Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Am J Transplant ; 16(3): 1021-30, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26523372

ABSTRACT

Clinical hepatocyte transplantation is hampered by low engraftment rates and gradual loss of function resulting in incomplete correction of the underlying disease. Preconditioning with partial hepatectomy improves engraftment in animal studies. Our aim was to study safety and efficacy of partial hepatectomy preconditioning in clinical hepatocyte transplantation. Two patients with Crigler-Najjar syndrome type I underwent liver resection followed by hepatocyte transplantation. A transient increase of hepatocyte growth factor was seen, suggesting that this procedure provides a regenerative stimulus. Serum bilirubin was decreased by 50%, and presence of bilirubin glucuronides in bile confirmed graft function in both cases; however, graft function was lost due to discontinuation of immunosuppressive therapy in one patient. In the other patient, serum bilirubin gradually increased to pretransplant concentrations after ≈600 days. In both cases, loss of graft function was temporally associated with emergence of human leukocyte antigen donor-specific antibodies (DSAs). In conclusion, partial hepatectomy in combination with hepatocyte transplantation was safe and induced a robust release of hepatocyte growth factor, but its efficacy on hepatocyte engraftment needs to be evaluated with additional studies. To our knowledge, this study provides the first description of de novo DSAs after hepatocyte transplantation associated with graft loss.


Subject(s)
Antibody Formation/immunology , Crigler-Najjar Syndrome/immunology , Graft Rejection/etiology , HLA Antigens/immunology , Hepatectomy/adverse effects , Hepatocytes/transplantation , Liver Transplantation/adverse effects , Tissue Donors , Adolescent , Adult , Child , Crigler-Najjar Syndrome/surgery , Female , Humans , Infant , Male , Postoperative Complications , Prognosis
2.
Gene Ther ; 21(2): 168-74, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24285217

ABSTRACT

Adenoviral (AdV) and Adenovirus-associated viral (AAV) vectors both are used for in vivo gene therapy of inherited liver disorders, such as Crigler-Najjar syndrome type 1. In a relevant animal model, the Gunn rat, both vectors efficiently correct the severe hyperbilirubinemia characteristic of this liver disorder. Although the clinical use of AAV is more advanced, as demonstrated by the successful phase 1 trial in hemophilia B patients, because of its large cloning capacity AdV remains an attractive option. A direct comparison of the efficacy of these two vectors in the liver in a relevant disease model has not been reported. Aim of this study was to compare the efficiency of clinically applicable doses of both vectors in the Gunn rat. AdV or scAAV (self-complimentary AAV) ferrying identical liver-specific expression cassettes of the therapeutic gene, UGT1A1, were injected into the tail vein. As the titration methods of these two vectors are very different, a comparison based on vector titers is not valid. Therefore, their efficacy was compared by determining the amount of vector genomes delivered to the liver required for therapeutic correction of serum bilirubin. Like AAV, the liver-specific first-generation AdV also provided sustained correction in this relevant disease model. UGT1A1 mRNA expression provided per genome was comparable for both vectors. Flanking the expression cassette in AdV with AAV-ITRs (inverted terminal repeats), increased UGT1A1 mRNA expression eightfold which resulted in a significant improvement of efficacy. Compared with AAV, less AdV genomes were needed for complete correction of hyperbilirubinemia.


Subject(s)
Adenoviridae/genetics , Dependovirus/genetics , Genetic Vectors/adverse effects , Glucuronosyltransferase/genetics , Liver/metabolism , Liver/virology , Animals , Bilirubin/metabolism , Crigler-Najjar Syndrome/genetics , Crigler-Najjar Syndrome/therapy , Disease Models, Animal , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/administration & dosage , Glucuronosyltransferase/metabolism , HEK293 Cells , Humans , Liver/pathology , Male , RNA, Messenger/genetics , Rats , Rats, Gunn
3.
Cancer Gene Ther ; 16(7): 585-97, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19180144

ABSTRACT

Survival of patients with pancreatic cancer is poor. Adenoviral (Ad) gene therapy employing the commonly used serotype 5 reveals limited transduction efficiency due to the low amount of coxsackie-adenovirus receptor on pancreatic cancer cells. To identify fiber-chimeric adenoviruses with improved gene transfer, a library of Ad vectors based on Ad5 and carrying fiber molecules consisting of 16 other serotypes were transduced to human pancreatic carcinoma cell lines. Adenoviruses containing fibers from serotype 16 and 50 showed increased gene transfer and were further analyzed. In a gene-directed prodrug activation system using cytosine deaminase, these adenoviruses proved to be effective in eradicating primary pancreatic tumor cells. Fiber-chimeric Ad5 containing fiber 16 and wild-type Ad5 were also transduced ex vivo to slices of normal human pancreatic tissue and pancreatic carcinoma tissue obtained during surgery. It was shown that fiber-chimeric Ad5 with fiber 16 revealed an improved gene delivery to primary pancreatic tumor tissue compared to Ad5. In conclusion, fiber-chimeric adenoviruses carrying fiber 16 and 50 reveal a significantly enhanced gene transfer and an increased specificity to human pancreatic adenocarcinoma compared to Ad5, whereas transduction to normal pancreatic tissue was decreased. These findings expand the therapeutic window of Ad gene therapy for pancreatic cancer.


Subject(s)
Adenoviridae/genetics , Adenoviridae/physiology , Genetic Therapy/methods , Genetic Vectors/genetics , Genetic Vectors/physiology , Pancreatic Neoplasms/therapy , Cell Line , Cell Line, Tumor , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Polymerase Chain Reaction , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombination, Genetic/genetics , Transduction, Genetic , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
4.
Gene Ther ; 12(17): 1347-52, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15877046

ABSTRACT

Replicating adenoviral vectors (RAds) hold great promise for the treatment of cancer. Significant therapeutic effects of these vectors do not only rely on tumor targeting but also on efficient release of viral progeny from host cells. Cytotoxic genes expressed late in the adenoviral life cycle can significantly enhance viral release and spreading. Therefore, an adenoviral cloning system that allows easy integration of established tumor targeting techniques together with late expression of transgenes can be a valuable tool for the development of RAds. We expanded the features of the widely used AdEasy adenoviral cloning system toward the production of tropism modified replicating adenoviral vectors that express transgenes late in the viral life cycle. Three vectors (pIRES, pFIBER and pAdEasy-Sce) that facilitate easy manipulation of the adenoviral fiber region were established. Unique BstBI and I-Sce-1 restriction sites facilitate the introduction of retargeting peptides in the fiber HI-loop and of genes of interest in the fiber transcription unit. We validated the system by constructing an E1-positive adenovirus with an RGD motif in the fiber HI-loop and green fluorescent protein (GFP) expressed from the fiber transcription unit (AdDelta24Fiber-rgd-GFP). Additionally, assessment of E1-negative replication-deficient vectors confirmed strict dependence upon E1 expression for the expression of transgenes inserted into the fiber transcription unit. This flexible cloning system allows for straightforward construction of tropism expanded replicating adenoviral vectors that express transgenes late in the adenoviral life cycle.


Subject(s)
Adenoviridae/physiology , Genetic Therapy/methods , Genetic Vectors/genetics , Neoplasms/therapy , Adenoviridae/genetics , Adenoviridae Infections , Adenovirus E1 Proteins/genetics , Adenovirus E3 Proteins/genetics , Capsid Proteins/genetics , Cloning, Molecular , Gene Expression , Genetic Engineering , Green Fluorescent Proteins/genetics , Humans , Oligopeptides/genetics , Transgenes , Tropism , Virus Replication
5.
Gene Ther ; 8(13): 969-76, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11438831

ABSTRACT

In this study we analyzed two ways of retargeting of Ad-vectors to human pancreatic carcinoma with the aim of enhancing the gene transfer efficiency. First, we analyzed the expression of the epidermal growth factor receptor (EGFR) on primary, as well as established pancreatic carcinoma cells by flow cytometry which revealed high expression levels of EGFR on the surface of these cells. We showed that EGFR-retargeted entry pathway using a bispecific fusion protein formed by a recombinant soluble form of truncated Coxsackie and Adenovirus Receptor (sCAR) genetically fused with human EGF (sCAR-EGF) redirects them to the EGFR leading to an enhanced gene transfer efficiency to pancreatic carcinoma cells. Since flow cytometry revealed absence of CAR expression, but the presence of at least one of both alphav integrins on the pancreatic carcinoma cells, a second way of targeting was investigated using a genetically modified Ad vector which has an RGD (Arg-Gly-Asp)-containing peptide inserted into the HI-loop of the fiber knob. This RGD targeted Ad (AdlucRGD) revealed efficient CAR-independent infection by allowing binding to cellular integrins resulting in a dramatic enhancement of gene transfer. These findings have direct relevance for Ad-vector based gene therapy strategies for pancreatic carcinoma.


Subject(s)
ErbB Receptors/metabolism , Gene Targeting/methods , Gene Transfer Techniques , Genetic Therapy/methods , Pancreatic Neoplasms/metabolism , Adenoviridae/genetics , Genetic Vectors , Humans , Integrins/metabolism , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/virology , Receptors, Vitronectin/metabolism , Tumor Cells, Cultured
6.
Gastroenterology ; 120(6): 1448-58, 2001 May.
Article in English | MEDLINE | ID: mdl-11313315

ABSTRACT

BACKGROUND & AIMS: We have specified the features of progressive familial intrahepatic cholestasis type 3 and investigated in 31 patients whether a defect of the multidrug resistance 3 gene (MDR3) underlies this phenotype. METHODS: MDR3 sequencing, liver MDR3 immunohistochemistry, and biliary phospholipid dosage were performed. RESULTS: Liver histology showed a pattern of biliary cirrhosis with patency of the biliary tree. Age at presentation ranged from the neonatal period to early adulthood. Sequence analysis revealed 16 different mutations in 17 patients. Mutations were identified on both alleles in 12 patients and only on 1 allele in 5. Four mutations lead to a frame shift, 2 are nonsense, and 10 are missense. An additional missense mutation probably representing a polymorphism was found in 5 patients. MDR3 mutations were associated with abnormal MDR3 canalicular staining and a low proportion of biliary phospholipids. Gallstones or episodes of cholestasis of pregnancy were found in patients or parents. Children with missense mutations had a less severe disease and more often a beneficial effect of ursodeoxycholic acid therapy. CONCLUSIONS: At least one third of the patients with a progressive familial intrahepatic cholestasis type 3 phenotype have a proven defect of MDR3. This gene defect should also be considered in adult liver diseases.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/genetics , ATP-Binding Cassette Transporters/genetics , Cholestasis, Intrahepatic/genetics , Liver Cirrhosis, Biliary/etiology , Mutation , ATP Binding Cassette Transporter, Subfamily B/analysis , ATP Binding Cassette Transporter, Subfamily B/deficiency , ATP-Binding Cassette Transporters/analysis , Adolescent , Adult , Bile/chemistry , Child , Child, Preschool , Cholestasis, Intrahepatic/drug therapy , Female , Genotype , Humans , Immunohistochemistry , Infant , Liver/chemistry , Liver/pathology , Male , Phenotype , Phospholipids/analysis , Polymorphism, Genetic , Ursodeoxycholic Acid/therapeutic use
7.
Hepatology ; 33(3): 627-32, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11230743

ABSTRACT

Although in Gilbert's syndrome (GS), bilirubin glucuronidation is impaired due to an extra TA in the TATA box of the promoter of the gene for bilirubin UDP-glucuronosyltransferase 1 (UGT1A1), many GS homozygotes lack unconjugated hyperbilirubinemia. Accordingly, an additional defect in bilirubin transport might be required for phenotypic expression. Plasma bilirubin and the early fractional hepatic uptake rate (BSP K(1)) of a low dose of tetrabromosulfophthalein (0.59 micromol/kg) were determined in (1) 15 unrelated patients with unconjugated hyperbilirubinemia plus 12 random controls; (2) 4 unrelated GS probands and 15 of their first-degree relatives; (3) 7 unrelated patients with hemolysis due to beta-Thalassemia minor. Subjects were classified by DNA sequencing of the promoter region of both UGT1A1 alleles. In group 1, GS homozygotes showed a highly significant negative linear correlation between plasma bilirubin levels and BSP K(1). BSP K(1) values overlapped considerably between GS and normal subjects, whereas, in group 2, they were clustered within, and sharply segregated among, families. Patients with hemolysis, despite elevated plasma bilirubin levels, had mean BSP K(1) values similar to the normal subjects. Within each GS subgroup with defined UGT1A1 mutations, the plasma bilirubin level is in part determined by the organic anion uptake rate, assessed by early plasma disappearance of low-dose BSP. The lower BSP uptake in GS is not secondary to the hyperbilirubinemia, but probably caused by (an) independent, genetically determined defect(s) in hepatic transport mechanism(s), shared by BSP and bilirubin, that are likely necessary for phenotypic expression of GS.


Subject(s)
Bilirubin/blood , Gilbert Disease/genetics , Gilbert Disease/metabolism , Glucuronosyltransferase/genetics , Indicators and Reagents/pharmacokinetics , Liver/metabolism , Mutation/physiology , Sulfobromophthalein/pharmacokinetics , Adult , Female , Gilbert Disease/blood , Hemolysis , Humans , Hyperbilirubinemia/blood , Hyperbilirubinemia/metabolism , Male , Middle Aged , Reference Values , beta-Thalassemia/blood , beta-Thalassemia/metabolism
8.
Cancer Gene Ther ; 8(12): 990-6, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11781662

ABSTRACT

Midkine (MK), a heparin binding growth factor, and cyclooxygenase-2 (COX-2), a key enzyme in the conversion of arachidonic acid to prostaglandin, are both up-regulated at the mRNA or protein level in many human malignant tumors. Here, we investigated the tumor specificity of both MK and COX-2 promoters in human pancreatic cancer, with the aim to improve the selectivity of therapeutic gene expression. We constructed recombinant adenoviral (Ad) vectors containing either the luciferase (Luc) reporter gene under the control of the COX-2 or MK promoter or the herpes simplex virus thymidine kinase (HSV Tk) gene under the control of the COX-2 promoter and compared the expression with the cytomegalovirus (CMV) promoter. AdMKLuc achieved moderate to relatively high activity upon infection to both primary and established pancreatic carcinoma cells. Of the two COX-2 promoter regions (COX-2M and COX-2L), both revealed a high activity in primary pancreatic carcinoma cells, whereas in the established pancreatic carcinoma cell lines, COX-2L has an approximately equal high activity compared to CMV. In addition, both AdCOX-2M Tk and AdCOX-2L Tk induced marked cell death in response to ganciclovir (GCV) in three of four established pancreatic carcinoma cell lines. From these results, and because it has been reported that AdMKTk and AdCOX-2L Tk in combination with GCV did not reveal significant liver toxicity, we conclude that the MK as well as the COX-2 promoters are promising tumor-specific promoters for Ad vector-based gene therapy of pancreatic cancer.


Subject(s)
Adenoviridae , Angiogenesis Inducing Agents/genetics , Carrier Proteins/genetics , Cytokines , Isoenzymes/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , Prostaglandin-Endoperoxide Synthases/genetics , Angiogenesis Inducing Agents/therapeutic use , Carrier Proteins/therapeutic use , Cyclooxygenase 2 , Genetic Therapy , Genetic Vectors , Humans , Isoenzymes/therapeutic use , Membrane Proteins , Midkine , Promoter Regions, Genetic/genetics , Prostaglandin-Endoperoxide Synthases/therapeutic use , Reassortant Viruses , Transfection , Tumor Cells, Cultured
9.
Gastroenterology ; 119(6): 1720-30, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11113093

ABSTRACT

BACKGROUND & AIMS: Patients with progressive familial intrahepatic cholestasis (PFIC) type 3 have a mutation in the MDR3 gene, encoding the hepatocanalicular phospholipid translocator. In general, liver failure develops within the first decade of life in these patients. Previous studies have shown that in the mdr2-knockout mouse, the animal model for this disease, the absence of phospholipids in bile causes chronic bile salt-induced damage to hepatocytes. We aimed to test the efficacy of hepatocyte transplantation and liver repopulation in this disease model. METHODS: Transgenic MDR3-expressing hepatocytes as well as normal mdr2(+/+) hepatocytes were transplanted in mdr2(-/-) mice, and liver repopulation was assessed by immunohistochemistry and measurement of biliary lipid secretion. RESULTS: Transplanted hepatocytes partially repopulated the liver, restored phospholipid secretion, and diminished liver pathology. Repopulation was stronger when hepatocellular damage was enhanced by a bile salt-supplemented diet. After 1 year, however, these animals developed multiple hepatic tumors, and biliary phospholipid secretion decreased. In transplanted animals receiving a control diet, repopulation was slower but eventually remained stable at 21%, while liver pathology was completely abrogated and tumor formation was prevented. CONCLUSIONS: These results suggest that moderate liver pathology is a safe condition for the induction of effective hepatocyte repopulation and that this therapy is potentially applicable to patients with PFIC type 3.


Subject(s)
Cholestasis/genetics , Cholestasis/surgery , Hepatocytes/transplantation , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP-Binding Cassette Transporters/genetics , Animals , Cholates/administration & dosage , Cholates/pharmacology , Cholestasis/metabolism , Cholestasis/pathology , Diet , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Neoplasms/etiology , Liver Neoplasms/prevention & control , Male , Mice , Mice, Knockout/genetics , Mice, Transgenic/genetics , Phospholipids/metabolism
10.
Hepatology ; 31(3): 684-93, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10706559

ABSTRACT

We have studied regulation of the multidrug resistance protein 2 (mrp2) during bile duct ligation (BDL) in the rat. In hepatocytes isolated after 16, 48, and 72 hours of BDL, mrp2-mediated dinitrophenyl-glutathione (DNP-GS) transport was decreased to 65%, 33%, and 33% of control values, respectively. The impaired mrp2-mediated transport coincided with strongly decreased mrp2 protein levels, without any significant changes in mrp2 RNA levels. Restoration of bile flow after a 48-hour BDL period resulted in a slow recovery of mrp2-mediated transport and protein levels. Immunohistochemical detection of the protein in livers of rats undergoing BDL showed strongly reduced mrp2 staining after 48 hours, which was initiated in the periportal areas of the liver lobule and progressed toward the pericentral areas after 96 hours. Immunofluorescent detection of mrp2 in livers of rats undergoing 48 hours of BDL revealed decreased staining accompanied by intracellular localization of the protein in pericanalicular vesicular structures. Within this intracellular compartment, mrp2 colocalized with the bile salt transporter (bsep) and was still active as shown by vesicular accumulation of the fluorescent organic anion glutathione-bimane (GS-B). We conclude that down-regulation of mrp2 during BDL-induced obstructive cholestasis is mainly posttranscriptionally regulated. We propose that this down-regulation is caused by endocytosis of apical transporters followed up by increased breakdown of mrp2, probably in lysosomes. This breakdown of mrp2 is more severe in the periportal areas of the liver lobule.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP-Binding Cassette Transporters/metabolism , Bile Ducts, Intrahepatic/metabolism , Liver/metabolism , ATP Binding Cassette Transporter, Subfamily B/analysis , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP-Binding Cassette Transporters/analysis , ATP-Binding Cassette Transporters/genetics , Animals , Bile/metabolism , Bile Ducts, Intrahepatic/surgery , Bilirubin/blood , Biological Transport , Bridged Bicyclo Compounds , Cells, Cultured , Down-Regulation , Fluorescent Antibody Technique , Fluorescent Dyes , Gene Expression , Glutathione/analogs & derivatives , Glutathione/metabolism , RNA/metabolism , Rats , Time Factors
11.
Cell Transplant ; 7(6): 607-16, 1998.
Article in English | MEDLINE | ID: mdl-9853589

ABSTRACT

Ex vivo gene therapy, in which hepatocytes are harvested from mutants, retrovirally transduced with a normal gene and transplanted back into the donor, has been used for correction of inherited metabolic defects of liver. Major drawbacks of this method include limited availability of autologous hepatocytes, inefficient retroviral transduction of primary hepatocytes, and the limited number of hepatocytes that can be transplanted safely. To obviate these problems, we transduced primary hepatocytes derived from inbred bilirubin-UDP-glucuronosyl-transferase (BUGT)-deficient Gunn rats by infection with a recombinant retrovirus expressing temperature-sensitive mutant SV40 large T antigen (tsT). The immortalized cells were then transduced with a second recombinant retrovirus expressing human B-UGT, and a clone expressing high levels of the enzyme was expanded by culturing at permissive temperature (33 degrees C). At 37 degrees C, tsT antigen was degraded and the cells expressed UGT activity toward bilirubin at a level approximately twice that present in normal rat liver homogenates. For seeding the cells into the liver bed, 1 x 10(7) cells were injected into the spleens of syngeneic Gunn rats five times at 10-day intervals. Excretion of bilirubin glucuronides in bile was demonstrated by HPLC analysis and serum bilirubin levels were reduced by 27 to 52% in 40 days after the first transplantation and remained so throughout the duration of the study (120 days). None of the transplanted Gunn rats or SCID mice transplanted with the immortalized cells developed tumors.


Subject(s)
Antigens, Viral, Tumor/genetics , Cell Transplantation , Gene Transfer Techniques , Genetic Therapy , Glucuronosyltransferase/genetics , Liver/pathology , Liver/physiology , Animals , Cell Transformation, Viral , Cells, Cultured , Glucuronosyltransferase/biosynthesis , Glucuronosyltransferase/deficiency , Humans , Liver Transplantation , Mice , Rats , Rats, Gunn , Simian virus 40
12.
Am J Hum Genet ; 62(3): 585-92, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9497253

ABSTRACT

Crigler-Najjar syndrome type 1 (CN-1) is a recessively inherited, potentially lethal disorder characterized by severe unconjugated hyperbilirubinemia resulting from deficiency of the hepatic enzyme bilirubin-UDP-glucuronosyltransferase. In all CN-1 patients studied, structural mutations in one of the five exons of the gene (UGT1A1) encoding the uridinediphosphoglucuronate glucuronosyltransferase (UGT) isoform bilirubin-UGT1 were implicated in the absence or inactivation of the enzyme. We report two patients in whom CN-1 is caused, instead, by mutations in the noncoding intronic region of the UGT1A1 gene. One patient (A) was homozygous for a G-->C mutation at the splice-donor site in the intron, between exon 1 and exon 2. The other patient (B) was heterozygous for an A-->G shift at the splice-acceptor site in intron 3, and in the second allele a premature translation-termination codon in exon 1 was identified. Bilirubin-UGT1 mRNA is difficult to obtain, since it is expressed in the liver only. To determine the effects of these splice-junction mutations, we amplified genomic DNA of the relevant splice junctions. The amplicons were expressed in COS-7 cells, and the expressed mRNAs were analyzed. In both cases, splice-site mutations led to the use of cryptic splice sites, with consequent deletions in the processed mRNA. This is the first report of intronic mutations causing CN-1 and of the determination of the consequences of these mutations on mRNA structure, by ex vivo expression.


Subject(s)
Crigler-Najjar Syndrome/genetics , Glucuronosyltransferase/genetics , Mutation , RNA Splicing/genetics , Adult , Humans , Infant , Introns , Liver/enzymology , Male , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Nuclear/genetics
13.
Liver Transpl Surg ; 4(1): 78-88, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9457971

ABSTRACT

Conjugation with glucuronic acid, mediated by bilirubin-uridinediphosphoglucuronate glucuronosyltransferase (bilirubin-UGT), is essential for efficient biliary excretion of bilirubin. Inherited absence of this enzyme activity results in the potentially lethal Crigler-Najjar syndrome type I in humans and lifelong hyperbilirubinemia in Gunn rats. To develop a gene therapy for bilirubin-UGT deficiency, we constructed a high-titer replication-deficient amphotropic recombinant retrovirus (MFG-S hB-UGT1) capable of transferring the gene encoding bilirubin-UGT1, the principal bilirubin-UGT isoform in human liver. To stimulate hepatocyte proliferation, Gunn rats were subjected to 66% hepatectomy. After 24 hours, the portal vein, the hepatic artery, and the inferior vena cava above and below the hepatic vein were clamped, and the portal vein and the isolated segment of the vena cava were cannulated. The liver was perfused with the MFG-S hB-UGT1 preparation through the portal vein at 5 ml/min for 10 minutes, then circulation was restored. Control rat livers were perfused with a recombinant retrovirus expressing Escherichia coli beta-galactosidase. In MFG-S hB-UGT1-perfused rats, but not in controls, expression of human bilirubin-UGT1 was shown by immunotransblotting, bilirubin-UGT assay of liver homogenates, and biliary excretion of bilirubin diglucuronide and monoglucuronide. Mean serum bilirubin levels decreased by 20% to 25% in 3 weeks and remained at that level throughout the study period (18 months). This is the first report of long-term amelioration of inherited jaundice by retrovirus-directed gene therapy in an animal model for Crigler-Najjar syndrome.


Subject(s)
Bilirubin/blood , Glucuronosyltransferase/therapeutic use , Hyperbilirubinemia/therapy , Animals , Bile/chemistry , Bilirubin/metabolism , Cells, Cultured , Female , Fluorescent Antibody Technique, Indirect , Gene Transfer Techniques , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , Immunoblotting , Liver/enzymology , Male , Perfusion , RNA, Messenger/metabolism , Rats , Rats, Gunn , Rats, Wistar , Time Factors , Tissue Distribution , beta-Galactosidase/metabolism
14.
Proc Natl Acad Sci U S A ; 95(1): 282-7, 1998 Jan 06.
Article in English | MEDLINE | ID: mdl-9419367

ABSTRACT

Class III multidrug resistance (MDR) P-glycoproteins (P-gp), mdr2 in mice and MDR3 in man, mediate the translocation of phosphatidylcholine across the canalicular membrane of the hepatocyte. Mice with a disrupted mdr2 gene completely lack biliary phospholipid excretion and develop progressive liver disease, characterized histologically by portal inflammation, proliferation of the bile duct epithelium, and fibrosis. This disease phenotype is very similar to a subtype of progressive familial intrahepatic cholestasis, hallmarked by a high serum gamma-glutamyltransferase (gamma-GT) activity. We report immunohistochemistry for MDR3 P-gp, reverse transcription-coupled PCR sequence analysis, and genomic DNA analysis of MDR3 from two progressive familial intrahepatic cholestasis patients with high serum gamma-GT. Canalicular staining for MDR3 P-gp was negative in liver tissue of both patients. Reverse transcription-coupled PCR sequencing of the first patient's sequence demonstrated a homozygous 7-bp deletion, starting at codon 132, which results in a frameshift and introduces a stop codon 29 codons downstream. The second patient is homozygous for a nonsense mutation in codon 957 (C --> T) that introduces a stop codon (TGA). Our results demonstrate that mutations in the human MDR3 gene lead to progressive familial intrahepatic cholestasis with high serum gamma-GT. The histopathological picture in these patients is very similar to that in the corresponding mdr2(-/-) mouse, in which mdr2 P-gp deficiency induces complete absence of phospholipid in bile.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B , ATP-Binding Cassette Transporters/genetics , Cholestasis, Intrahepatic/genetics , Drug Resistance, Multiple/genetics , Mutation , Animals , Child , Cholestasis, Intrahepatic/enzymology , Cholestasis, Intrahepatic/pathology , Female , Humans , Liver/pathology , Male , Mice , gamma-Glutamyltransferase/blood
15.
J Hepatol ; 27(1): 1-5, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9252066

ABSTRACT

BACKGROUND/AIMS: Gilbert's syndrome is genetically characterized by an extra TA element in the TATAA-box of the promotor region upstream of the bilirubin UDP-glucuronosyltransferase (UGT1A) coding region (Bosma et al. N Engl J Med 1995; 333: 1171-5). Persistent unconjugated hyperbilirubinemia is occasionally observed in liver transplant recipients with an otherwise normal liver function. We postulate that these patients could have received a liver from a donor with the Gilbert's syndrome genotype. Therefore, we investigated the UGT1A-gene TATAA-box in DNA from liver graft donors of jaundiced and non-jaundiced recipients. METHODS: DNA was obtained from stored donor lymphocytes and the number of TA elements in the TATAA-box of the UGT1A-gene promotor region was analyzed by polymerase chain-reaction. RESULTS: We observed two liver transplant recipients with persistent unconjugated hyperbilirubinemia. They received liver grafts from donors who were homozygous for an abnormal A(TA)7TAA-box in the UGT1A-gene. Four of 10 non-jaundiced recipients received livers from donors who were homozygous for the normal A(TA)6TAA-box and six received livers from donors who were heterozygous with a normal A(TA)6TAA-box on one allele and a prolonged A(TA)7TAA-box on the other allele. CONCLUSIONS: This study shows that liver graft recipients with persistent unconjugated hyperbilirubinemia may have received a liver from a donor with an abnormal TATAA-box in the bilirubin UGT1A-gene promotor region.


Subject(s)
Gilbert Disease/etiology , Glucuronosyltransferase/genetics , Liver Transplantation/adverse effects , Liver/enzymology , Promoter Regions, Genetic , Adult , Female , Gilbert Disease/enzymology , Glucuronosyltransferase/deficiency , Humans , Male , Middle Aged , TATA Box , Tissue Donors
16.
Hepatology ; 25(6): 1539-42, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9185779

ABSTRACT

The human Dubin-Johnson syndrome (DJS) is a rare autosomal recessive liver disorder characterized by chronic conjugated hyperbilirubinemia. Patients have impaired hepatobiliary transport of non-bile salt organic anions. A highly similar phenotype has been described for a mutant Wistar rat strain, the transport-deficient (TR-) rat, which is defective in the canalicular multispecific organic anion transporter (cmoat). This protein mediates adenosine triphosphate-dependent transport of a broad range of endogenous and xenobiotic compounds across the (apical) canalicular membrane of the hepatocyte. The complementary DNA (cDNA) encoding rat cmoat has recently been cloned, and the mutation underlying the defect in TR- rats has been identified. In the present study, we have isolated the human homologue of rat cmoat, human cMOAT, and analyzed the corresponding cDNA from fibroblasts of a DJS patient for mutations. Our results show that a mutation in this gene is the cause of DJS.


Subject(s)
Carrier Proteins/genetics , Genes , Jaundice, Chronic Idiopathic/genetics , Mutation , Amino Acid Sequence , Anion Transport Proteins , Antibodies, Monoclonal , Base Sequence , Carrier Proteins/metabolism , DNA, Complementary/genetics , Female , Humans , Immunohistochemistry/methods , Jaundice, Chronic Idiopathic/metabolism , Liver/metabolism , Middle Aged , Molecular Sequence Data , Reference Values , Tissue Distribution
17.
Hum Gene Ther ; 8(1): 27-36, 1997 Jan 01.
Article in English | MEDLINE | ID: mdl-8989992

ABSTRACT

The end product of the breakdown of the heme group of hemoglobin and other heme-containing proteins is bilirubin. Bilirubin is hydrophobic and cannot be excreted as such. Therefore, mammals have a liver enzyme bilirubin UDP-glucuronosyltransferase (B-UGT), which conjugates bilirubin with glucuronic acid, thereby making the molecule much more water soluble. Bilirubin glucuronides are secreted into bile. Patients with Crigler-Najjar (CN) disease have a deficiency in bilirubin UDP-glucuronosyltransferase and accumulate high serum levels of bilirubin. An animal model for CN disease is the Gunn rat. The obvious target for gene therapy for CN disease is the liver, but because liver cells do only divide infrequently, they are difficult to transduce. To investigate whether cells that are easily transduced can be used to develop gene therapy for CN disease, we have transduced Gunn rat fibroblasts with B-UGT, using a recombinant retrovirus. Gunn rat fibroblasts expressing B-UGT were able to glucuronidate bilirubin present in cell culture media. In this study, we describe the intraperitoneal transplantation of Gunn rats with Gunn rat fibroblasts expressing B-UGT. Transplantation of the fibroblasts corrected the genetic deficiency of the Gunn rats, serum bilirubin concentrations of the transplanted Gunn rats were reduced to normal, and bilirubin glucuronides appeared in bile. However, due to the prolonged period of cell culture, the transplanted fibroblasts were transformed, and the experimental animals developed tumors after transplantation.


Subject(s)
Genetic Therapy , Glucuronosyltransferase/metabolism , Transplantation, Autologous , Animals , Bile/chemistry , Bile/metabolism , Bilirubin/blood , Blotting, Western , Chromatography, High Pressure Liquid , Crigler-Najjar Syndrome/genetics , Crigler-Najjar Syndrome/therapy , Disease Models, Animal , Fibroblasts , Gene Expression/genetics , Genetic Vectors , Glucuronidase/metabolism , Immunohistochemistry , Neoplasms, Experimental/metabolism , Rats , Rats, Gunn , Rats, Wistar , Retroviridae/genetics , Transformation, Genetic/genetics
18.
Eur J Biochem ; 241(2): 393-402, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8917435

ABSTRACT

Experiments were designed to clarify the role of c-Jun/c-Fos and of putative phorbol 12-myristate-13-acetate-(PMA)-responsive elements (TREs) in the induction of plasminogen-activator inhibitor 1 (PAI-1) gene transcription in the human hepatoma cell line HepG2 by activators of protein kinase C (PKC). Treatment of HepG2 cells with the phorbol ester PMA or serum rapidly and transiently increased c-Jun and c-Fos mRNA and protein levels prior to PAI-1 induction. This induction of PAI-1 gene transcription was found to be dependent on ongoing protein synthesis. An essential role of c-Jun and c-Fos in basal and PMA-stimulated transcription of the PAI-1 gene is demonstrated by our finding that antisense c-jun and c-fos oligodeoxynucleotides both strongly reduced basal and PMA-stimulated PAI-1 synthesis. Since it has already been shown that two TREs between positions -58 and -50 and between -79 and -72 of the PAI-1 promoter are essential for basal and PMA-induced PAI-1 promoter activity ([16]), we examined binding of nuclear proteins to these elements. The protein-binding activity to the TRE between positions -79 and -72 shows very strong PMA induction of an unknown factor, which is not related to c-Jun or c-Fos. The TRE binding between positions -58 and -50 forms two complexes, both containing c-Jun protein. The faster migrating complex primarily contains c-Jun homodimers. The amount of the faster migrating complex is enhanced more than 30-fold in PMA-treated cells, due to a strongly increased binding of c-Jun homodimers and, to a minor extent, to binding of c-Jun/c-Fos heterodimers. Dissociation experiments suggest that the c-Jun/c-Fos heterodimers bind with much lower affinity compared to binding of c-Jun homodimers. Together with the finding that both antisense c-jun and antisense c-fos oligodeoxynucleotides reduced the amount of c-Jun homodimer, we conclude that binding of c-Jun homodimer to the TRE at positions -58 to -50 is important in the basal activity and PMA activation of the PAI-1 promoter in HepG2 cells.


Subject(s)
Gene Expression Regulation/drug effects , Genes, jun , Plasminogen Activator Inhibitor 1/genetics , Tetradecanoylphorbol Acetate/pharmacology , Base Sequence , Binding Sites/genetics , Cell Line , Culture Media , Cycloheximide/pharmacology , Genes, fos/drug effects , Genes, jun/drug effects , Humans , Kinetics , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-jun/biosynthesis , Proto-Oncogene Proteins c-jun/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factor AP-1/metabolism
19.
FEBS Lett ; 390(3): 294-8, 1996 Jul 29.
Article in English | MEDLINE | ID: mdl-8706880

ABSTRACT

Crigler-Najjar (CN) disease is caused by a deficiency of the hepatic enzyme, bilirubin UDP-glucuronosyltransferase (B-UGT). We have found two CN type II patients, who were homozygous for a leucine to arginine transition at position 15 of B-UGT1. This mutation is expected to disrupt the hydrophobic core of the signal peptide of B-UGT1. Wild type and mutant B-UGT cDNAs were transfected in COS cells. Mutant and wild type mRNA were formed in equal amounts. The mutant protein was expressed with 0.5% efficiency, as compared to wild type. Mutant and wild type mRNAs were translated in vitro. Wild type transferase is processed by microsomes, no processing of the mutant protein was observed.


Subject(s)
Crigler-Najjar Syndrome/genetics , Endoplasmic Reticulum/enzymology , Glucuronosyltransferase/genetics , Liver/enzymology , Protein Sorting Signals/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Crigler-Najjar Syndrome/enzymology , Gene Expression , Genes, Recessive , Glucuronosyltransferase/chemistry , Glucuronosyltransferase/metabolism , Homozygote , Humans , Hyperbilirubinemia/enzymology , Hyperbilirubinemia/genetics , Molecular Sequence Data , Point Mutation , Protein Biosynthesis , Protein Sorting Signals/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Analysis , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...