Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Hepatology ; 49(3): 950-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19152424

ABSTRACT

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.


Subject(s)
Apolipoprotein A-II/metabolism , Cell Transplantation/methods , Embolization, Therapeutic/methods , Hepatocytes/metabolism , Hepatocytes/transplantation , Animals , Apolipoprotein A-II/genetics , Cell Proliferation , Gene Expression Regulation , Genetic Therapy , Hepatocytes/pathology , Humans , Lentivirus/genetics , Liver/metabolism , Liver/pathology , Liver/surgery , Liver Regeneration/physiology , Macaca mulatta , Models, Animal , Portal Vein/surgery , Transgenes/genetics
2.
Curr Gene Ther ; 9(2): 136-49, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19355871

ABSTRACT

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.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/genetics , Hepatocytes/transplantation , Metabolic Diseases/therapy , Retroviridae/genetics , Animals , Genetic Vectors/metabolism , Humans , Liver/metabolism , Liver Diseases/therapy , Retroviridae/metabolism
3.
Liver Transpl ; 15(1): 7-14, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19109838

ABSTRACT

More than 30 years after the first hepatocyte transplant to treat the Gunn rat, the animal model for Crigler-Najjar syndrome, there are still a number of impediments to hepatocyte transplantation. Numerous animal models are still used in work aimed at improving hepatocyte engraftment and/or long-term function. Although other cell sources, particularly hepatic and extrahepatic stem cells, are being explored, adult hepatocytes remain the cells of choice for the treatment of liver diseases by cell therapy. In recent years, diverse approaches have been developed in various animal models to enhance hepatocyte transduction and amplification in vitro and cell engraftment and functionality in vivo. They have led to significant progress in hepatocyte transplantation for the treatment of patients with metabolic diseases and for bridging patients with acute injury until their own livers regenerate. This review presents and considers the results of this work with a special emphasis on procedures that might be clinically applicable.


Subject(s)
Hepatocytes/cytology , Animals , Cell Transplantation , Cell- and Tissue-Based Therapy , Crigler-Najjar Syndrome/pathology , Disease Models, Animal , Humans , Liver/pathology , Liver Regeneration , Mice , Rats , Stem Cells/cytology , Syndrome , Tissue Engineering/methods
4.
Methods Mol Biol ; 481: 83-96, 2009.
Article in English | MEDLINE | ID: mdl-19096791

ABSTRACT

The poor hepatocyte engraftment efficiency and the low level of their expansion in the host liver are a major limitation to cell therapy for the treatment of life-threatening liver diseases. Many rodent models have shown that liver repopulation via transplanted hepatocytes occurs only when liver growth capacity is impaired for an extended period of time. However, these models are not transposable to the clinics and to date there is no safe method to achieve this result in a clinical setting.Therefore, it is necessary to define on large animal models strategies that provide to transplanted hepatocytes sufficient proliferation stimuli to induce their division and that could permit a direct extrapolation to humans. Such procedures should be transposable to patients. We have defined a protocol of liver partial portal branch embolisation and shown that it induces the proliferation of transplanted hepatocytes in non-human primates (Macaca mulatta). This animal model is also appropriate to evaluate the lentiviral-mediated ex vivo gene therapy approach, since simian hepatocytes are efficiently transduced by HIV-1-derived lentivirus vectors.


Subject(s)
Hepatocytes/transplantation , Liver Transplantation/methods , Macaca mulatta , Models, Animal , Animals , Cell Culture Techniques , Cell Separation , Hemostasis, Surgical/methods , Hepatocytes/cytology , Liver/blood supply , Liver/cytology , Models, Biological , Portal Vein/surgery , Retroviridae/genetics , Tissue and Organ Harvesting/methods , Transduction, Genetic/methods
5.
Transplantation ; 82(8): 1067-73, 2006 Oct 27.
Article in English | MEDLINE | ID: mdl-17060856

ABSTRACT

BACKGROUND: Hepatocyte transplantation could be an alternative to whole liver transplantation for the treatment of metabolic liver diseases. However, the results of clinical investigations suggest that the number of engrafted hepatocytes was insufficient to correct metabolic disorders. This may partly result from a lack of proliferation of transplanted hepatocytes. In rodents, portal ligation enhances hepatocyte engraftment after transplantation. We investigated the effects of partial portal ligation and embolization on engraftment and proliferation of transplanted hepatocytes in primates. METHODS: Hepatocyte autotransplantation was performed in Macaca monkeys. The left lateral lobe was resected for hepatocyte isolation. The first group of monkeys underwent surgical ligation of the left and right anterior portal branches; in the second group, the same portal territories were obstructed by embolization with biological glue. To evaluate the proportion of cell engraftment hepatocytes were Hoechst-labeled and transplanted via the portal vein. Cell proliferation was measured by BrdU incorporation. RESULTS: Hepatocyte proliferation was induced by both procedures but it was significantly higher after partial portal embolization (23.5% and 11.2% of dividing hepatocytes on days 3 and 7) than after ligation (3% and 0.8%). Hepatocytes engrafted more efficiently after embolization than after ligation. They proliferated and participated to liver regeneration representing 10% of the liver mass on day seven and their number remained constant on day 15. CONCLUSIONS: These data suggest that partial portal embolization of the recipient liver improves engraftment of transplanted hepatocytes in a primate preclinical model providing a new strategy for hepatocyte transplantation.


Subject(s)
Cell Transplantation/methods , Embolization, Therapeutic , Hepatocytes/cytology , Portal Vein/pathology , Animals , Bromodeoxyuridine/pharmacology , Cell Proliferation , Hepatocytes/metabolism , Liver/pathology , Liver Diseases/therapy , Liver Regeneration , Macaca , Primates , Treatment Outcome
6.
Transplantation ; 100(2): 344-54, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26757049

ABSTRACT

BACKGROUND: Hepatocyte transplantation has been proposed as an alternative to orthotopic liver transplantation to treat metabolic liver diseases. This approach requires preconditioning of the host liver to enhance engraftment of transplanted hepatocytes. Different methods are currently used in preclinical models: partial hepatectomy, portal ligature or embolization, and radiotherapy or chemotherapeutic drugs. However, these methods carry high risks of complications and are problematic for use in clinical practice. Here, we developed an innovative method called volumetric (distal, partial, and random) portal embolization (VPE), which preserves total liver volume. METHODS: Embolization was performed in the portal trunk of C57BL6 adult mice with polyester microspheres, to ensure a bilateral and distal distribution. The repartition of microspheres was studied by angiographic and histological analyses. Liver regeneration was evaluated by Ki67 labeling. Optimal conditions for VPE were determined, and the resulting regeneration was compared with that after partial hepatectomy (70%). Labeled adult hepatocytes were then transplanted, and engraftment was compared between embolized (n = 19) and nonembolized mice (n = 8). Engraftment was assessed in vivo and histologically by tracking labeled cells at day 5. RESULTS: The best volumetric embolization conditions, which resulted in the regeneration of 5% of total liver, were 8 × 10 ten-micron microspheres infused with a 29 G needle directly into the portal trunk at 3.3 µL/s. In these conditions, transplanted hepatocytes engraftment was significantly higher than that in control conditions (3 vs 0.65%). CONCLUSIONS: The VPE is a new, minimally invasive, and efficient technique to prepare the host liver for cell transplantation.


Subject(s)
Embolization, Therapeutic/methods , Hepatocytes/transplantation , Liver Regeneration , Liver/blood supply , Polyesters/administration & dosage , Portal Vein , Animals , Biomarkers/metabolism , Cell Survival , Cell Tracking , Female , Graft Survival , Hepatectomy/methods , Hepatocytes/metabolism , Hepatocytes/pathology , Injections, Intravenous , Ki-67 Antigen/metabolism , Liver/metabolism , Liver/pathology , Liver/physiopathology , Liver/surgery , Male , Mice, Inbred C57BL , Microspheres , Organ Size , Portal Vein/diagnostic imaging , Radiography , Time Factors
7.
Mol Endocrinol ; 18(3): 733-46, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14694083

ABSTRACT

The mechanisms of the basolateral targeting of G protein-coupled receptors remain largely unknown. Mutagenesis experiments have allowed us to identify the basolateral sorting signals of the TSH and LH receptors expressed in Madin-Darby canine kidney cells and thyroid follicular FRT cells. Unexpectedly these signals (amino acids 731-746 and 672-689, respectively) share an unusual localization in the distal part of the intracellular domain of the receptors at a marked distance from the membrane. When grafted onto the p75-neurotropin receptor, these signals redirect this normally apically expressed protein to the basolateral cell surface. They are independent of the endocytosis signal. The basolateral sorting signals of TSH, LH, and FSH receptors do not exhibit primary sequence homology with each other or with any other known signal. Furthermore, circular dichroism studies show that the three signals exhibit distinct secondary structures. The TSH receptor has a stable helical structure, the LH receptor has both helix and beta-sheet structures, and the FSH receptor sorting signal has a main random coil structure. This means that even in closely-related receptors different secondary structures can be found for basolateral signals unrelated to internalization signals. This observation contrasts with what is known about basolateral signals related to internalization signals for which a common beta-turn structure has been described. Deletion of the basolateral sorting signals results in apical targeting of the receptors, suggesting the existence of apical sorting information. However, a soluble form of the TSH receptor, which harbors all N- and putative O-linked oligosaccharides, is secreted in a nonpolarized fashion. This implies that apical sorting information must be located elsewhere, either in the transmembrane or in the intracellular domains of the receptor.


Subject(s)
Protein Sorting Signals/physiology , Receptors, LH/chemistry , Receptors, LH/metabolism , Receptors, Thyrotropin/chemistry , Receptors, Thyrotropin/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Circular Dichroism , Dogs , GTP-Binding Protein alpha Subunits, Gs/metabolism , Hydrophobic and Hydrophilic Interactions , Intracellular Space/metabolism , Molecular Sequence Data , Protein Conformation , Rats , Receptor, Nerve Growth Factor/genetics , Receptor, Nerve Growth Factor/metabolism , Receptors, FSH/chemistry , Receptors, FSH/metabolism , Receptors, LH/genetics , Receptors, Thyrotropin/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tyrosine/chemistry , Tyrosine/metabolism
8.
Transplantation ; 99(1): 36-40, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25340599

ABSTRACT

INTRODUCTION: Hepatocyte transplantation could be an alternative to liver transplantation for the treatment of metabolic diseases. However, rodent models have shown that engraftment of transplanted cells in the liver is low and requires deposition of cells in hepatic sinusoids. Splanchnic vasodilatators improved hepatocyte engraftment in a rat model. Therefore, we investigated the effect of glyceryl trinitrate (GTN) on the efficacy of cell engraftment and on liver repopulation in the mdr2-knockout mouse, a model for progressive familial intrahepatic cholestasis type 3. METHODS: Congenic normal mdr2 (+/+) hepatocytes were isolated by two-step collagenase perfusion and transplanted into mdr2(-/-) mice livers through the portal vein in the presence or absence of GTN. Liver repopulation was assessed by immunohistochemistry, and transplanted hepatocyte function was assessed at different times after transplantation by measurement of biliary lipid secretion and quantification of fibrosis. RESULTS: The number of engrafted cells in GTN-treated mice was significantly higher than that in control mice, and transplanted hepatocytes were found in a greater number of distal sinusoids. Levels of phospholipid secretion were significantly higher than those in the control group 3 months after hepatocyte transplantation (18.3 ± 2.3 vs. 5.2 ± 3.9 nmol/min/100 g, P < 0.0001), and the ratio of phospholipids to bile salt was greater (6.8 ± 1.3 vs. 3.2 ± 1.6, P = 0.03). The percentage area of liver fibrosis was also significantly reduced in GTN-treated mice (5.7% ± 2.3% vs. 12.4% ± 2.9%, P = 0.016). CONCLUSION: The use of GTN improves hepatocyte engraftment and correction of metabolic disease in mdr2 (-/-) mice. This approach might be beneficial in hepatocyte transplantation for the treatment of patients with liver diseases.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/deficiency , Cholestasis, Intrahepatic/surgery , Hepatocytes/drug effects , Hepatocytes/transplantation , Liver Transplantation/methods , Nitroglycerin/pharmacology , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Bile Acids and Salts/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cholestasis, Intrahepatic/genetics , Cholestasis, Intrahepatic/metabolism , Disease Models, Animal , Hepatocytes/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/surgery , Male , Mice, Knockout , Phospholipids/metabolism , Time Factors , ATP-Binding Cassette Sub-Family B Member 4
9.
Cell Med ; 4(2): 85-98, 2012 Feb.
Article in English | MEDLINE | ID: mdl-26858856

ABSTRACT

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.
Hum Mol Genet ; 16(22): 2683-92, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17720887

ABSTRACT

Heterozygous mutations in JAGGED1, encoding a single-pass transmembrane ligand for the Notch receptors, cause Alagille syndrome (AGS), a polymalformative disorder affecting the liver, heart, eyes and skeleton and characterized by a peculiar facies. Most of the JAGGED1 mutations generate premature termination codons, and as a result, two pathogenic mechanisms causing AGS have been proposed: haploinsufficiency or a dominant-negative effect of putative truncated proteins. To determine whether missense or protein-truncating mutations in JAGGED1 can lead to the synthesis and function of abnormal proteins, we performed cell culture experiments. We showed that human JAGGED1 undergoes a metalloprotease-dependent cleavage resulting in the shedding of its extracellular domain and that this domain seems able to fulfill a biological function in vitro, probably by antagonizing Notch signaling. Moreover, the soluble form of JAGGED1 was able to compete with the transmembrane ligand. Mutant proteins with missense or nonsense mutations were synthesized and gave rise to a chord-like phenotype and a migration defect when expressed by stably transfected cells. These chord-like structures were similar to the phenotype exhibited by fibroblasts isolated from a fetus with a protein-truncating mutation. Results obtained from Notch signaling inhibition and Notch reporter assays showed that this chord-like phenotype, exhibited by mutant JAGGED1 transfectants, may result from an inhibitory effect on Notch signaling. Altogether, our results favor a dominant-negative mechanism of some JAGGED1 mutations in AGS.


Subject(s)
Alagille Syndrome/metabolism , Calcium-Binding Proteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Mutation/genetics , Receptors, Notch/genetics , Signal Transduction , Animals , Blotting, Western , COS Cells , Cell Differentiation , Cell Line , Cell Movement , Chlorocebus aethiops , Fluorescent Antibody Technique , Humans , Jagged-1 Protein , Luciferases/metabolism , Mice , NIH 3T3 Cells , Receptors, Notch/metabolism , Serrate-Jagged Proteins , Transfection
11.
J Hepatol ; 45(1): 99-107, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16723167

ABSTRACT

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.


Subject(s)
HIV-1/genetics , Hepatocytes/physiology , Lentivirus/genetics , Actins/genetics , Animals , Apolipoprotein A-II/genetics , DNA Primers , Gene Transfer Techniques , Genetic Vectors , Hepatocytes/virology , Macaca fascicularis , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL