Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Int J Mol Sci ; 23(17)2022 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-36077097

RESUMEN

Insulin-producing cells derived from induced pluripotent stem cells (iPSCs) are promising candidates for ß cell replacement in type 1 diabetes. However, the risk of teratoma formation due to residual undifferentiated iPSCs contaminating the differentiated cells is still a critical concern for clinical application. Here, we hypothesized that pretreatment of iPSC-derived insulin-producing cells with an anti-CD30 antibody−drug conjugate could prevent in vivo teratoma formation by selectively killing residual undifferentiated cells. CD30 is expressed in all human iPSCs clones tested by flow cytometry (n = 7) but not in iPSC-derived ß cells (ißs). Concordantly, anti-CD30 treatment in vitro for 24 h induced a dose-dependent cell death (up to 90%) in human iPSCs while it did not kill ißs nor had an impact on iß identity and function, including capacity to secrete insulin in response to stimuli. In a model of teratoma assay associated with iß transplantation, the pretreatment of cells with anti-CD30 for 24 h before the implantation into NOD-SCID mice completely eliminated teratoma development (0/10 vs. 8/8, p < 0.01). These findings suggest that short-term in vitro treatment with clinical-grade anti-CD30, targeting residual undifferentiated cells, eliminates the tumorigenicity of iPSC-derived ß cells, potentially providing enhanced safety for iPSC-based ß cell replacement therapy in clinical scenarios.


Asunto(s)
Antineoplásicos , Inmunoconjugados , Células Madre Pluripotentes Inducidas , Teratoma , Animales , Antineoplásicos/farmacología , Diferenciación Celular , Humanos , Inmunoconjugados/farmacología , Insulina/metabolismo , Antígeno Ki-1/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Teratoma/etiología , Teratoma/metabolismo , Teratoma/prevención & control
2.
JCI Insight ; 6(7)2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33830086

RESUMEN

Human pluripotent stem cells (PSCs), which are composed of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), provide an opportunity to advance cardiac cell therapy-based clinical trials. However, an important hurdle that must be overcome is the risk of teratoma formation after cell transplantation due to the proliferative capacity of residual undifferentiated PSCs in differentiation batches. To tackle this problem, we propose the use of a minimal noncardiotoxic doxorubicin dose as a purifying agent to selectively target rapidly proliferating stem cells for cell death, which will provide a purer population of terminally differentiated cardiomyocytes before cell transplantation. In this study, we determined an appropriate in vitro doxorubicin dose that (a) eliminates residual undifferentiated stem cells before cell injection to prevent teratoma formation after cell transplantation and (b) does not cause cardiotoxicity in ESC-derived cardiomyocytes (CMs) as demonstrated through contractility analysis, electrophysiology, topoisomerase activity assay, and quantification of reactive oxygen species generation. This study establishes a potentially novel method for tumorigenic-free cell therapy studies aimed at clinical applications of cardiac cell transplantation.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Doxorrubicina/administración & dosificación , Células Madre Embrionarias/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Células Madre Pluripotentes/citología , Animales , Apoptosis/efectos de los fármacos , Cardiotoxicidad/etiología , Cardiotoxicidad/prevención & control , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Tratamiento Basado en Trasplante de Células y Tejidos/efectos adversos , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Células Madre Embrionarias/trasplante , Regulación de la Expresión Génica/efectos de los fármacos , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/efectos de los fármacos , Humanos , Ratones SCID , Especies Reactivas de Oxígeno/metabolismo , Teratoma/prevención & control
3.
Nat Commun ; 11(1): 2713, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32483127

RESUMEN

Despite their rapidly-expanding therapeutic potential, human pluripotent stem cell (hPSC)-derived cell therapies continue to have serious safety risks. Transplantation of hPSC-derived cell populations into preclinical models has generated teratomas (tumors arising from undifferentiated hPSCs), unwanted tissues, and other types of adverse events. Mitigating these risks is important to increase the safety of such therapies. Here we use genome editing to engineer a general platform to improve the safety of future hPSC-derived cell transplantation therapies. Specifically, we develop hPSC lines bearing two drug-inducible safeguards, which have distinct functionalities and address separate safety concerns. In vitro administration of one small molecule depletes undifferentiated hPSCs >106-fold, thus preventing teratoma formation in vivo. Administration of a second small molecule kills all hPSC-derived cell-types, thus providing an option to eliminate the entire hPSC-derived cell product in vivo if adverse events arise. These orthogonal safety switches address major safety concerns with pluripotent cell-derived therapies.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/genética , Edición Génica/métodos , Células Madre Pluripotentes/metabolismo , Trasplante de Células Madre/métodos , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Tacrolimus/análogos & derivados , Tacrolimus/farmacología , Teratoma/genética , Teratoma/metabolismo , Teratoma/prevención & control
4.
Nutrients ; 12(3)2020 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-32182802

RESUMEN

Induced pluripotent stem cells (iPSCs) have similar properties to embryonic stem cells in terms of indefinite self-renewal and differentiation capacity. After in vitro differentiation of iPSCs, undifferentiated iPSCs (USCs) may exist in cell therapy material and can form teratomas after in vivo transplantation. Selective elimination of residual USCs is, therefore, very important. Prunellae Spica (PS) is a traditional medicinal plant that has been shown to exert anti-cancer, antioxidant, and anti-inflammatory activities; however, its effects on iPSCs have not been previously characterized. In this study, we find that ethanol extract of PS (EPS) effectively induces apoptotic cell death of USCs through G2/M cell cycle arrest, generation of intracellular reactive oxygen species, alteration of mitochondrial membrane potentials, and caspase activation of USCs. In addition, EPS increases p53 accumulation and expression of its downstream targets. In p53 knockout (KO) iPSCs, the EPS did not induce apoptosis, indicating that EPS-mediated apoptosis of USCs was p53-dependent. In addition, EPS was not genotoxic towards iPSCs-derived differentiated cells. EPS treatment before injection efficiently prevented in ovo teratoma formation of p53 wild-type (WT) iPSCs but not p53KO iPSCs. Collectively, these results indicate that EPS has potent anti-teratoma activity and no genotoxicity to differentiated cells. It can, therefore, be used in the development of safe and efficient iPSC-based cell therapies.


Asunto(s)
Apoptosis/efectos de los fármacos , Extractos Vegetales/farmacología , Células Madre Pluripotentes/efectos de los fármacos , Prunella/química , Teratoma/prevención & control , Proteína p53 Supresora de Tumor/metabolismo , Células Cultivadas , Humanos
5.
Phytomedicine ; 69: 153198, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32151917

RESUMEN

BACKGROUND: Induced pluripotent stem cells (iPSCs) are regarded as the best potential cell source for cell-based regenerative medicine. To develop a safe and efficient iPSC-based cell therapy, it is very important to avoid possible teratoma formation, which can arise from undifferentiated iPSCs (USCs) remaining among differentiated cell products. Dried bark of Magnolia officinalis (Magnolia cortex, MC) has long been used in traditional medicine to treat gastrointestinal ailments and allergic diseases, and has shown have various pharmacological activities, including anti-bacterial, anti-inflammatory, and anti-cancer effects. However, its effects on iPSCs have not yet been examined. PURPOSE: In this study, we investigated the selective cytotoxic effects of ethanol extract of MC (EEMC) on undifferentiated iPSCs and elucidated the underlying apoptotic mechanisms in detail. We also investigated the inhibitory effects of EEMC on teratoma formation via in ovo experiments. RESULTS: We found that EEMC greatly reduced cell growth and induced apoptotic cell death in USCs, but not in differentiated or normal cells. EEMC caused G2/M cell cycle arrest, mitochondrial damage, and caspase activation of USCs, accompanied by p53 accumulation. In p53KO human iPSCs, EEMC had no cytotoxicity, reinforcing that EEMC-mediated apoptosis of USCs is p53-dependent. EEMC did not cause DNA damage in iPSC-derived differentiated cells. In ovo teratoma formation assay revealed that EEMC treatment before injection efficiently eliminated USCs and prevented teratoma formation. CONCLUSIONS: These results collectively indicate that EEMC has potent anti-teratoma activity, and therefore can be used for the development of safe iPSC-based therapy.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Magnolia/química , Extractos Vegetales/farmacología , Teratoma/prevención & control , Animales , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Embrión de Pollo , Etanol/química , Hepatocitos/citología , Hepatocitos/fisiología , Humanos , Células Madre Pluripotentes Inducidas/patología , Células Madre Pluripotentes Inducidas/fisiología , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Extractos Vegetales/química , Teratoma/patología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
6.
FEBS J ; 287(21): 4783-4800, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32056377

RESUMEN

Antiepileptic/teratogen valproate (VPA) is a histone deacetylase inhibitor/epigenetic drug proposed for the antitumor therapy where it is generally crucial to target poorly or undifferentiated cells to prevent a recurrence. Transplanted rodent gastrulating embryos-proper (primitive streak and three germ layers) are the source of teratoma/teratocarcinoma tumors. Human primitive-streak remnants develop sacrococcygeal teratomas that may recur even when benign (well differentiated). To screen for unknown VPA impact on teratoma-type tumors, we used original 2-week embryo-derived teratoma in vitro biological system completed by a spent media metabolome analysis. Gastrulating 9.5-day-old rat embryos-proper were cultivated in Eagle's minimal essential medium (MEM) with 50% rat serum (controls) or with the addition of 2 mmVPA. Spent media metabolomes were analyzed by FTIR. Compared to controls, VPA acetylated histones; significantly diminished overall teratoma growth, impaired survival, increased the apoptotic index, and decreased proliferation index and incidence of differentiated tissues (e.g., neural tissue). Control teratomas continued to grow and differentiate for 14 days in isotransplants in vivo, but in vitro VPA-treated teratomas resorbed. Principal component analysis of FTIR results showed that spent media metabolomes formed well-separated clusters reflecting the treatment and day of cultivation. In metabolomes of VPA-treated teratomas, we found elevation of previously described histone acetylation biomarkers [amide I α-helix and A(CH3 )/A(CH2 )]) with apoptotic biomarkers within the amide I region for ß-sheets, and unordered and CH2 vibrations of lipids. VPA may be proposed for therapy of the undifferentiated component of teratoma tumors and this biological system completed by metabolome analysis, for a faster dual screening of antitumor/embryotoxic agents.


Asunto(s)
Embrión de Mamíferos/efectos de los fármacos , Histonas/metabolismo , Teratoma/prevención & control , Ácido Valproico/farmacología , Acetilación/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Femenino , Inhibidores de Histona Desacetilasas/farmacología , Masculino , Ratas , Espectroscopía Infrarroja por Transformada de Fourier , Teratoma/metabolismo , Teratoma/patología
7.
Front Immunol ; 10: 2580, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31787975

RESUMEN

The safe utilization of induced pluripotent stem cell (iPSC) derivatives in clinical use is attributed to the complete elimination of the risk of forming teratomas after transplantation. The extent by which such a risk exists in immune-competent hosts is mostly unknown. Here, using humanized mice reconstituted with fetal hematopoietic stem cells and autologous thymus tissue (bone-liver-thymus humanized mice [Hu-BLT]) or following the adoptive transfer of peripheral blood mononuclear cells(PBMCs) (Hu-AT), we evaluated the capacity of immune cells to prevent or eliminate teratomas derived from human iPSCs (hiPSCs). Our results showed that the injection of hiPSCs failed to form teratomas in Hu-AT mice reconstituted with allogeneic or autologous PBMCs or purified natural killer (NK) cells alone. However, teratomas were observed in Hu-AT mice reconstituted with autologous PBMCs depleted from NK cells. In line with these results, Hu-BLT, which do not have functional NK cells, could not prevent the growth of teratomas. Finally, we found that established teratomas were not targeted by NK cells and instead were efficiently rejected by allogeneic but not autologous T cells in Hu-AT mice. Overall, our findings suggest that autologous hiPSC-derived therapies are unlikely to form teratomas in the presence of NK cells.


Asunto(s)
Células Asesinas Naturales/inmunología , Células Madre Pluripotentes/inmunología , Teratoma/prevención & control , Traslado Adoptivo/efectos adversos , Adulto , Animales , Humanos , Leucocitos Mononucleares/trasplante , Ratones , Ratones Endogámicos NOD , Ratones SCID , Linfocitos T/inmunología , Teratoma/etiología , Teratoma/inmunología , Trasplante Heterólogo
8.
Int J Exp Pathol ; 99(3): 131-144, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-30066346

RESUMEN

We screened for the impact of hyperthermal regimes varying in the cumulative equivalent minutes at 43°C (CEM43°C) and media composition on tumour development using an original teratoma in vitro model. Rat embryos (three germ layers) were microsurgically isolated and cultivated at the air-liquid interface. During a two week period, ectodermal, mesodermal and endodermal derivatives developed within trilaminar teratomas. Controls were grown at 37°C. Overall growth was measured, and teratoma survival and differentiation were histologically assessed. Cell proliferation was stereologically quantified by the volume density of Proliferating Cell Nuclear Antigen. Hyperthermia of 42°C, applied for 15 minutes after plating (CEM43°C 3.75 minutes), diminished cell proliferation (P Ë‚ .0001) and enhanced differentiation of both myotubes (P Ë‚ .01) and cylindrical epithelium (P Ë‚ .05). Hyperthermia of 43°C applied each day for 30 minutes during the first week (CEM43°C 210 minutes) impaired overall growth (P Ë‚ .01) and diminished cell proliferation (P Ë‚ .0001). Long-term hyperthermia of 40.5°C applied for two weeks (CEM43°C 630 minutes) significantly impaired survival (P Ë‚ .005). Long-term hyperthermia of 40.5°C applied from the second day when differentiation of tissues begins (CEM43°C 585 minutes) impaired survival (P Ë‚ .0001), overall growth (P Ë‚ .01) and cartilage differentiation (P Ë‚ .05). No teratomas survived extreme regimes: 43°C for 24 hours (CEM43°C 1440 minutes), hyperthermia in the scant serum-free medium (CEM43°C 630 minutes) or treatment with an anti-HSP70 antibody before long-term hyperthermia 40.5°C from the second day (CEM43°C 585 minutes). This in vitro research provided novel insights into the impact of hyperthermia on the development of experimental teratomas from their undifferentiated sources and are thus of potential interest for future therapeutic strategies in corresponding in vivo models.


Asunto(s)
Embrión de Mamíferos/patología , Hipertermia Inducida/métodos , Teratoma/patología , Teratoma/prevención & control , Animales , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Técnicas de Cultivo de Embriones , Embrión de Mamíferos/metabolismo , Femenino , Embarazo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas Endogámicas F344 , Teratoma/metabolismo , Factores de Tiempo
9.
BMC Vet Res ; 14(1): 213, 2018 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-29954396

RESUMEN

BACKGROUND: Articular cartilage lacks a regenerative response. Embryonic stem cells (ESCs) are a source of pluripotent cells for cartilage regeneration. Their use, however, is associated with a risk of teratoma development, which depends on multiple factors including the number of engrafted cells and their degree of histocompatibility with recipients, the immunosuppression of the host and the site of transplantation. Colonies of sheep embryonic stem-like (ES-like) cells from in vitro-produced embryos, positive for stage-specific embryonic antigens (SSEAs), alkaline phosphatase (ALP), Oct 4, Nanog, Sox 2 and Stat 3 gene expression, and forming embryoid bodies, were pooled in groups of two-three, embedded in fibrin glue and engrafted into osteochondral defects in the left medial femoral condyles of 3 allogeneic ewes (ES). Empty defects (ED) and defects filled with cell-free glue (G) in the condyles of the controlateral stifle joint served as controls. After euthanasia at 4 years post-engraftment, the regenerated tissue was evaluated by macroscopic, histological and immunohistochemical (collagen type II) examinations and fluorescent in situ hybridization (FISH) assay to prove the ES-like cells origin of the regenerated tissue. RESULTS: No teratoma occurred in any of the ES samples. No statistically significant macroscopic or histological differences were observed among the 3 treatment groups. FISH was positive in all the 3 ES samples. CONCLUSIONS: This in vivo preclinical study allowed a long-term evaluation of the occurrence of teratoma in non-immunosuppressed allogeneic adult sheep engrafted with allogeneic ES-like cells, supporting the safe and reliable application of ES cells in the clinic.


Asunto(s)
Células Madre Embrionarias/trasplante , Fémur/lesiones , Ovinos/lesiones , Animales , Neoplasias Óseas/prevención & control , Neoplasias Óseas/veterinaria , Trasplante Óseo/efectos adversos , Trasplante Óseo/métodos , Trasplante Óseo/veterinaria , Femenino , Fémur/patología , Fémur/cirugía , Estudios de Seguimiento , Hibridación Fluorescente in Situ/veterinaria , Masculino , Ovinos/cirugía , Teratoma/prevención & control , Teratoma/veterinaria
10.
Sci Rep ; 7(1): 5289, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28706279

RESUMEN

An important safety concern in the use of human pluripotent stem cells (hPSCs) is tumorigenic risk, because these cells can form teratomas after an in vivo injection at ectopic sites. Several thousands of undifferentiated hPSCs are sufficient to induce teratomas in a mouse model. Thus, it is critical to remove all residue-undifferentiated hPSCs that have teratoma potential before the clinical application of hPSC-derived cells. In this study, our data demonstrated the cytotoxic effects of cardiac glycosides, such as digoxin, lanatoside C, bufalin, and proscillaridin A, in human embryonic stem cells (hESCs). This phenomenon was not observed in human bone marrow mesenchymal stem cells (hBMMSCs). Most importantly, digoxin and lanatoside C did not affect the stem cells' differentiation ability. Consistently, the viability of the hESC-derived MSCs, neurons, and endothelium cells was not affected by the digoxin and lanatoside C treatment. Furthermore, the in vivo experiments demonstrated that digoxin and lanatoside C prevented teratoma formation. To the best of our knowledge, this study is the first to describe the cytotoxicity and tumor prevention effects of cardiac glycosides in hESCs. Digoxin and lanatoside C are also the first FDA-approved drugs that demonstrated cytotoxicity in undifferentiated hESCs.


Asunto(s)
Adipogénesis/efectos de los fármacos , Glicósidos Cardíacos/farmacología , Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias Humanas/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Teratoma/prevención & control , Animales , Técnicas de Cultivo de Célula , Células Cultivadas , Células Madre Embrionarias Humanas/patología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Teratoma/metabolismo , Teratoma/patología
11.
Stem Cells Transl Med ; 6(2): 382-393, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28191782

RESUMEN

Iatrogenic tumorigenesis is a major limitation for the use of human induced pluripotent stem cells (hiPSCs) in hematology. The teratoma risk comes from the persistence of hiPSCs in differentiated cell populations. Our goal was to evaluate the best system to purge residual hiPSCs before graft without compromising hematopoietic repopulation capability. Teratoma risk after systemic injection of hiPSCs expressing the reporter gene luciferase was assessed for the first time. Teratoma formation in immune-deficient mice was tracked by in vivo bioimaging. We observed that systemic injection of hiPSCs produced multisite teratoma as soon as 5 weeks after injection. To eliminate hiPSCs before grafting, we tested the embryonic-specific expression of suicide genes under the control of the pmiR-302/367 promoter. This promoter was highly active in hiPSCs but not in differentiated cells. The gene/prodrug inducible Caspase-9 (iCaspase-9)/AP20187 was more efficient and rapid than thymidine kinase/ganciclovir, fully specific, and without bystander effect. We observed that iCaspase-9-expressing hiPSCs died in a dose-dependent manner with AP20187, without reaching full eradication in vitro. Unexpectedly, nonspecific toxicity of AP20187 on iCaspase-9-negative hiPSCs and on CD34+ cells was evidenced in vitro. This toxic effect strongly impaired CD34+ -derived human hematopoiesis in adoptive transfers. Survivin inhibition is an alternative to the suicide gene approach because hiPSCs fully rely on survivin for survival. Survivin inhibitor YM155 was more efficient than AP20187/iCaspase-9 for killing hiPSCs, without toxicity on CD34+ cells, in vitro and in adoptive transfers. hiPSC purge by survivin inhibitor fully eradicated teratoma formation in immune-deficient mice. This will be useful to improve the safety management for hiPSC-based medicine. Stem Cells Translational Medicine 2017;6:382-393.


Asunto(s)
Caspasa 9/genética , Transformación Celular Neoplásica/efectos de los fármacos , Genes Transgénicos Suicidas , Enfermedades Hematológicas/cirugía , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/efectos de los fármacos , Imidazoles/farmacología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/trasplante , Naftoquinonas/farmacología , Medicina Regenerativa/métodos , Survivin/antagonistas & inhibidores , Tacrolimus/análogos & derivados , Teratoma/prevención & control , Animales , Caspasa 9/metabolismo , Línea Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Relación Dosis-Respuesta a Droga , Regulación Neoplásica de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Ratones Endogámicos NOD , Ratones SCID , Fenotipo , Medición de Riesgo , Survivin/metabolismo , Tacrolimus/farmacología , Teratoma/genética , Teratoma/metabolismo , Teratoma/patología , Factores de Tiempo , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Curr Res Transl Med ; 64(2): 97-106, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27316393

RESUMEN

Stem cell-based therapy is currently tested in several trials of chronic heart failure. The main question is to determine how its implementation could be extended to standard clinical practice. To answer this question, it is helpful to capitalize on the three main lessons drawn from the accumulated experience, both in the laboratory and in the clinics. Regarding the cell type, the best outcomes seem to be achieved by cells the phenotype of which closely matches that of the target tissue. This argues in favor of the use of cardiac-committed cells among which the pluripotent stem cell-derived cardiac progeny is particularly attractive. Regarding the mechanism of action, there has been a major paradigm shift whereby cells are no longer expected to structurally integrate within the recipient myocardium but rather to release biomolecules that foster endogenous repair processes. This implies to focus on early cell retention, rather than on sustained cell survival, so that the cells reside in the target tissue long enough and in sufficient amounts to deliver the factors underpinning their action. Biomaterials are here critical adjuncts to optimize this residency time. Furthermore, the paracrine hypothesis gives more flexibility for using allogeneic cells in that targeting an only transient engraftment requires to delay, and no longer to avoid, rejection, which, in turn, should simplify immunomodulation regimens. Regarding manufacturing, a broad dissemination of cardiac cell therapy requires the development of automated systems allowing to yield highly reproducible cell products. This further emphasizes the interest of allogeneic cells because of their suitability for industrially-relevant and cost-effective scale-up and quality control procedures. At the end, definite confirmation that the effects of cells can be recapitulated by the factors they secrete could lead to acellular therapies whereby factors alone (possibly clustered in extracellular vesicles) would be delivered to the patient. The production process of these cell-derived biologics would then be closer to that of a pharmaceutical compound, which could streamline the manufacturing and regulatory paths and thereby facilitate an expended clinical use.


Asunto(s)
Insuficiencia Cardíaca/terapia , Trasplante de Células Madre , Aloinjertos , Animales , Separación Celular/métodos , Células Madre Embrionarias/trasplante , Vesículas Extracelulares/trasplante , Supervivencia de Injerto , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Ratones , MicroARNs/fisiología , MicroARNs/uso terapéutico , Miocardio/citología , Miocitos Cardíacos/trasplante , Comunicación Paracrina , Células Madre Pluripotentes/trasplante , Ensayos Clínicos Controlados Aleatorios como Asunto , Trasplante de Células Madre/efectos adversos , Trasplante de Células Madre/economía , Trasplante de Células Madre/métodos , Células Madre/metabolismo , Teratoma/etiología , Teratoma/prevención & control , Ingeniería de Tejidos
13.
Oncotarget ; 7(3): 2684-95, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26646796

RESUMEN

Although pluripotent stem cell (PSC) therapy has advantages for clinical applications because of the self-renewal and multi-lineage differentiation abilities of PSCs, it also has disadvantages in terms of the potential for PSCs to undergo malignant transformation or unexpected differentiation. The prevention of teratoma formation is the largest hurdle of all. Despite intensive studies that have investigated ways to block teratomas, such methods have yet to be further developed for clinical use. Here, a new approach has focused on exerting anti-tumorigenic effects using a novel mica fine particle (MFP) designated STB-HO. Treatment with STB-HO regulated pluripotency- and apoptosis-related genes in differentiating human embryonic stem (hES) cells, while there is no effects in undifferentiated hES cells. In particular, STB-HO blocked the anti-apoptotic gene BIRC5 and activated p53, p21 and the pro-apoptotic proteins Bim, Puma and p-Bad during early spontaneous differentiation. Moreover, STB-HO-pretreated differentiating hES cells did not give rise to teratomas following in vivo stem cell transplantation. Our in vitro and in vivo results suggest a method for teratoma prevention in the context of PSC-derived cell transplantation. This novel MFP could break through the limitations of PSC therapy.


Asunto(s)
Silicatos de Aluminio/farmacología , Transformación Celular Neoplásica/patología , Células Madre Embrionarias Humanas/citología , Nanopartículas/administración & dosificación , Células Madre Pluripotentes/citología , Trasplante de Células Madre , Teratoma/prevención & control , Animales , Western Blotting , Transformación Celular Neoplásica/efectos de los fármacos , Células Cultivadas , Femenino , Células Madre Embrionarias Humanas/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Madre Pluripotentes/efectos de los fármacos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Teratoma/patología
14.
Stem Cell Reports ; 5(6): 1067-1080, 2015 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-26584542

RESUMEN

Stem-toxic small molecules have been developed to induce selective cell death of pluripotent stem cells (PSCs) to lower the risk of teratoma formation. However, despite their high efficacies, chemical-based approaches may carry unexpected toxicities on specific differentiated cell types. Herein, we took advantage of KillerRed (KR) as a suicide gene, to selectively induce phototoxicity using visible light via the production of reactive oxygen species. PSCs in an undifferentiated state that exclusively expressed KR (KR-PSCs) were eliminated by a single exposure to visible light. This highly selective cell death in KR-PSCs was exploited to successfully inhibit teratoma formation. In particular, endothelial cells from KR-mPSCs remained fully functional in vitro and sufficient to repair ischemic injury in vivo regardless of light exposure, suggesting that a genetic approach in which KR is expressed in a tightly controlled manner would be a viable strategy to inhibit teratoma formation for future safe PSC-based therapies.


Asunto(s)
Células Endoteliales/trasplante , Isquemia/terapia , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de la radiación , Teratoma/prevención & control , Animales , Muerte Celular/efectos de la radiación , Diferenciación Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Células Endoteliales/citología , Femenino , Miembro Posterior/irrigación sanguínea , Luz , Ratones , Ratones Desnudos , Células Madre Pluripotentes/metabolismo , Especies Reactivas de Oxígeno/metabolismo
15.
Stem Cell Res Ther ; 6: 69, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25879223

RESUMEN

INTRODUCTION: Human parthenogenetic embryonic stem cells (hpESCs) are generated from artificially activated oocytes, however, the issue of whether hpESCs have equivalent differentiation ability to human fertilized embryonic stem cells remains controversial. METHODS: hpESCs were injected into male severe combined immunodeficiency (SCID) mice and the efficiency of teratoma formation was calculated. Then the gene expression and methylation modification were detected by real time-PCR and bisulfate methods. RESULTS: Comparison of five hpESCs with different differentiation abilities revealed that levels of paternal genes in the Dlk1-Dio3 region on chromosome 14 in the hpESCs with high differentiation potential are enhanced, but strictly methylated and silenced in the hpESCs with lower differentiation potential. Treatment with ascorbic acid, rescued their ability to support teratoma formation and altered the expression profiles of paternally expressed genes in hpESCs that could not form teratoma easily. No differences in the expression of other imprinting genes were evident between hpESCs with higher and lower differentiation potential, except for those in the Dlk1-Dio3 region. CONCLUSIONS: The Dlk1-Dio3 imprinting gene cluster distinguishes the differentiation ability of hpESCs. Moreover, modification by ascorbic acid may facilitate application of hpESCs to clinical settings in the future by enhancing their pluripotency.


Asunto(s)
Ácido Ascórbico/farmacología , Células Madre Embrionarias/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/genética , Yoduro Peroxidasa/genética , Proteínas de la Membrana/genética , Teratoma/prevención & control , Animales , Proteínas de Unión al Calcio , Diferenciación Celular , Metilación de ADN/genética , Técnicas de Cultivo de Embriones , Expresión Génica/genética , Perfilación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Yoduro Peroxidasa/biosíntesis , Masculino , Proteínas de la Membrana/biosíntesis , Ratones , Ratones SCID , Familia de Multigenes/genética , Partenogénesis , Células Madre Pluripotentes/efectos de los fármacos , Teratoma/genética
16.
Expert Rev Clin Immunol ; 11(6): 709-20, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25873365

RESUMEN

The treatment of psoriasis in pregnant women can be challenging. Psoriasis generally improves during pregnancy; however, many pregnant patients still require treatment. In treating pregnant patients, the benefits of treatment and risks to the mother and the fetus must be considered. For localized psoriasis, topical corticosteroids are the treatment of choice. Other topical agents that are approved for the treatment of psoriasis, such as topical tar products and topical tazarotene, should be avoided during pregnancy because of unclear risks of teratogenicity. For moderate-to-severe psoriasis, ultraviolet B phototherapy is preferred. Despite limited safety data, biologics are favored over other systemic medications when needed. While there are new treatment options for psoriasis, there is limited information on the safety of medications during pregnancy.


Asunto(s)
Corticoesteroides/uso terapéutico , Ácidos Nicotínicos , Fototerapia , Psoriasis/terapia , Teratoma/prevención & control , Animales , Contraindicaciones , Femenino , Humanos , Masculino , Ácidos Nicotínicos/efectos adversos , Embarazo , Psoriasis/complicaciones , Riesgo , Teratoma/etiología , Rayos Ultravioleta
17.
J Neurosci ; 34(24): 8318-23, 2014 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-24920634

RESUMEN

The heparan sulfate proteoglycan Glypican 4 (Gpc4) is strongly expressed in mouse embryonic stem (ES) cells where it controls the maintenance of self-renewal by modulating Wnt/ß-catenin signaling activities. Here we show that mouse ES cells carrying a hypomorphic Gpc4 allele, in a single-step neuronal differentiation protocol, show increased differentiation into dopaminergic neurons expressing tyrosine hydroxylase (TH) and nuclear receptor related-1 protein (Nurr1) 1. In contrast to wild-type cells, these differentiating Gpc4-mutant cells expressed high levels of DOPA decarboxylase and the dopamine transporter, two markers expressed by fully mature dopaminergic neurons. Intrastriatal transplantation of Gpc4 hypomorphic cells into a 6-OHDA rat model for Parkinson's disease improved motor behavior in the cylinder test and amphetamine-induced rotations at a higher level than transplanted wild-type cells. Importantly, Gpc4 hypomorphic cell grafts, in contrast to wild-type cells, did not generate teratomas in the host brains, leading to strongly enhanced animal survival. Therefore, control of Gpc4 activity level represents a new potential strategy to reduce ES cell tumorigenic features while at the same time increasing neuronal differentiation and integration.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Células Madre Embrionarias/trasplante , Glipicanos/metabolismo , Enfermedad de Parkinson/fisiopatología , Enfermedad de Parkinson/cirugía , Teratoma/prevención & control , Animales , Calbindinas/metabolismo , Recuento de Células , Diferenciación Celular , Modelos Animales de Enfermedad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Glipicanos/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Factor Nuclear 3-beta del Hepatocito/metabolismo , Humanos , Ratones , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Ratas , Receptores de Dopamina D2/metabolismo , Recuperación de la Función/fisiología , Teratoma/etiología , Tirosina 3-Monooxigenasa/metabolismo
18.
PLoS One ; 9(3): e85039, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24647085

RESUMEN

A major concern in Pluripotent Stem Cell (PSC)-derived cell replacement therapy is the risk of teratoma formation from contaminating undifferentiated cells. Removal of undifferentiated cells from differentiated cultures is an essential step before PSC-based cell therapies can be safely deployed in a clinical setting. We report a group of novel small molecules that are cytotoxic to PSCs. Our data indicates that these molecules are specific and potent in their activity allowing rapid eradication of undifferentiated cells. Experiments utilizing mixed PSC and primary human neuronal and cardiomyocyte cultures demonstrate that up to a 6-fold enrichment for specialized cells can be obtained without adversely affecting cell viability and function. Several structural variants were synthesized to identify key functional groups and to improve specificity and efficacy. Comparative microarray analysis and ensuing RNA knockdown studies revealed involvement of the PERK/ATF4/DDIT3 ER stress pathway. Surprisingly, cell death following ER stress induction was associated with a concomitant decrease in endogenous ROS levels in PSCs. Undifferentiated cells treated with these molecules preceding transplantation fail to form teratomas in SCID mice. Furthermore, these molecules remain non-toxic and non-teratogenic to zebrafish embryos suggesting that they may be safely used in vivo.


Asunto(s)
Citotoxinas/farmacología , Células Madre Pluripotentes/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Teratoma/prevención & control , Factor de Transcripción Activador 4/antagonistas & inhibidores , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Animales , Diferenciación Celular , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citotoxinas/síntesis química , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/fisiología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Regulación de la Expresión Génica , Humanos , Ratones , Ratones SCID , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Especificidad de Órganos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Bibliotecas de Moléculas Pequeñas/síntesis química , Trasplante de Células Madre , Relación Estructura-Actividad , Factor de Transcripción CHOP/antagonistas & inhibidores , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Pez Cebra , eIF-2 Quinasa/antagonistas & inhibidores , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo
19.
Cell Stem Cell ; 14(1): 121-30, 2014 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-24388175

RESUMEN

Human embryonic stem cells (hESCs) hold great promise for cell therapy as a source of diverse differentiated cell types. One key bottleneck to realizing such potential is allogenic immune rejection of hESC-derived cells by recipients. Here, we optimized humanized mice (Hu-mice) reconstituted with a functional human immune system that mounts a vigorous rejection of hESCs and their derivatives. We established knockin hESCs that constitutively express CTLA4-Ig and PD-L1 before and after differentiation, denoted CP hESCs. We then demonstrated that allogenic CP hESC-derived teratomas, fibroblasts, and cardiomyocytes are immune protected in Hu-mice, while cells derived from parental hESCs are effectively rejected. Expression of both CTLA4-Ig, which disrupts T cell costimulatory pathways, and PD-L1, which activates T cell inhibitory pathway, is required to confer immune protection, as neither was sufficient on their own. These findings are instrumental for developing a strategy to protect hESC-derived cells from allogenic immune responses without requiring systemic immune suppression.


Asunto(s)
Diferenciación Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Células Madre Embrionarias/citología , Células Madre Embrionarias/inmunología , Rechazo de Injerto/prevención & control , Terapia de Inmunosupresión , Abatacept , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Biomarcadores/análisis , Western Blotting , Proliferación Celular , Células Cultivadas , Células Madre Embrionarias/metabolismo , Feto/citología , Feto/metabolismo , Fibroblastos/citología , Fibroblastos/inmunología , Fibroblastos/metabolismo , Rechazo de Injerto/inmunología , Humanos , Inmunoconjugados/genética , Inmunoconjugados/metabolismo , Hígado/citología , Hígado/metabolismo , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Teratoma/inmunología , Teratoma/patología , Teratoma/prevención & control , Trasplante Homólogo
20.
Stem Cells Transl Med ; 1(10): 709-18, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23197662

RESUMEN

Pluripotent stem cells have been the focus of bioengineering efforts designed to generate regenerative products, yet harnessing therapeutic capacity while minimizing risk of dysregulated growth remains a challenge. The risk of residual undifferentiated stem cells within a differentiated progenitor population requires a targeted approach to eliminate contaminating cells prior to delivery. In this study we aimed to validate a toxicity strategy that could selectively purge pluripotent stem cells in response to DNA damage and avoid risk of uncontrolled cell growth upon transplantation. Compared with somatic cell types, embryonic stem cells and induced pluripotent stem cells displayed hypersensitivity to apoptotic induction by genotoxic agents. Notably, hypersensitivity in pluripotent stem cells was stage-specific and consistently lost upon in vitro differentiation, with the mean half-maximal inhibitory concentration increasing nearly 2 orders of magnitude with tissue specification. Quantitative polymerase chain reaction and Western blotting demonstrated that the innate response was mediated through upregulation of the BH3-only protein Puma in both natural and induced pluripotent stem cells. Pretreatment with genotoxic etoposide purged hypersensitive pluripotent stem cells to yield a progenitor population refractory to teratoma formation upon transplantation. Collectively, this study exploits a hypersensitive apoptotic response to DNA damage within pluripotent stem cells to decrease risk of dysregulated growth and augment the safety profile of transplant-ready, bioengineered progenitor cells.


Asunto(s)
Apoptosis , Daño del ADN , Células Madre Pluripotentes/trasplante , Teratoma/prevención & control , Animales , Anexina A5/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Western Blotting , Diferenciación Celular , Células Cultivadas , Cuerpos Embrioides/citología , Cuerpos Embrioides/efectos de los fármacos , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Etopósido/farmacología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Citometría de Flujo , Inestabilidad Genómica , Concentración 50 Inhibidora , Lentivirus/genética , Lentivirus/metabolismo , Ratones , Ratones Desnudos , Pruebas de Mutagenicidad/métodos , Especificidad de Órganos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Factores de Riesgo , Teratoma/patología , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...