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Ratones , Ratones Desnudos , Pentosiltransferasa/genética , Neoplasias de la Próstata/inducido químicamente
13.
Mol Cancer ; 9: 129, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20509882

RESUMEN

BACKGROUND: Human mesenchymal stromal cells (MSC) hold a promise for future cell-based therapies due to their immunomodulatory properties and/or secretory activity. Nevertheless non-neoplastic tumor compartment could also originate from MSC. We aimed to show whether multipotent MSC derived from human adipose tissue (AT-MSC) could create tumor cell-protective milieu and affect tumor cell behaviour in vitro and in vivo. RESULTS: Here we have demonstrated tumor-promoting effect of AT-MSC on human melanoma A375 cells. AT-MSC coinjection mediated abrogation of tumor latency and supported subcutaneous xenotransplant growth from very low melanoma cell doses. Tumor incidence was also significantly increased by AT-MSC-derived soluble factors. AT-MSC supported proliferation, suppressed apoptosis and modulated melanoma cell responses to cytotoxic drugs in vitro. Expression and multiplex cytokine assays confirmed synergistic increase in VEGF that contributed to the AT-MSC-mediated support of A375 xenotransplant growth. Production of G-CSF and other factors implicated in formation of supportive proinflammatory tumor cell microenvironment was also confirmed. SDF-1alpha/CXCR4 signalling contributed to tumor-promoting effect of systemic AT-MSC administration on A375 xenotransplants. However, no support was observed for human glioblastoma cells 8MGBA co-injected along with AT-MSC that did not sustain tumor xenotransplant growth in vivo. Tumor-inhibiting response could be attributed to the synergistic action of multiple cytokines produced by AT-MSC on glioblastoma cells. CONCLUSIONS: Herein we provide experimental evidence for MSC-mediated protective effect on melanoma A375 cells under nutrient-limiting and hostile environmental conditions resulting from mutual crosstalk between neoplastic and non-malignant cells. This tumor-favouring effect was not observed for the glioblastoma cells 8MGBA. Collectively, our data further strengthen the need for unravelling mechanisms underlying MSC-mediated modulation of tumor behaviour for possible future MSC clinical use in the context of malignant disease.


Asunto(s)
Comunicación Celular/fisiología , Citocinas/biosíntesis , Melanoma Experimental/patología , Células Madre Mesenquimatosas , Receptor Cross-Talk/fisiología , Tejido Adiposo/citología , Animales , Línea Celular Tumoral , Técnicas de Cocultivo , Expresión Génica , Perfilación de la Expresión Génica , Glioblastoma/patología , Humanos , Melanoma Experimental/metabolismo , Ratones , Ratones Desnudos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cancers (Basel) ; 12(5)2020 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-32354013

RESUMEN

Mesenchymal stem/stromal cells (MSCs) prepared from various human tissues were stably transduced with the suicide gene herpes simplex virus thymidine kinase (HSVTK) by means of retrovirus infection. HSVTK-transduced MSCs express the suicide gene and in prodrug ganciclovir (GCV) presence induced cell death by intracellular conversion of GCV to GCV-triphosphate. The homogenous population of HSVTK-MSCs were found to release exosomes having mRNA of the suicide gene in their cargo. The exosomes were easily internalized by the tumor cells and the presence of ganciclovir caused their death in a dose-dependent manner. Efficient tumor cell killing of glioma cell lines and primary human glioblastoma cells mediated by HSVTK-MSC exosomes is reported. Exosomes produced by suicide gene transduced MSCs represent a new class of highly selective tumor cell targeted drug acting intracellular with curative potential.

15.
Cancer Res ; 67(13): 6304-13, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17616689

RESUMEN

Human adipose tissue-derived mesenchymal stem cells (AT-MSC) are considered to be a promising source of autologous stem cells in personalized cell-based therapies. Tumor tracking properties of MSC provide an attractive opportunity for targeted transgene delivery into the sites of tumor formation. In the present study, we addressed whether the suicide gene introduction into human AT-MSC could produce a tumor-specific prodrug converting cellular vehicle for targeted chemotherapy. We prepared yeast fusion cytosine deaminase::uracil phosphoribosyltransferase gene-expressing cells [cytosine deaminase (CD)-expressing AT-MSC (CD-AT-MSC)] by retrovirus transduction. We explored their therapeutic potential on a model of human colon cancer in the presence of prodrug 5-fluorocytosine (5-FC). Gene manipulation of human AT-MSC did not sensitize CD-AT-MSC to 5-FC, thus overcoming the inherent disadvantage of suicide effect on cellular vehicle. CD-AT-MSC in combination with 5-FC augmented the bystander effect and selective cytotoxicity on target tumor cells HT-29 in direct coculture in vitro. We confirmed directed migration ability of AT-MSC and CD-AT-MSC toward tumor cells HT-29 in vitro. Moreover, we achieved significant inhibition of s.c. tumor xenograft growth by s.c. or i.v. administered CD-AT-MSC in immunocompromised mice treated with 5-FC. We confirmed the ability of CD-AT-MSC to deliver the CD transgene to the site of tumor formation and mediate strong antitumor effect in vivo. Taken together, these data characterize MSC derived from adipose tissue as suitable delivery vehicles for prodrug converting gene and show their utility for a personalized cell-based targeted cancer gene therapy.


Asunto(s)
Tejido Adiposo/metabolismo , Antineoplásicos/farmacología , Flucitosina/farmacología , Regulación de la Expresión Génica , Terapia Genética/métodos , Células Madre Mesenquimatosas/citología , Neoplasias/tratamiento farmacológico , Profármacos/farmacología , Animales , Línea Celular Tumoral , Citosina Desaminasa/genética , Citosina Desaminasa/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias
16.
Methods Mol Biol ; 1895: 75-85, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30539530

RESUMEN

Exosomes derived from human mesenchymal stem cells (MSCs) engineered to express the suicide gene yeast cytosine deaminase::uracil phosphoribosyl transferase (yCD::UPRT) represent a new therapeutic approach for tumor-targeted innovative therapy. The yCD::UPRT-MSC-exosomes carry mRNA of the suicide gene in their cargo. Upon internalization by tumor cells, the exosomes inhibit the growth of broad types of cancer cells in vitro, in the presence of a prodrug. Here we describe the method leading to the production and testing of these therapeutic exosomes. The described steps include the preparation of replication-deficient retrovirus possessing the yCD::UPRT suicide gene, and the preparation and selection of MSCs transduced with yCD::UPRT suicide gene. We present procedures to obtain exosomes possessing the ability to induce the death of tumor cells. In addition, we highlight methods for the evaluation of the suicide gene activity of yCD::UPRT-MSC-exosomes.


Asunto(s)
Exosomas , Genes Transgénicos Suicidas , Terapia Genética/métodos , Neoplasias/terapia , Retroviridae/genética , Animales , Línea Celular , Citosina Desaminasa/genética , Portadores de Fármacos , Proteínas Fúngicas/genética , Vectores Genéticos , Humanos , Células Madre Mesenquimatosas , Ratones , Pentosiltransferasa/genética , Levaduras/enzimología
17.
J Gene Med ; 10(10): 1071-82, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18671316

RESUMEN

BACKGROUND: Previously, we validated capability of human adipose tissue-derived mesenchymal stem cells (AT-MSC) to serve as cellular vehicles for gene-directed enzyme prodrug molecular chemotherapy. Yeast fusion cytosine deaminase : uracil phosphoribosyltransferase expressing AT-MSC (CD y-AT-MSC) combined with systemic 5-fluorocytosine (5FC) significantly inhibited growth of human colon cancer xenografts. We aimed to determine the cytotoxic efficiency to other tumour cells both in vitro and in vivo. METHODS: CD y-AT-MSC/5FC-mediated proliferation inhibition against a panel of human tumour cells lines was evaluated in direct and indirect cocultures in vitro. Antitumour effect was tested on immunodeficient mouse model in vivo. RESULTS: Although culture expansion of CD y-AT-MSC sensitized these cells to 5FC mediated suicide effect, expanded CD y-AT-MSC/5FC still exhibited strong bystander cytotoxic effect towards human melanoma, glioblastoma, colon, breast and bladder carcinoma in vitro. Most efficient inhibition (91%) was observed in melanoma A375 cell line when directly cocultured with 2% of therapeutic cells CD y-AT-MSC/5FC. The therapeutic paradigm of the CD y -AT-MSC/5FC system was further evaluated on melanoma A375 xenografts on nude mice in vivo. Complete regression in 89% of tumours was achieved when 20% CD y-AT-MSC/5FC were co-injected along with tumour cells. More importantly, systemic CD y-AT-MSC administration resulted in therapeutic cell homing into subcutaneous melanoma and mediated tumour growth inhibition. CONCLUSIONS: CD y-AT-MSC capability of targeting subcutaneous melanoma offers a possibility to selectively produce cytotoxic agent in situ. Our data further demonstrate beneficial biological properties of AT-MSC as a cellular vehicle for enzyme/prodrug therapy approach to molecular chemotherapy.


Asunto(s)
Citosina Desaminasa/genética , Melanoma Experimental/tratamiento farmacológico , Células Madre Mesenquimatosas/enzimología , Tejido Adiposo/metabolismo , Adulto , Animales , Apoptosis , Vacunas contra el Cáncer/genética , Línea Celular Tumoral , Citosina Desaminasa/metabolismo , Flucitosina/metabolismo , Flucitosina/farmacología , Terapia Genética , Vectores Genéticos/administración & dosificación , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Pentosiltransferasa/genética , Pentosiltransferasa/metabolismo , Profármacos/farmacología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción Genética
18.
Stem Cells Int ; 2018: 8973613, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29760738

RESUMEN

Stem cells derived from human dental pulp tissue (DP-MSC) differ from the other mesenchymal stem cells prepared from bone marrow or adipose tissue due to their embryonic origin from the neural crest and are of special interest because of their neurotropic character. Furthermore, the therapeutic potential of DP-MSCs is realized through paracrine action of extracellularly released components, for which exosomes play an important role. In this review, we intend to explore the properties of these cells with an emphasis on exosomes. The therapeutic applicability of these cells and exosomes in dental practice, neurodegenerative diseases, and many other difficultly treatable diseases, like myocardial infarction, focal cerebral ischemia, acute lung or brain injury, acute respiratory distress syndrome, acute inflammation, and several others is concisely covered. The use of cellular exosomes as an important diagnostic marker and indicator of targeted cancer therapies is also discussed, while the importance of stem cells from human exfoliated deciduous teeth as a source of evolutionally young cells for future regenerative therapies is stressed. We conclude that exosomes derived from these cells are potent therapeutic tools for regenerative medicine in the near future as clinical administration of DP-MSC-conditioned medium and/or exosomes is safer and more practical than stem cells.

19.
Dose Response ; 14(4): 1559325816673901, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27867323

RESUMEN

Prior findings showed that serum from DBA/2 mice that had been given whole-body irradiation for 1 hour at a low dose rate (LDR) of 30 cGy/h induced protection against radiation in reporter cells by a mechanism depending on transforming growth factor ß3 and inducible nitric oxide synthase activity. In the present study, the effect of the 1 hour of LDR irradiation on the response of the preirradiated mice to a subsequent lethal dose and on the life span is examined. These DBA/2 mice were prime irradiated for 1 hour at 30 cGy/h. Two experiments with 9 and 9.5 Gy challenge doses given 6 weeks after priming showed increased survival in primed mice compared to unprimed mice followed up to 225 and 81 days after challenge irradiation, respectively. There was no overall significant difference in life span between primed and unprimed mice when no challenge irradiation was given. The males seemed to have a slight increase in lifespan after priming while the opposite was seen for the females.

20.
Dose Response ; 13(1)2015.
Artículo en Inglés | MEDLINE | ID: mdl-26673923

RESUMEN

Prior findings in vitro of a TGF-ß3 dependent mechanism induced by low dose-rate irradiation and resulting in increased radioresistance and removal of low dose hyper-radiosensitivity (HRS) was tested in an in vivo model. DBA/2 mice were given whole-body irradiation for 1 h at low dose-rates (LDR) of 0.3 or 0.03 Gy/h. Serum was harvested and added to RPMI (4% mouse serum and 6% bovine serum).This medium was transferred to reporter cells (T-47D breast cancer cells or T98G glioblastoma cells). The response to subsequent challenge irradiation of the reporter cells was measured by the colony assay. While serum from unirradiated control mice had no effect on the radiosensitivity in the reporter cells, serum from mice given 0.3 Gy/h or 0.03 Gy/h for 1 h removed HRS and also increased survival in response to doses up to 5 Gy. The effect lasted for at least 15 months after irradiation. TGF-ß3 neutralizer added to the medium containing mouse serum inhibited the effect. Serum from mice given irradiation of 0.3 Gy/h for 1 h and subsequently treated with iNOS inhibitor 1400W did not affect radiosensitivity in reporter cells; neither did serum from the unirradiated progeny of mice given 1h LDR whole-body irradiation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA