Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
BMC Biol ; 11: 86, 2013 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-23870169

RESUMEN

BACKGROUND: Human pluripotent stem cells (hPSCs) hold great promise for applications in regenerative medicine. However, the safety of cell therapy using differentiated hPSC derivatives must be improved through methods that will permit the transplantation of homogenous populations of a specific cell type. To date, purification of progenitors and mature cells generated from either embryonic or induced pluripotent stem cells remains challenging with use of conventional methods. RESULTS: We used lentivectors encoding green fluorescent protein (GFP) driven by the liver-specific apoliprotein A-II (APOA-II) promoter to purify human hepatic progenitors. We evaluated both integrating and integration-defective lentivectors in combination with an HIV integrase inhibitor. A human embryonic stem cell line was differentiated into hepatic progenitors using a chemically defined protocol. Subsequently, cells were transduced and sorted at day 16 of differentiation to obtain a cell population enriched in hepatic progenitor cells. After sorting, more than 99% of these APOA-II-GFP-positive cells expressed hepatoblast markers such as α-fetoprotein and cytokeratin 19. When further cultured for 16 days, these cells underwent differentiation into more mature cells and exhibited hepatocyte properties such as albumin secretion. Moreover, they were devoid of vector DNA integration. CONCLUSIONS: We have developed an effective strategy to purify human hepatic cells from cultures of differentiating hPSCs, producing a novel tool that could be used not only for cell therapy but also for in vitro applications such as drug screening. The present strategy should also be suitable for the purification of a broad range of cell types derived from either pluripotent or adult stem cells.


Asunto(s)
Diferenciación Celular , Separación Celular/métodos , Células Madre Embrionarias/citología , Vectores Genéticos/genética , Hepatocitos/citología , Lentivirus/genética , Integración Viral/fisiología , Apolipoproteína A-II/genética , Biomarcadores/metabolismo , Línea Celular , Citocromo P-450 CYP3A/metabolismo , ADN Viral/metabolismo , Citometría de Flujo , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Hepatocitos/metabolismo , Humanos , Hígado/citología , Especificidad de Órganos , Regiones Promotoras Genéticas/genética , Transducción Genética
2.
Hepatology ; 51(5): 1754-65, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20301097

RESUMEN

UNLABELLED: Generation of hepatocytes from human embryonic stem cells (hESCs) could represent an advantageous source of cells for cell therapy approaches as an alternative to orthotopic liver transplantation. However, the generation of differentiated hepatocytes from hESCs remains a major challenge, especially using a method compatible with clinical applications. We report a novel approach to differentiate hESCs into functional hepatic cells using fully defined culture conditions, which recapitulate essential stages of liver development. hESCs were first differentiated into a homogenous population of endoderm cells using a combination of activin, fibroblast growth factor 2, and bone morphogenetic protein 4 together with phosphoinositide 3-kinase inhibition. The endoderm cells were then induced to differentiate further into hepatic progenitors using fibroblast growth factor 10, retinoic acid, and an inhibitor of activin/nodal receptor. After further maturation, these cells expressed markers of mature hepatocytes, including asialoglycoprotein receptor, tyrosine aminotransferase, alpha1-antitrypsin, Cyp7A1, and hepatic transcription factors such as hepatocyte nuclear factors 4alpha and 6. Furthermore, the cells generated under these conditions exhibited hepatic functions in vitro, including glycogen storage, cytochrome activity, and low-density lipoprotein uptake. After transduction with a green fluorescent protein-expressing lentivector and transplantation into immunodeficient uPA transgenic mice, differentiated cells engrafted into the liver, grew, and expressed human albumin and alpha1-antitrypsin as well as green fluorescent protein for at least 8 weeks. In addition, we showed that hepatic cells could be generated from human-induced pluripotent cells derived from reprogrammed fibroblasts, demonstrating the efficacy of this approach with pluripotent stem cells of diverse origins. CONCLUSION: We have developed a robust and efficient method to differentiate pluripotent stem cells into hepatic cells, which exhibit characteristics of human hepatocytes. Our approach should facilitate the development of clinical grade hepatocytes for transplantation and for research on drug discovery.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Hepatocitos/citología , Hígado/embriología , Activinas/farmacología , Animales , Benzamidas/farmacología , Proteína Morfogenética Ósea 4/farmacología , Diferenciación Celular/fisiología , Cromonas/farmacología , Dioxoles/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Morfolinas/farmacología , Células Madre Pluripotentes/citología , Tretinoina/farmacología
3.
Hepatology ; 49(3): 950-9, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19152424

RESUMEN

UNLABELLED: The feasibility of ex vivo gene therapy as an alternative to liver transplantation for the treatment of liver metabolic diseases needs to be analyzed in large animal models. This approach requires appropriate gene transfer vectors and effective hepatocyte engraftment. Lentiviral vectors have the ability to transduce nondividing differentiated cells, such as hepatocytes, and portal vein occlusion increases hepatocyte engraftment. We investigated whether reversible portal vein embolization combined with ex vivo lentivirus-mediated gene transfer is an effective approach for successful hepatocyte engraftment in nonhuman primates and whether the transgene remains expressed in the long term in transplanted hepatocytes in situ. Simian hepatocytes were isolated after left lobe resection, and the left and right anterior portal branches of animals were embolized with absorbable material. Isolated hepatocytes were labeled with Hoechst dye or transduced in suspension with lentiviruses expressing green fluorescent protein under the control of the human apolipoprotein A-II promoter and transplanted via the inferior mesenteric vein. The whole procedure was well tolerated. The embolized liver was revascularized within 2 weeks. The volume of nonembolized liver increased from 38.7% +/- 0.8% before embolization to 55.9% +/- 1% after embolization and hepatocytes significantly proliferated (10.5% +/- 0.4% on day 3 after embolization). Liver repopulation after transplantation with Hoechst-labeled hepatocytes was 7.4% +/- 1.2%. Liver repopulation was 2.1% +/- 0.2% with transduced hepatocytes, a proportion similar to that obtained with Hoechst-labeled cells, given that the mean transduction efficacy of simian hepatocyte population was 34%. Transgene expression persisted at 16 weeks after transplantation. CONCLUSION: We have developed a new approach to improve hepatocyte engraftment and to express a transgene in the long term in nonhuman primates. This strategy could be suitable for clinical applications.


Asunto(s)
Apolipoproteína A-II/metabolismo , Trasplante de Células/métodos , Embolización Terapéutica/métodos , Hepatocitos/metabolismo , Hepatocitos/trasplante , Animales , Apolipoproteína A-II/genética , Proliferación Celular , Regulación de la Expresión Génica , Terapia Genética , Hepatocitos/patología , Humanos , Lentivirus/genética , Hígado/metabolismo , Hígado/patología , Hígado/cirugía , Regeneración Hepática/fisiología , Macaca mulatta , Modelos Animales , Vena Porta/cirugía , Transgenes/genética
4.
Exp Cell Res ; 315(19): 3396-405, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19615360

RESUMEN

UNLABELLED: The success of hepatocyte transplantation has been limited by the low efficiency of transplanted cell integration into liver parenchyma. Human fetal hepatic progenitor cells (hepatoblasts) engraft more effectively than adult hepatocytes in mouse livers. However, the signals required for their integration are not yet fully understood. We investigated the role of HGF on the migration and invasive ability of human hepatic progenitors in vitro and in vivo. Hepatoblasts were isolated from the livers of human fetuses between 10 and 12 weeks of gestation. Their invasive ability was assessed in the presence or absence of HGF. These cells were also transplanted into immunodeficient mice and analyzed by immunohistochemistry. In contrast to TNF-alpha, HGF increased the motogenesis and invasiveness of hepatoblasts, but not of human adult hepatocytes, via phosphorylation of extracellular signal-regulated kinase (ERK) 1/2. The invasive ability of human hepatoblasts correlated with the expression and secretion of matrix metalloproteinases (MMPs). Hepatoblasts stimulated with HGF prior transplantation into newborn mice migrated from the portal area into the hepatic parenchyma. CONCLUSIONS: In contrast to adult hepatocytes, hepatoblasts display invasive ability that can be modulated by HGF in vitro and in vivo.


Asunto(s)
Movimiento Celular , Factor de Crecimiento de Hepatocito/fisiología , Hepatocitos/trasplante , Trasplante de Células Madre/métodos , Células Madre/fisiología , Animales , Proliferación Celular , Feto , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Ratones , Ratones SCID , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Células Madre/citología , Trasplante Heterólogo
5.
Curr Gene Ther ; 9(2): 136-49, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19355871

RESUMEN

Transplantation of hepatocytes, whether genetically modified or not, has become an alternative to orthotopic liver transplantation for the treatment of patients with metabolic disease. However, more than ten years after the first clinical trial of ex vivo gene therapy to treat patients with Familial Hypercholesterolemia, there are still a number of impediments to these approaches. Numerous animal models are still being developed on the one hand to improve hepatocyte integration within hepatic parenchyma and function, and on the other hand to develop vectors that drive long-term transgene expression in situ. These include large animal models such as non-human primates, which have recently led to significant progress in hepatocyte transplantation. Simultaneous development of lentiviral vectors from different lentivirus species has permitted the transfer of genes into mitotically-quiescent primary cells including differentiated hepatocytes. Particularly third generation vectors derived from HIV-1 lentivirus are the most widely used and have significantly improved the safety and efficiency of these vectors. Given the shortage of organs and problems related to immunosuppression on one hand, and recent progresses in hepatocyte transduction and transplantation on the other hand, ex vivo approach is becoming a real alternative to allogeneic hepatocyte transplantation. We review the present progresses and limits of the ex vivo liver gene therapy approach in different animal models, emphasizing clinically relevant procedures.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/genética , Hepatocitos/trasplante , Enfermedades Metabólicas/terapia , Retroviridae/genética , Animales , Vectores Genéticos/metabolismo , Humanos , Hígado/metabolismo , Hepatopatías/terapia , Retroviridae/metabolismo
6.
Stem Cells Int ; 2016: 6323486, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27057173

RESUMEN

In line with the search of effective stem cell population that would progress liver cell therapy and because the rate and differentiation potential of mesenchymal stem cells (MSC) decreases with age, the current study investigates the hepatogenic differentiation potential of human fetal liver MSCs (FL-MSCs). After isolation from 11-12 gestational weeks' human fetal livers, FL-MSCs were shown to express characteristic markers such as CD73, CD90, and CD146 and to display adipocytic and osteoblastic differentiation potential. Thereafter, we explored their hepatocytic differentiation potential using the hepatogenic protocol applied for adult human liver mesenchymal cells. FL-MSCs differentiated in this way displayed significant features of hepatocyte-like cells as demonstrated in vitro by the upregulated expression of specific hepatocytic markers and the induction of metabolic functions including CYP3A4 activity, indocyanine green uptake/release, and glucose 6-phosphatase activity. Following transplantation, naive and differentiated FL-MSC were engrafted into the hepatic parenchyma of newborn immunodeficient mice and differentiated in situ. Hence, FL-MSCs appeared to be interesting candidates to investigate the liver development at the mesenchymal compartment level. Standardization of their isolation, expansion, and differentiation may also support their use for liver cell-based therapy development.

7.
Hum Gene Ther ; 15(12): 1219-28, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15684698

RESUMEN

Transplantation of hepatocytes is a promising alternative to liver transplantation for the treatment of severe liver diseases. However, this approach is hampered by the shortage of donor organs and intrinsic limitations of adult hepatocytes. To investigate whether most of the hurdles faced with adult hepatocytes could be surmounted by the use of human fetal hepatoblasts, we have developed a method to isolate, transduce, and cryopreserve hepatoblasts from human livers at an early stage of development (11-13 weeks of gestation). Cells were characterized in vitro for expression of specific markers, and in vivo for their proliferation and differentiation potential after transplantation into athymic mice. Most of the cells (80-90%) harbored a bipotent phenotype, expressing cytokeratins 8/18, albumin, and CK19. They proliferated spontaneously in culture and were efficiently transduced by a beta-galactosidase-expressing retrovirus (90%). After transplantation, cryopreserved cells engrafted into the liver of athymic mice and proliferated, resulting in up to 10% repopulation. Engrafted cells expressed markers of differentiated adult hepatocytes including albumin, alpha1-antitrypsin, cytochrome P450 3A4, and alpha-glutathione-S-transferase. When retrovirally transduced before transplantation they expressed the transgene in vivo. In summary, early human fetal hepatoblasts engraft, proliferate, and mature in athymic mouse liver, without conditioning the donor.


Asunto(s)
Criopreservación , Trasplante de Tejido Fetal , Hepatocitos/citología , Hígado/citología , Trasplante de Células Madre , Células Madre/citología , Albúminas/análisis , Albúminas/genética , Animales , Biomarcadores/análisis , División Celular , Proteínas Fluorescentes Verdes/metabolismo , Hepatocitos/trasplante , Humanos , Inmunohistoquímica , Queratinas/análisis , Queratinas/metabolismo , Hígado/embriología , Hígado/metabolismo , Ratones , Ratones Desnudos , Retroviridae/genética , Células Madre/metabolismo , Factores de Tiempo , Transducción Genética , Trasplante Heterólogo
8.
Stem Cells Transl Med ; 3(6): 686-91, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24736403

RESUMEN

The use of synthetic messenger RNAs to generate human induced pluripotent stem cells (iPSCs) is particularly appealing for potential regenerative medicine applications, because it overcomes the common drawbacks of DNA-based or virus-based reprogramming strategies, including transgene integration in particular. We compared the genomic integrity of mRNA-derived iPSCs with that of retrovirus-derived iPSCs generated in strictly comparable conditions, by single-nucleotide polymorphism (SNP) and copy number variation (CNV) analyses. We showed that mRNA-derived iPSCs do not differ significantly from the parental fibroblasts in SNP analysis, whereas retrovirus-derived iPSCs do. We found that the number of CNVs seemed independent of the reprogramming method, instead appearing to be clone-dependent. Furthermore, differentiation studies indicated that mRNA-derived iPSCs differentiated efficiently into hepatoblasts and that these cells did not load additional CNVs during differentiation. The integration-free hepatoblasts that were generated constitute a new tool for the study of diseased hepatocytes derived from patients' iPSCs and their use in the context of stem cell-derived hepatocyte transplantation. Our findings also highlight the need to conduct careful studies on genome integrity for the selection of iPSC lines before using them for further applications.


Asunto(s)
Reprogramación Celular , Fibroblastos/metabolismo , Vectores Genéticos , Células Madre Pluripotentes Inducidas/metabolismo , ARN Mensajero/metabolismo , Retroviridae/genética , Factores de Transcripción/metabolismo , Transfección/métodos , Diferenciación Celular , Células Cultivadas , Variaciones en el Número de Copia de ADN , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Genotipo , Hepatocitos/metabolismo , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Polimorfismo de Nucleótido Simple , Factores de Transcripción/genética
9.
Cell Med ; 4(2): 85-98, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26858856

RESUMEN

Innovative cell-based therapies are considered as alternatives to liver transplantation. Recent progress in lentivirus-mediated hepatocyte transduction has renewed interest in cell therapy for the treatment of inherited liver diseases. However, hepatocyte transplantation is still hampered by inefficient hepatocyte engraftment. We previously showed that partial portal vein embolization (PVE) improved hepatocyte engraftment in a nonhuman primate model. We developed here an ex vivo approach based on PVE and lentiviral-mediated transduction of hepatocytes from normal (New Zealand White, NZW) and Watanabe heritable hyperlipidemic (WHHL) rabbits: the large animal model of familial hypercholesterolemia type IIa (FH). FH is a life-threatening human inherited autosomal disease caused by a mutation in the low-density lipoprotein receptor (LDLR) gene, which leads to severe hypercholesterolemia and premature coronary heart disease. Rabbit hepatocytes were isolated from the resected left liver lobe, and the portal branches of the median lobes were embolized with Histoacryl® glue under radiologic guidance. NZW and WHHL hepatocytes were each labeled with Hoechst dye or transduced with lentivirus expressing GFP under the control of a liver-specific promoter (mTTR, a modified murine transthyretin promoter) and were then immediately transplanted back into donor animals. In our conditions, 65-70% of the NZW and WHHL hepatocytes were transduced. Liver repopulation after transplantation with the Hoechst-labeled hepatocytes was 3.5 ± 2%. It was 1.4 ± 0.6% after transplantation with either the transduced NZW hepatocytes or the transduced WHHL hepatocytes, which was close to that obtained with Hoechst-labeled cells, given the mean transduction efficacy. Transgene expression persisted for at least 8 weeks posttransplantation. Transduction of WHHL hepatocytes with an LDLR-encoding vector resulted in phenotypic correction in vitro as assessed by internalization of fluorescent LDL ligands. In conclusion, our results have applications for the treatment of inherited metabolic liver diseases, such as FH, by transplantation of lentivirally transduced hepatocytes.

10.
Methods Mol Biol ; 640: 41-55, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20645045

RESUMEN

Hepatocyte transplantation has become an alternative to orthotopic liver transplantation for the treatment of liver metabolic diseases. However, there is an increasing lack of donor organs and isolated mature hepatocytes are difficult to manipulate and cannot be expanded in vitro. It is therefore necessary to find alternative sources of hepatocytes, and different approaches to evaluate the therapeutic potential of stem cells of different origins are being developed. Hepatic progenitors (hepatoblasts) and/or foetal hepatocytes isolated from foetal livers may be one potential source to generate fully differentiated hepatocytes. We have reported that human foetal liver cells can be isolated and cultured. These cells also engraft and differentiate into mature hepatocytes in situ after transplantation into immunodeficient mice. Foetal cell populations could also be used as targets for gene therapy since efficient gene transfer is achieved with retroviral vectors. Use of such experimental approaches will help design strategies for clinical applications of liver cell therapy with hepatic progenitors.


Asunto(s)
Separación Celular/métodos , Feto/citología , Hepatocitos/citología , Hepatocitos/trasplante , Animales , Diferenciación Celular , Células Cultivadas , Femenino , Vectores Genéticos/genética , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Lentivirus/genética , Ratones , Ratones SCID , Embarazo , Transducción Genética
11.
Pathol Biol (Paris) ; 54(2): 58-63, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16545631

RESUMEN

Transplantation of genetically modified or unmodified hepatocytes appears to be a less invasive alternative to liver transplantation. However, clinical trials performed for the treatment of metabolic deficiencies resulted in a partial and transitory correction due to an insufficient number of engrafted and functional hepatocytes. In vitro, adult hepatocytes do not proliferate and the lack of organ donors limits their availability. Concomitantly, numerous works on hepatocyte transplantation in rodents have shown that cell engraftment was inefficient in normal livers. It is therefore necessary to explore the therapeutic potential of new cell sources such as stem cells and to develop pre-clinical models of transplantation. Foetal liver progenitor cells (hepatoblasts) are bipotent and express markers of both foetal hepatocytes and cholangiocytes. We have immortalized one clone of primate hepatoblasts using a retroviral vector expressing SV40 Large T and have characterized the cells at different population doublings (PDs). After 500 days in culture, immortalized cells remained bipotent and kept contact inhibition, in spite of numerous chromosomal rearrangements. After transplantation into athymic mice, the cells expressed hepatocyte functions but did not proliferate. We isolated, phenotypically characterized, transduced and cryopreserved early human hepatoblasts. These cells repopulate up to 7% of recipient immunodeficient mouse livers. This indicates that early progenitor cells display molecular characteristics related to proliferation and migration that allow these cells to engraft within hepatic parenchyma more efficiently than adult hepatocytes.


Asunto(s)
Primates/embriología , Trasplante de Células Madre , Animales , División Celular , Células Madre Hematopoyéticas/citología , Ratones , Trasplante Heterólogo
12.
J Hepatol ; 45(1): 99-107, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16723167

RESUMEN

BACKGROUND/AIMS: Lentivirus-mediated ex vivo gene therapy is becoming a promising approach for the treatment of liver metabolic disorders. However, the feasibility of this approach needs to be studied in large animal models. The purpose of this study was to evaluate the efficacy of ex vivo gene transfer into Macaca hepatocytes with two different HIV-1 derived lentiviral vectors. METHODS: A self-inactivating lentivector was constructed to express GFP under the control of the hepatic apolipoprotein A-II promoter. Freshly isolated and thawed hepatocytes were transduced in suspension with lentiviral vectors expressing the GFP gene under the control of a ubiquitous promoter (EF1-alpha) and the apolipoprotein A-II promoter. Transduced thawed hepatocytes were transplanted into the spleen of newborn mice, and livers analyzed 4 and 12 weeks after transplantation. RESULTS: We show that lentivectors are efficient in transducing hepatocytes in suspension either freshly isolated or cryopreserved. We also show that thawed and transduced hepatocytes engrafted and participated in liver growth after transplantation into newborn mice and that the apolipoprotein A-II promoter is functional. CONCLUSIONS: Our data show that transplantation of transduced hepatocytes into monkeys should allow to evaluate the fate of transplanted cells and transgene expression in a pre-clinical model of ex vivo gene therapy.


Asunto(s)
VIH-1/genética , Hepatocitos/fisiología , Lentivirus/genética , Actinas/genética , Animales , Apolipoproteína A-II/genética , Cartilla de ADN , Técnicas de Transferencia de Gen , Vectores Genéticos , Hepatocitos/virología , Macaca fascicularis , Regiones Promotoras Genéticas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA