Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Stem Cell Res Ther ; 15(1): 71, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38475825

RESUMO

BACKGROUND: Liver transplantation remains the only curative treatment for end-stage liver diseases. Unfortunately, there is a drastic organ donor shortage. Hepatocyte transplantation emerged as a viable alternative to liver transplantation. Considering their unique expansion capabilities and their potency to be driven toward a chosen cell fate, pluripotent stem cells are extensively studied as an unlimited cell source of hepatocytes for cell therapy. It has been previously shown that freshly prepared hepatocyte-like cells can cure mice from acute and chronic liver failure and restore liver function. METHODS: Human PSC-derived immature hepatic progenitors (GStemHep) were generated using a new protocol with current good manufacturing practice compliant conditions from PSC amplification and hepatic differentiation to cell cryopreservation. The therapeutic potential of these cryopreserved cells was assessed in two clinically relevant models of acute liver failure, and the mode of action was studied by several analytical methods, including unbiased proteomic analyses. RESULTS: GStemHep cells present an immature hepatic phenotype (alpha-fetoprotein positive, albumin negative), secrete hepatocyte growth factor and do not express major histocompatibility complex. A single dose of thawed GStemHep rescue mice from sudden death caused by acetaminophen and thioacetamide-induced acute liver failure, both in immunodeficient and immunocompetent animals in the absence of immunosuppression. Therapeutic biological effects were observed as soon as 3 h post-cell transplantation with a reduction in serum transaminases and in liver necrosis. The swiftness of the therapeutic effect suggests a paracrine mechanism of action of GStemHep leading to a rapid reduction of inflammation as well as a rapid cytoprotective effect with as a result a proteome reprograming of the host hepatocytes. The mode of action of GStemHep relie on the alleviation of inhibitory factors of liver regeneration, an increase in proliferation-promoting factors and a decrease in liver inflammation. CONCLUSIONS: We generated cryopreserved and current good manufacturing practice-compliant human pluripotent stem cell-derived immature hepatic progenitors that were highly effective in treating acute liver failure through rapid paracrine effects reprogramming endogenous hepatocytes. This is also the first report highlighting that human allogeneic cells could be used as cryopreserved cells and in the absence of immunosuppression for human PSC-based regenerative medicine for acute liver failure.


Assuntos
Falência Hepática Aguda , Células-Tronco Pluripotentes , Humanos , Animais , Camundongos , Proteômica , Fígado/metabolismo , Hepatócitos/metabolismo , Falência Hepática Aguda/terapia , Diferenciação Celular , Inflamação/metabolismo
2.
Biomacromolecules ; 24(2): 789-796, 2023 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-36655630

RESUMO

Macroporous hydrogels have great potential for biomedical applications. Liquid or gel-like pores were created in a photopolymerizable hydrogel by forming water-in-water emulsions upon mixing aqueous solutions of gelatin and a poly(ethylene oxide) (PEO)-based triblock copolymer. The copolymer constituted the continuous matrix, which dominated the mechanical properties of the hydrogel once photopolymerized. The gelatin constituted the dispersed phase, which created macropores in the hydrogel. The microstructures of the porous hydrogel were determined by the volume fraction of the gelatin phase. When volume fractions were close to 50 v%, free-standing hydrogels with interpenetrated morphology can be obtained thanks to the addition of a small amount of xanthan. The hydrogels displayed Young's moduli ranging from 5 to 30 kPa. They have been found to be non-swellable and non-degradable in physiological conditions. Preliminary viability tests with hepatic progenitor cells embedded in monophasic PEO-based hydrogels showed rapid mortality of the cells, whereas encouraging viability was observed in PEO-based triblock copolymer/gelatin macroporous hydrogels. The latter has the potential to be used in cell therapy.


Assuntos
Gelatina , Hidrogéis , Hidrogéis/química , Gelatina/química , Óxido de Etileno , Encapsulamento de Células , Polietilenoglicóis/química , Polímeros , Células-Tronco , Água
3.
Hepatology ; 75(4): 866-880, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34687060

RESUMO

BACKGROUND AND AIMS: Pluripotent stem cell-derived hepatocytes differentiated in monolayer culture are known to have more fetal than adult hepatocyte characteristics. If numerous studies tend to show that this immature phenotype might not necessarily be an obstacle to their use in transplantation, other applications such as drug screening, toxicological studies, or bioartificial livers are reliant on hepatocyte functionality and require full differentiation of hepatocytes. New technologies have been used to improve the differentiation process in recent years, usually evaluated by measuring the albumin production and CYP450 activity. Here we used the complex production and most importantly the activity of the coagulation factor IX (FIX) produced by mature hepatocytes to assess the differentiation of hemophilia B (HB) patient's induced pluripotent stem cells (iPSCs) in both monolayer culture and organoids. APPROACH AND RESULTS: Indeed, HB is an X-linked monogenic disease due to an impaired activity of FIX synthesized by hepatocytes in the liver. We have developed an in vitro model of HB hepatocytes using iPSCs generated from fibroblasts of a severe HB patient. We used CRISPR/Cas9 technology to target the genomic insertion of a coagulation factor 9 minigene bearing the Padua mutation to enhance FIX activity. Noncorrected and corrected iPSCs were differentiated into hepatocytes under both two-dimensional and three-dimensional differentiation protocols and deciphered the production of active FIX in vitro. Finally, we assessed the therapeutic efficacy of this approach in vivo using a mouse model of HB. CONCLUSIONS: Functional FIX, whose post-translational modifications only occur in fully mature hepatocytes, was only produced in corrected iPSCs differentiated in organoids. Immunohistochemistry analyses of mouse livers indicated a good cell engraftment, and the FIX activity detected in the plasma of transplanted animals confirmed rescue of the bleeding phenotype.


Assuntos
Hemofilia B , Células-Tronco Pluripotentes Induzidas , Fígado Artificial , Animais , Biomarcadores , Diferenciação Celular , Fator IX/genética , Hemofilia B/genética , Hemofilia B/terapia , Hepatócitos , Humanos
4.
Xenotransplantation ; 27(1): e12544, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31342573

RESUMO

Pluripotent stem cells have been investigated as a renewable source of therapeutic hepatic cells, in order to overcome the lack of transplantable donor hepatocytes. Whereas different studies were able to correct hepatic defects in animal models, they focused on the most mature phenotype of hepatocyte-like cells (HLCs) derived from pluripotent stem cells and needed freshly prepared cells, which limits clinical applications of HLCs. Here, we report the production of hepatic stem cells (pHSCs) from human-induced pluripotent stem cells (hiPSCs) in xeno-free, feeder-free, and chemically defined conditions using as extracellular matrix a recombinant laminin instead of Matrigel, an undefined animal-derived matrix. Freshly prepared and frozen pHSCs were transplanted via splenic injection in Gunn rats, the animal model for Crigler-Najjar syndrome. Following cell transplantation and daily immunosuppression treatment, bilirubinemia was significantly decreased (around 30% decrease, P < .05) and remained stable throughout the 6-month study. The transplanted pHSCs underwent maturation in vivo to restore the deficient metabolic hepatic function (bilirubin glucuronidation by UGT1A1). In conclusion, we demonstrate for the first time the differentiation of hiPSCs into pHSCs that (a) are produced using a differentiation protocol compatible with Good Manufacturing Practices, (b) can be frozen, and (c) are sufficient to demonstrate in vivo therapeutic efficacy to significantly lower hyperbilirubinemia in a model of inherited liver disease, despite their immature phenotype. Thus, our approach provides major advances toward future clinical applications and would facilitate cell therapy manufacturing from human pluripotent stem cells.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Síndrome de Crigler-Najjar/terapia , Hepatócitos/fisiologia , Hiperbilirrubinemia/terapia , Células-Tronco Pluripotentes Induzidas/fisiologia , Fígado/fisiologia , Transplante de Células-Tronco/métodos , Animais , Diferenciação Celular , Células Cultivadas , Criopreservação , Modelos Animais de Doenças , Humanos , Fígado/cirurgia , Ratos , Ratos Gunn , Medicina Regenerativa/métodos , Transplante Heterólogo
5.
Stem Cell Res Ther ; 10(1): 221, 2019 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-31358055

RESUMO

BACKGROUND: Familial hypercholesterolemia type IIA (FH) is due to mutations in the low-density lipoprotein receptor (LDLR) resulting in elevated levels of low-density lipoprotein cholesterol (LDL-c) in plasma and in premature cardiovascular diseases. As hepatocytes are the only cells capable of metabolizing cholesterol, they are therefore the target cells for cell/gene therapy approaches in the treatment of lipid metabolism disorders. Furthermore, the LDLR has been reported to be involved in hepatitis C virus (HCV) entry into hepatocytes; however, its role in the virus infection cycle is still disputed. METHODS: We generated induced pluripotent stem cells (iPSCs) from a homozygous LDLR-null FH-patient (FH-iPSCs). We constructed a correction cassette bearing LDLR cDNA under the control of human hepatic apolipoprotein A2 promoter that targets the adeno-associated virus integration site AAVS1. We differentiated both FH-iPSCs and corrected FH-iPSCs (corr-FH-iPSCs) into hepatocytes to study statin-mediated regulation of genes involved in cholesterol metabolism. Upon HCV particle inoculation, viral replication and production were quantified in these cells. RESULTS: We showed that FH-iPSCs displayed the disease phenotype. Using homologous recombination mediated by the CRISPR/Cas9 system, FH-iPSCs were genetically corrected by the targeted integration of a correction cassette at the AAVS1 locus. Both FH-iPSCs and corr-FH-iPSCs were then differentiated into functional polarized hepatocytes using a stepwise differentiation approach (FH-iHeps and corr-FH-iHeps). The correct insertion and expression of the correction cassette resulted in restoration of LDLR expression and function (LDL-c uptake) in corr-FH-iHeps. We next demonstrated that pravastatin treatment increased the expression of genes involved in cholesterol metabolism in both cell models. Moreover, LDLR expression and function were also enhanced in corr-FH-iHeps after pravastatin treatment. Finally, we demonstrated that both FH-iHeps and corr-FH-iHeps were as permissive to viral infection as primary human hepatocytes but that virus production in FH-iHeps was significantly decreased compared to corr-FH-iHeps, suggesting a role of the LDLR in HCV morphogenesis. CONCLUSIONS: Our work provides the first LDLR-null FH cell model and its corrected counterpart to study the regulation of cholesterol metabolism and host determinants of HCV life cycle, and a platform to screen drugs for treating dyslipidemia and HCV infection.


Assuntos
Sistemas CRISPR-Cas/genética , Edição de Genes , Hepatite C/patologia , Hiperlipoproteinemia Tipo II/patologia , Receptores de LDL/genética , Antivirais/farmacologia , Antivirais/uso terapêutico , Apolipoproteína A-II/genética , Diferenciação Celular , Colesterol/metabolismo , Hepacivirus/efeitos dos fármacos , Hepacivirus/fisiologia , Hepatite C/tratamento farmacológico , Hepatite C/virologia , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Hiperlipoproteinemia Tipo II/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Fenótipo , Plasmídeos/genética , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Pró-Proteína Convertase 9/genética , Pró-Proteína Convertase 9/metabolismo , Receptores de LDL/metabolismo , Sofosbuvir/farmacologia , Sofosbuvir/uso terapêutico , Proteína de Ligação a Elemento Regulador de Esterol 2/genética , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo
6.
Sci Rep ; 8(1): 8222, 2018 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-29844473

RESUMO

Despite decades of investigation on the proliferation of adult human primary hepatocytes, their expansion in vitro still remains challenging. To later be able to consider hepatocytes as a cell therapy alternative or bridge to liver transplantation, dramatically impeded by a shortage in liver donors, the first step is having an almost unlimited source of these cells. The banking of transplantable hepatocytes also implies a protocol for their expansion that can be compatible with large-scale production. We show that adult human primary hepatocytes when grown in 3D organoids are easily amplified, providing a substantial source of functional hepatocytes ready for transplantation. Following their plating, differentiated human hepatocytes are amplified during a transient and reversible step as liver progenitors, and can subsequently be converted back to mature differentiated hepatocytes. The protocol we propose is not only compatible with automated and high-throughput cell culture systems, thanks to the expansion of hepatocytes in suspension, but also guarantees the generation of a high number of functional cells from the same patient sample, with a relatively easy set up.


Assuntos
Hepatócitos/citologia , Organoides/citologia , Células-Tronco/citologia , Adulto , Idoso , Diferenciação Celular , Células Cultivadas , Colágeno , Combinação de Medicamentos , Feminino , Humanos , Laminina , Masculino , Proteoglicanas , Engenharia Tecidual
7.
Transplantation ; 101(1): 12-16, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27653295

RESUMO

"When Stem Cells Meet Immunology" has been the topic of the 21st annual "Nantes Actualités en Transplantation" meeting (June 9-10, 2016, Nantes, France). This meeting brought together pioneers and leading experts in the fields of stem cells, biomaterials and immunoregulation. Presentations covered multipotent (mesenchymal and hematopoietic) and pluripotent stem cells (embryonic and induced) for regenerative medicine of incurable diseases, immunotherapy and blood transfusions. An additional focus had been immune rejections and responses of allogeneic or autologous stem cells. Conversely, stem cells are also able to directly modulate the immune response through the production of immunoregulatory molecules. Moreover, stem cells may also provide an unlimited source of immune cells (DCs, NK cells, B cells, and T cells) that can operate as "super" immune cells, for example, through genetic engineering with chimeric antigen receptors.This meeting report puts presentations into an overall context highlighting new potential biomarkers for potency prediction of mesenchymal stem cell-derived and pluripotent stem cell-derived multicellular organoids. Finally, we propose future directions arising from the flourishing encounter of stem cell and immune biology.


Assuntos
Alergia e Imunologia , Pesquisa Biomédica/métodos , Medicina Regenerativa/métodos , Pesquisa com Células-Tronco , Transplante de Células-Tronco , Células-Tronco/imunologia , Animais , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto , Humanos
8.
Curr Gene Ther ; 15(5): 447-59, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26264708

RESUMO

BACKGROUND: Polarized airway epithelial cell cultures modelling Cystic Fibrosis Transmembrane conductance Regulator (CFTR) defect are crucial for CF and biomedical research. RNA interference has proven its value to generate knockdown models for various pathologies. More recently, genome editing using CRISPR-Cas9 artificial endonuclease was a valuable addition to the toolbox of gene inactivation. METHODS: Calu-3 cells and primary HAECs were transduced with HIV-1-derived lentiviral vectors (LVV) encoding small hairpin RNA (shRNA) sequence or CRISPR-Cas9 components targeting CFTR alongside GFP. After sorting of GFP-positive cells, CFTR expression was measured by RT-qPCR and Western blot in polarized or differentiated cells. CFTR channel function was assessed in Ussing chambers. Il-8 secretion, proliferation and cell migration were also studied in transduced cells. RESULTS: shRNA interference and CRISPRCas9 strategies efficiently decreased CFTR expression in Calu-3 cells. Strong CFTR knockdown was confirmed at the functional level in CRISPR-Cas9-modified cells. CFTR-specific shRNA sequences did not reduce gene expression in primary HAECs, whereas CRISPR-Cas9-mediated gene modification activity was correlated with a reduction of transepithelial secretion and response to a CFTR inhibitor. CFTR inactivation in the CRISPR-Cas9-modified Calu-3 cells did not affect migration and proliferation but slightly increased basal interleukin-8 secretion. CONCLUSION: We generated CFTR inactivated cell lines and demonstrated that CRISPR-Cas9 vectorised in a single LVV efficiently promotes CFTR inactivation in primary HAECs. These results provide a new protocol to engineer CF primary epithelia with their isogenic controls and pave the way for manipulation of CFTR expression in these cultures.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Células Epiteliais/metabolismo , Vetores Genéticos/genética , Lentivirus/genética , Interferência de RNA/fisiologia , Sistema Respiratório/metabolismo , Linhagem Celular , Movimento Celular/genética , Proliferação de Células/genética , Fibrose Cística/genética , Expressão Gênica/genética , Terapia Genética/métodos , Genoma/genética , Humanos , Interleucina-8/genética , RNA Interferente Pequeno/genética
9.
Liver Transpl ; 21(6): 801-11, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25821167

RESUMO

Crigler-Najjar type 1 disease is a rare inherited metabolic disease characterized by high levels of unconjugated bilirubin due to the complete absence of hepatic uridine diphosphoglucuronate-glucuronosyltransferase activity. Hepatocyte transplantation (HT) has been proposed as an alternative treatment for Crigler-Najjar syndrome, but it is still limited by the quality and the low engraftment and repopulation ability of the cells used. Because of their attachment capability and expression of adhesion molecules as well as the higher proportion of hepatic progenitor cells, neonatal hepatocytes may have an advantage over adult cells. Adult or neonatal hepatocytes were transplanted into Gunn rats, a model for Crigler-Najjar disease. Engraftment and repopulation were studied and compared by immunofluorescence (IF). Additionally, the serum bilirubin levels, the presence of bilirubin conjugates in rat serum, and the expression of uridine diphosphate glucuronosyltransferase 1 family polypeptide A1 (UGT1A1) in rat liver samples were also analyzed. Here we show that neonatal HT results in long-term correction in Gunn rats. In comparison with adult cells, neonatal cells showed better engraftment and repopulation capability 3 days and 6 months after transplantation, respectively. Bilirubinemia decreased in the transplanted animals during the whole experimental follow-up (6 months). Bilirubin conjugates were also present in the serum of the transplanted animals. Western blots and IF confirmed the presence and expression of UGT1A1 in the liver. This work is the first to demonstrate the advantage of using neonatal hepatocytes for the treatment of Crigler-Najjar in vivo.


Assuntos
Síndrome de Crigler-Najjar/terapia , Hepatócitos/transplante , Regeneração Hepática , Idoso , Idoso de 80 Anos ou mais , Animais , Bilirrubina/sangue , Proliferação de Células , Feminino , Glucuronosiltransferase/metabolismo , Humanos , Recém-Nascido , Fígado/metabolismo , Masculino , Pessoa de Meia-Idade , Propranolol , Ratos Gunn
10.
Hum Mol Genet ; 24(8): 2287-96, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25561689

RESUMO

Glycogen storage disease type 1a (GSD1a) is a rare disease due to the deficiency in the glucose-6-phosphatase (G6Pase) catalytic subunit (encoded by G6pc), which is essential for endogenous glucose production. Despite strict diet control to maintain blood glucose, patients with GSD1a develop hepatomegaly, steatosis and then hepatocellular adenomas (HCA), which can undergo malignant transformation. Recently, gene therapy has attracted attention as a potential treatment for GSD1a. In order to maintain long-term transgene expression, we developed an HIV-based vector, which allowed us to specifically express the human G6PC cDNA in the liver. We analysed the efficiency of this lentiviral vector in the prevention of the development of the hepatic disease in an original GSD1a mouse model, which exhibits G6Pase deficiency exclusively in the liver (L-G6pc(-/-) mice). Recombinant lentivirus were injected in B6.G6pc(ex3lox/ex3lox). SA(creERT2/w) neonates and G6pc deletion was induced by tamoxifen treatment at weaning. Magnetic resonance imaging was then performed to follow up the development of hepatic tumours. Lentiviral gene therapy restored glucose-6 phosphatase activity sufficient to correct fasting hypoglycaemia during 9 months. Moreover, lentivirus-treated L-G6pc(-/-) mice presented normal hepatic triglyceride levels, whereas untreated mice developed steatosis. Glycogen stores were also decreased although liver weight remained high. Interestingly, lentivirus-treated L-G6pc(-/-) mice were protected against the development of hepatic tumours after 9 months of gene therapy while most of untreated L-G6pc(-/-) mice developed millimetric HCA. Thus the treatment of newborns by recombinant lentivirus appears as an attractive approach to protect the liver from the development of steatosis and hepatic tumours associated to GSD1a pathology.


Assuntos
Terapia Genética , Doença de Depósito de Glicogênio Tipo I/genética , Doença de Depósito de Glicogênio Tipo I/terapia , Lentivirus/genética , Neoplasias Hepáticas/prevenção & controle , Animais , Modelos Animais de Doenças , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Glucose-6-Fosfatase/genética , Glucose-6-Fosfatase/metabolismo , Doença de Depósito de Glicogênio Tipo I/complicações , Doença de Depósito de Glicogênio Tipo I/enzimologia , Humanos , Lentivirus/metabolismo , Fígado/enzimologia , Neoplasias Hepáticas/etiologia , Camundongos , Camundongos Knockout
11.
Hum Gene Ther Methods ; 25(3): 181-6, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24593043

RESUMO

Helper-dependent adenoviral (HDAd) vectors are attractive for liver-directed gene therapy because they can drive sustained high levels of transgene expression without chronic toxicity. However, high vector doses are required to achieve efficient hepatic transduction by systemic delivery because of a nonlinear dose response. Unfortunately, such high doses result in systemic vector dissemination and dose-dependent acute toxicity with potential lethal consequences. We have previously shown in nonhuman primates that delivery of HDAd in surgically isolated livers resulted in a significantly higher hepatic transduction with reduced systemic vector dissemination compared with intravenous delivery and multiyear transgene expression. Encouraged by these data, we have now employed a surgical vector delivery method in the Gunn rat, an animal model for Crigler-Najjar syndrome. After vector delivery into the surgically isolated liver, we show phenotypic correction at the low and clinically relevant vector dose of 1 × 10(11) vp/kg. Correction of hyperbilirubinemia and increased glucuronidation of bilirubin in bile was achieved for up to 1 year after vector administration. Surgical delivery of the vector was well tolerated without signs of acute or chronic toxicity. This method of delivery could thereby be a safer alternative to liver transplantation for long-term treatment of Crigler-Najjar syndrome type I.


Assuntos
Adenoviridae/genética , Vetores Genéticos/metabolismo , Hiperbilirrubinemia/terapia , Animais , Bilirrubina/sangue , Síndrome de Crigler-Najjar/terapia , Terapia Genética , Vetores Genéticos/genética , Glucuronosiltransferase/genética , Humanos , Fígado/metabolismo , Fígado/cirurgia , Regiões Promotoras Genéticas , Ratos , Ratos Gunn , Transdução Genética
12.
PLoS One ; 7(12): e51952, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23251659

RESUMO

Precise control of transgene expression in a tissue-specific and temporally regulated manner is desirable for many basic and applied investigations gene therapy applications. This is important to regulate dose of transgene products and minimize unwanted effects. Previously described methods have employed tissue specific promoters, miRNA-based transgene silencing or tetR-KRAB-mediated suppression of transgene promoters. To improve on versatility of transgene expression control, we have developed expression systems that use combinations of a tetR-KRAB artificial transgene-repressor, endogenous miRNA silencing machinery and tissue specific promoters. Precise control of transgene expression was demonstrated in liver-, macrophage- and muscle-derived cells. Efficiency was also demonstrated in vivo in murine muscle. This multicomponent and modular regulatory system provides a robust and easily adaptable method for achieving regulated transgene expression in different tissue types. The improved precision of regulation will be useful for many gene therapy applications requiring specific spatiotemporal transgene regulation.


Assuntos
Vetores Genéticos/genética , Fatores de Transcrição Kruppel-Like/genética , MicroRNAs/genética , Proteínas Repressoras/genética , Transcrição Gênica , Transgenes , Animais , Linhagem Celular , Linhagem Celular Tumoral , Inativação Gênica , Células HEK293 , Humanos , Fígado/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Células Musculares/metabolismo , Regiões Promotoras Genéticas
13.
Curr Pharm Des ; 17(24): 2516-27, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21774771

RESUMO

Retroviral vectors have been used for several decades for the transfer of therapeutic genes to various cells or organs including the liver. Initial studies aimed at treating inherited liver deficiencies were carried out with murine oncoretroviral vectors either delivered directly to the organ or using an ex vivo strategy that entailed harvest of the hepatocytes, transduction during a culture phase and further reinfusion to the patient. However, although a clinical trial was performed in the early 1990s, a complete cure of animal models of metabolic diseases was rarely achieved. The advent of lentiviral vectors derived from HIV1 profoundly changed the field and this vector type now appears to be of the most attractive for liver directed gene therapy. Indeed, lentiviral vectors do not require complete cell division to transduce the target cells. There are however still bottlenecks that limit the clinical development of gene therapy using retroviral vectors. In the present review we will specifically focus on specific aspects such as the risk of insertional mutagenesis, the potential requirement of cell cycle activation to enhance transduction and the major issue of an immune response directed against the transgene as well as some specific aspects of ex vivo gene transfer. Finally we will briefly consider the future developments of these vectors made possible by the availability of new techniques in cell and molecular biology.


Assuntos
Terapia Genética/métodos , Doenças Metabólicas/terapia , Retroviridae/genética , Animais , Ciclo Celular , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Lentivirus/genética , Fígado/metabolismo , Biologia Molecular/métodos , Transgenes
14.
Gastroenterology ; 139(3): 999-1007, 1007.e1-2, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20546738

RESUMO

BACKGROUND & AIMS: Crigler-Najjar type 1 (CN-I) is an inherited liver disease caused by an absence of bilirubin-uridine 5'-diphosphate-glucuronosyltransferase (UGT1A1) activity. It results in life-threatening levels of unconjugated bilirubin, and therapeutic options are limited. We used adult Gunn rats (an animal model of the disease) to evaluate the efficiency of lentiviral-based gene therapy to express UGT1A1 in liver. METHODS: Gunn rats were given intraportal injections of VSVG-pseudotyped lentiviral vectors that encode UGT1A1 under the control of a liver-specific transthyretin promoter (mTTR.hUGT1A1); this vector does not contain target sequences for miR-142, a microRNA that is expressed specifically in hematopoietic cells. Rats were also injected with the vector mTTR.hUGT1A1.142T, which contains 4 copies of the miR-142 target sequences; its messenger RNA should be degraded in antigen-presenting cells. Bilirubinemia was monitored, and the presence of transduced hepatocytes was analyzed by quantitative polymerase chain reaction. Vector expression was tested in vitro in rat hematopoietic cells. RESULTS: In Gunn rats, bilirubin levels normalized 2 weeks after administration of mTTR.hUGT1A1. However, hyperbilirubinemia resumed 8 weeks after vector administration, concomitant with the induction of an immune response. In contrast, in rats injected with mTTR-UGT1A1.142T, bilirubin levels normalized for up to 6 months and transduced cells were not eliminated. CONCLUSIONS: Lentiviral vectors that express UGT1A1 reduce hyperbilirubinemia in immunocompetent Gunn rats for at least 6 months. The immune response against virally expressed UGT1A1 can be circumvented by inclusion of miR-142 target sequences, which reduce vector expression in antigen-presenting cells. This lentiviral-based gene therapy approach might be developed to treat patients with CN-I.


Assuntos
Síndrome de Crigler-Najjar/terapia , Terapia Genética/métodos , Vetores Genéticos , Glucuronosiltransferase/genética , Lentivirus/genética , Fígado/enzimologia , MicroRNAs/metabolismo , Animais , Anticorpos/sangue , Células Apresentadoras de Antígenos/imunologia , Bilirrubina/sangue , Biomarcadores/sangue , Síndrome de Crigler-Najjar/enzimologia , Síndrome de Crigler-Najjar/genética , Síndrome de Crigler-Najjar/imunologia , Modelos Animais de Doenças , Glucuronosiltransferase/biossíntese , Glucuronosiltransferase/imunologia , Células HeLa , Humanos , Masculino , Pré-Albumina/genética , Regiões Promotoras Genéticas , Estabilidade de RNA , RNA Mensageiro/metabolismo , Ratos , Ratos Gunn , Fatores de Tempo , Transdução Genética
15.
Methods Mol Biol ; 597: 109-25, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20013229

RESUMO

Lentiviral vectors are now well recognized as good vehicles for gene delivery. This is because they can efficiently transduce both dividing and post-mitotic cells, and stably integrate into the host genome allowing for long-term expression of the transgene. Their potential utility for the generation of transgenic animals has been recognized as an attractive and promising alternative to the conventional DNA-microinjection method which lacks efficiency. The initial success of lentiviral transgenesis in mice considerably broadened its use in other species, in which classical transgenic techniques are difficult, such as in the rat.In this chapter, we describe detailed procedures for both the production of human immunodeficiency virus-1 (HIV-1)-derived lentiviral vectors and for the generation of transgenic rats by injection of these vectors into the perivitelline space of fertilized one-cell eggs.


Assuntos
Vetores Genéticos/genética , Lentivirus/genética , Ratos Transgênicos/genética , Animais , Linhagem Celular , Transferência Embrionária/métodos , Embrião de Mamíferos/citologia , Feminino , Técnicas de Transferência de Genes , HIV-1/genética , Humanos , Glicoproteínas de Membrana/genética , Microinjeções/métodos , Ratos , Ratos Transgênicos/virologia , Transdução Genética/métodos , Transfecção/métodos , Proteínas do Envelope Viral/genética
16.
J Gene Med ; 11(8): 689-96, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19455564

RESUMO

BACKGROUND: In vivo adeno-associated virus (AAV) delivery to adult liver results in sustained expression of the transgene. However, it has been suggested that AAV delivery to the newborn liver may result in transient expression. In the present study, we analysed transgene expression after AAV8 delivery of a therapeutic or a marker gene to newborn rat liver. METHODS: Recombinant AAV 8 vectors carrying either the human UGT1A1 cDNA or the lacZ gene were injected intravenously in 2-day-old Gunn or Wistar rats. Serum bilirubin level was recorded in Gunn rats and beta-galactosidase expression was monitored by immunohistochemistry or enzyme activity. The molecular forms of AAV genome were analysed by the polymerase chain reaction and Southern blotting in whole liver and by the quantitative polymerase chain reaction in macroscopically dissected beta-galactosidase clusters. RESULTS: In Gunn rat, complete serum bilirubin normalization occurred after AAV delivery but hyperbilirubinemia resumed thereafter. Similarly, beta-galactosidase expression was maximum at day 7, but only a few (less than 1%) beta-galactosidase positive cells were recorded at 1 or 3 months. These cells gathered in small clusters and the AAV copy number was 75-fold higher in positive cell clusters than in the surrounding parenchyma. CONCLUSIONS: The results obtained in the present study show that in vivo AAV delivery to newborn rats results in transient expression in most hepatocytes. Expression of the trangene was persistent in small clusters of cells and preliminary data support the hypothesis that integration of the viral genome occurs in these clusters. Altogether, our data confirm the low efficiency of AAV vectors for gene therapy of liver diseases when delivered in the newborn period.


Assuntos
DNA Recombinante/metabolismo , Dependovirus/genética , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Fígado/metabolismo , Animais , Animais Recém-Nascidos , Bilirrubina/sangue , Southern Blotting , Ensaio de Imunoadsorção Enzimática , Vetores Genéticos/administração & dosagem , Glucuronosiltransferase/metabolismo , Humanos , Injeções , Reação em Cadeia da Polimerase , Ratos , Ratos Gunn , Fatores de Tempo , Integração Viral , beta-Galactosidase/metabolismo
17.
Curr Gene Ther ; 9(2): 136-49, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19355871

RESUMO

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.


Assuntos
Terapia Genética/métodos , Vetores Genéticos/genética , Hepatócitos/transplante , Doenças Metabólicas/terapia , Retroviridae/genética , Animais , Vetores Genéticos/metabolismo , Humanos , Fígado/metabolismo , Hepatopatias/terapia , Retroviridae/metabolismo
18.
J Pediatr Surg ; 44(3): 517-22, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19302851

RESUMO

PURPOSE: Metabolic inherited liver diseases are attractive targets for gene therapy. Recombinant lentiviruses are very powerful viral vectors able to infect nonmitotic cells. We wanted to develop a new surgical approach to improve gene transfer in adult liver using low viral doses. MATERIALS AND METHODS: Adult rats were injected with 2.108 infectious particles of lentiviral vectors encoding the green fluorescent protein marker gene under control of a liver-specific promoter transthyretin. In the control group (n = 5), gene delivery was performed by inflow intraportal injection. In the surgical group (n = 5), liver was completely excluded from systemic circulation before viral injection in infrahepatic vena cava with high pressure. RESULTS: At day 9, transduction efficiency was 14.35% in the surgical group 3 and 0.39% in the control group (P = .016). At month 2, the number of transduced hepatocytes decreased in the most part of rats, except in half of rats in the surgical group. Antibodies against green fluorescent protein were detected in all rats at month 2, except one in the surgical group. CONCLUSIONS: We developed a new surgical approach allowing an efficient transduction of hepatocytes in adult rats using lentivirus at low viral doses. We have now to control the immune response to permit long-term expression of transgene.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos , Lentivirus/genética , Animais , Contagem de Células , Células Cultivadas , Feminino , Proteínas de Fluorescência Verde/genética , Hepatócitos/metabolismo , Imuno-Histoquímica , Fígado/irrigação sanguínea , Circulação Hepática , Masculino , Plasmídeos , Ratos , Ratos Gunn , Transdução Genética , Veias Cavas
19.
Expert Rev Gastroenterol Hepatol ; 1(1): 155-71, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19072443

RESUMO

The liver is the site of numerous metabolic inherited diseases. It has unique features that make it compliant to various gene therapy approaches. Many vector types and gene delivery strategies have been evaluated during the past 20 years in a number of animal models of metabolic liver diseases. However, the complete cure of inherited liver deficiencies by gene therapy in relevant animal models were only reported recently. These successes were achieved thanks to major advances in vector technology. In this review, we will focus on Crigler-Najjar disease and hereditary tyrosinemia, two paradigmatic examples of the two categories of enzymatic liver deficiencies: type I, in which the genetic defect does not affect liver histology; and type II, in which liver lesions are present.


Assuntos
Síndrome de Crigler-Najjar/terapia , Terapia Genética , Tirosinemias/terapia , Animais , Modelos Animais de Doenças , Glucuronosiltransferase/deficiência , Humanos , Tirosina Transaminase/deficiência
20.
J Hepatol ; 43(6): 1031-7, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16169114

RESUMO

BACKGROUND/AIMS: Liver failure is a life threatening condition currently treated by palliative measures and, when applicable, organ transplantation. The use of a bioartificial organ capable of fulfilling the main functions of the liver would represent an attractive alternative. However, the shortage of suitable donor cells, and their limited growth ability have impeded the development of this strategy. We investigated whether lentiviral vectors allow for conditional immortalization of human hepatocytes and whether these immortalized hepatocytes could reverse lethal acute liver failure. METHODS: We exposed primary human hepatocytes to Cre-excisable lentiviral vectors coding for SV40T Antigen, telomerase, and/or Bmi-1 and tested the functionality of the resulting cell lines. Therapeutic potential of immortalized hepatocytes were tested in a murine model of acetaminophen-induced hepatic injury. RESULTS: The immortalized hepatocytes grew continuously yet were non-tumorigenic, stopped proliferating when exposed to Cre recombinase, and conserved defining properties of primary hepatocytes, including the ability to secrete liver-specific proteins and to detoxify drugs. The implantation of encapsulated immortalized human hepatocytes rescued mice from lethal doses of acetaminophen. CONCLUSIONS: Lentiviral vectors represent tools of choice for immortalization of non-dividing primary cells, and lentivirally immortalized human hepatocytes are promising reagents for cell-based therapy of acute liver failure.


Assuntos
Acetaminofen/efeitos adversos , Analgésicos não Narcóticos/efeitos adversos , Transplante de Células/métodos , Falência Hepática Aguda/terapia , Fígado Artificial , Adulto , Animais , Linhagem Celular Transformada , Modelos Animais de Doenças , Vetores Genéticos , Hepatócitos/transplante , Humanos , Lentivirus , Falência Hepática Aguda/induzido quimicamente , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA