Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Pediatr Transplant ; 16(4): 379-84, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22489822

ABSTRACT

Atopic disease occurs in solid organ transplant recipients with an increasingly recognized frequency. The time course for the development of these atopic diseases in liver transplantation has not been described. The objective was to characterize the atopic manifestations of children receiving chronic immunosuppression after orthotopic liver transplantation (OLT). Chart review and follow-up questionnaire were utilized for 176 OLT pediatric recipients at a single institution for manifestations of allergic disease. Atopic disease was present in 25 (14.2%) patients. Median age at transplant was 16 months with a median follow-up of 63 months. Food allergy and non-food related atopic symptoms presented at a median of 11.5 (IQR, 6-28) and 19 (IQR, 5-41) months post-transplantation, respectively. The median age at transplant of the non-atopic children was 72 months, higher than patients with atopy (p < 0.001). Food allergy and atopic skin disease symptoms were present in 40% and 56% of cases, respectively. Asthma, allergic rhinitis, or both were found in 66% of cases. The onset of symptoms of food allergy and eczema (median, 12 months post-transplantation) preceded symptoms of allergic rhinitis and asthma. (median of 27 and 30 months post-transplantation, respectively). Atopy occurs in ∼14% of pediatric liver transplant recipients, with manifestations including food allergy, eczema, allergic rhinitis, and asthma.


Subject(s)
Hypersensitivity, Immediate/etiology , Liver Transplantation , Postoperative Complications , Age Factors , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Hypersensitivity, Immediate/epidemiology , Infant , Male , Postoperative Complications/epidemiology , Retrospective Studies , Surveys and Questionnaires
2.
Clin Pharmacol Ther ; 87(4): 473-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20200515

ABSTRACT

Members of the nuclear receptor (NR) superfamily of ligand-activated transcription factors are players of substantial relevance in the regulation of hepatic gene expression. NRs direct normal physiology and metabolism, adaptations to liver disease, and responses to inflammation and toxins.They also contribute to the regenerative response. In this review, we summarize currently available experimental and clinical data, focusing on the role of NRs in cholestasis and nonalcoholic fatty liver disease (NAFLD). We also highlight the potential of NRs as targets for safe and effective therapeutic interventions.


Subject(s)
Cholestasis/physiopathology , Fatty Liver/physiopathology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cholestasis/drug therapy , Drug Delivery Systems , Fatty Liver/drug therapy , Humans , Inflammation/drug therapy , Inflammation/physiopathology , Receptors, Cytoplasmic and Nuclear/drug effects
3.
Xenobiotica ; 38(7-8): 1043-71, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18668439

ABSTRACT

In recent years the discovery of a number of major transporter proteins expressed in the liver and intestine specifically involved in bile acid transport has led to improved understanding of bile acid homeostasis and the enterohepatic circulation. Sodium (Na(+))-dependent bile acid uptake from portal blood into the liver is mediated primarily by the Na(+) taurocholate co-transporting polypeptide (NTCP), while secretion across the canalicular membrane into the bile is carried out by the bile salt export pump (BSEP). In the ileum, absorption of bile acids from the lumen into epithelial cells is mediated by the apical Na(+) bile salt transporter (ASBT), whereas exit into portal blood across the basolateral membrane is mediated by the organic solute transporter alpha/beta (OSTalpha/beta) heterodimer. Regulation of transporter gene expression and function occurs at several different levels: in the nucleus, members of the nuclear receptor superfamily, regulated by bile acids and other ligands are primarily involved in controlling gene expression, while cell signalling events directly affect transporter function, and subcellular localization. Polymorphisms, dysfunction, and impaired adaptive responses of several of the bile acid transporters, e.g. BSEP and ASBT, results in liver and intestinal disease. Bile acid transporters are now understood to play central roles in driving bile flow, as well as adaptation to various pathological conditions, with complex regulation of activity and function in the nucleus, cytoplasm, and membrane.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Gene Expression Regulation , Intestinal Diseases/metabolism , Liver Diseases/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Symporters/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/genetics , Bile Acids and Salts/metabolism , Biological Transport/genetics , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cytoplasm/genetics , Cytoplasm/metabolism , Gene Expression Regulation/genetics , Humans , Intestinal Diseases/genetics , Liver Diseases/genetics , Organic Anion Transporters, Sodium-Dependent/genetics , Polymorphism, Genetic , Signal Transduction , Symporters/genetics
4.
Transpl Infect Dis ; 8(2): 108-12, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16734634

ABSTRACT

BACKGROUND: Polymorphisms of the solute carrier family 11 member 1 (Slc11a1) gene have previously been associated with susceptibility to infectious disease, anti-tumor defenses, and autoimmune diseases. We postulated that polymorphisms of the gene may also be associated with susceptibility to post-transplant lymphoproliferative disease (PTLD), a disease thought to be related to an impaired immune response to Epstein-Barr virus (EBV) in immunosuppressed patients. METHODS: Whole blood samples were obtained from 45 pediatric patients who underwent liver transplantation. Polymerase chain reaction (PCR) was used to amplify a 3' region of the gene that includes an exon 15 single-nucleotide substitution (referred to as D543N) and a 4-bp deletion polymorphism (referred to as 3'-UTR). PCR products were digested using AvaII and FokI restriction enzymes for the D543N and 3'-UTR polymorphisms, respectively. PTLD disease status and EBV virus serum titers of all patients were obtained from hospital records. RESULTS: Six of the 45 pediatric transplant recipients developed PTLD. An association was found between 3'-UTR polymorphisms of Slc11a1 and incidence of PTLD after liver transplantation (P = 0.005). In addition, post-transplant serum EBV titers were higher (P = 0.009) for recipients with certain Slc11a1 polymorphisms. No association was found between the D543N polymorphism and incidence of PTLD. CONCLUSION: 3'-UTR polymorphisms of the Slc11a1 gene appear to be associated with susceptibility to PTLD and the immune response to EBV in pediatric liver transplant recipients. Genotyping of pediatric patients undergoing liver transplantation may enable early identification of patients at high risk for developing high EBV titers and/or PTLD.


Subject(s)
Cation Transport Proteins/genetics , Liver Transplantation/physiology , Lymphoproliferative Disorders/genetics , 3' Untranslated Regions/genetics , Adolescent , Cation Transport Proteins/immunology , Child , Child, Preschool , Epstein-Barr Virus Infections/blood , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/immunology , Female , Genetic Predisposition to Disease , Humans , Immunity, Cellular/immunology , Infant , Liver Transplantation/adverse effects , Liver Transplantation/immunology , Lymphoproliferative Disorders/etiology , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/virology , Male , Polymorphism, Genetic
5.
Eur J Pediatr Surg ; 15(6): 434-6, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16418964

ABSTRACT

We report here on a newborn infant who initially presented with a history of gastroschisis, abdominal distension, and jaundice. Further studies revealed that the child had findings consistent with extrahepatic biliary atresia (EHBA). The child later developed hepatic failure and subsequently expired. The purpose of this case report is to discuss the pathogenesis of each disease process and to identify any commonality between the pathogenesis of gastroschisis and EHBA.


Subject(s)
Biliary Atresia/diagnosis , Gastroschisis/diagnosis , Biliary Atresia/etiology , Biliary Atresia/pathology , Fatal Outcome , Female , Gastroschisis/epidemiology , Gastroschisis/etiology , Humans , Infant, Newborn , Risk Factors
6.
Biochem Biophys Res Commun ; 309(3): 612-8, 2003 Sep 26.
Article in English | MEDLINE | ID: mdl-12963034

ABSTRACT

BACKGROUND/AIMS: Molecular mechanisms underlying transcriptional rat multidrug-resistance protein 2 (Mrp2, Abcc2) gene regulation are mostly unclear. Given the presence of putative binding sites for the Y-box binding protein YB-1 in the regulatory sequence, its trans-regulatory influence was analyzed. METHODS: Reporter assays in HepG2 cells with various Mrp2 deletion constructs in the absence and presence of co-transfected YB-1 were performed. DNA binding studies with recombinant YB-1 protein and nuclear extracts obtained from HepG2 cells and rat liver tissue were carried out. RESULTS: The minimal promoter sequence was confined to the proximal 186 bp. A YB-1 responsive element, Mrp2 YRE-1, was mapped at -186/-157, which exhibits specific YB-1 binding. YB-1 acts as a potent repressor of Mrp2 promoter activity in vitro. CONCLUSIONS: Constitutive Mrp2 gene expression is conferred through the proximal -186 bp. YB-1 acts as a repressor in vitro by specific binding to a defined element in the proximal promoter sequence.


Subject(s)
ATP-Binding Cassette Transporters , CCAAT-Enhancer-Binding Proteins/metabolism , Carrier Proteins/genetics , DNA-Binding Proteins , Down-Regulation , Repressor Proteins/metabolism , Transcription Factors , Animals , Base Sequence , Binding Sites , Carrier Proteins/metabolism , Male , Molecular Sequence Data , NFI Transcription Factors , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley , Regulatory Sequences, Nucleic Acid , Response Elements , Sequence Homology, Nucleic Acid , Transcription, Genetic , Tumor Cells, Cultured , Y-Box-Binding Protein 1
9.
Gastroenterology ; 121(1): 140-7, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11438503

ABSTRACT

BACKGROUND AND AIMS: Hepatic bile acid homeostasis is regulated by negative feedback inhibition of genes involved in the uptake and synthesis of bile acids. Bile acids down-regulate the rate-limiting gene for bile acid synthesis, cholesterol 7alpha-hydroxylase (cyp7a), via bile acid receptor (fxr) activation of an inhibitory nuclear receptor, shp. We hypothesized that shp would also mediate negative feedback regulation of ntcp, the principal hepatic bile acid transporter. METHODS: Primary rat hepatocytes or transfected HepG2 and Cos cells were treated with retinoids with or without bile acids, and effects on bile acid transport and ntcp and shp gene expression and promoter activity were determined. Gel shift assays were performed using synthetic fxr, rxr, and rar proteins. RESULTS: Bile acid treatment of primary rat hepatocytes prevented retinoid activation of ntcp gene expression and function; this corresponded temporally with shp gene activation. Bile acid-mediated down-regulation occurred via fxr-dependent suppression of the ntcp RXR:RAR response element. Moreover, cotransfected shp directly inhibited retinoid activation of the ntcp promoter. CONCLUSIONS: These studies show negative feedback regulation of ntcp by bile acid-activated fxr via induction of shp. This novel regulatory pathway provides a means for coordinated down-regulation of bile acid import and synthesis, thereby protecting the hepatocyte from bile acid-mediated damage in cholestatic conditions.


Subject(s)
Bacterial Proteins/genetics , Bile Acids and Salts/genetics , Carrier Proteins/genetics , Cytochrome c Group/genetics , Liver Neoplasms/genetics , Membrane Transport Proteins , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Bacterial Proteins/pharmacology , Bile Acids and Salts/biosynthesis , Carcinoma, Hepatocellular/genetics , Cytochrome c Group/pharmacology , Humans , Male , Organic Anion Transporters, Sodium-Dependent , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/drug effects , Symporters , Tumor Cells, Cultured
10.
J Clin Invest ; 107(11): 1451-8, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11390427

ABSTRACT

Children with chronic inflammatory diseases experience growth failure and wasting. This may be due to growth hormone resistance caused by cytokine-induced suppression of growth hormone receptor (GHR) gene expression. However, the factors governing inflammatory regulation of GHR are not known. We have reported that Sp1 and Sp3 regulate hepatic GHR expression. We hypothesized that TNF-alpha suppresses GHR expression by inhibiting Sp1/Sp3 transactivators. LPS administration significantly reduced murine hepatic GHR expression, as well as Sp1 and Sp3 binding to GHR promoter cis elements. TNF-alpha was integral to this response, as LPS did not affect hepatic Sp1/Sp3 binding or GHR expression in TNF receptor 1-deficient mice. TNF-alpha treatment of BNL CL.2 mouse liver cells reduced Sp1 and Sp3 binding to a GHR promoter cis element and downregulated activity of a GHR promoter-driven luciferase reporter. Combined mutations within adjacent Sp elements eliminated GHR promoter suppression by TNF-alpha without affecting overall nuclear levels of Sp1 or Sp3 proteins. These studies demonstrate that murine GHR transcription is downregulated by LPS, primarily via TNF-alpha-dependent signaling. Evidence suggests that inhibition of Sp transactivator binding is involved. Further investigation of these mechanisms may identify novel strategies for preventing inflammatory suppression of growth.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , Liver/metabolism , Podophyllin/analogs & derivatives , Receptors, Somatotropin/metabolism , Sp1 Transcription Factor/metabolism , Trans-Activators/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Child , DNA-Binding Proteins/genetics , Genes, Reporter/genetics , Hepatocytes/drug effects , Humans , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Nuclear Proteins/metabolism , Plasmids/genetics , Plasmids/metabolism , Podophyllotoxin/analogs & derivatives , Promoter Regions, Genetic/genetics , Receptors, Somatotropin/genetics , Sp1 Transcription Factor/genetics , Trans-Activators/genetics
12.
Am J Physiol Gastrointest Liver Physiol ; 279(2): G347-55, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10915644

ABSTRACT

The divergent homeobox gene Hex is expressed in both developing and mature liver. A putative Hex binding site was identified in the promoter region of the liver-specific Na(+)-bile acid cotransporter gene (ntcp), and we hypothesized that Hex regulates the ntcp promoter through this site. Successive 5'-deletions of the ntcp promoter in a luciferase reporter construct transfected into Hep G2 cells confirmed a Hex response element (HRE) within the ntcp promoter (nt -733/-714). Moreover, p-CMHex transactivated a heterologous promoter construct containing HRE multimers (p4xHRELUC), whereas a 5-bp mutation of the core HRE eliminated transactivation. A dominant negative form of Hex (p-Hex-DN) suppressed basal luciferase activity of p-4xHRELUC and inhibited activation of this construct by p-CMHex. Interestingly, p-CMHex transactivated the HRE in Hep G2 cells but not in fibroblast-derived COS cells, suggesting the possibility that Hex protein requires an additional liver cell-specific factor(s) for full activity. Electrophoretic mobility shift assays confirmed that liver and Hep G2 cells contain a specific nuclear protein that binds the native HRE. We have demonstrated that the liver-specific ntcp gene promoter is the first known target of Hex and is a useful tool for evaluating function of the Hex protein.


Subject(s)
Carrier Proteins/genetics , Homeodomain Proteins/genetics , Liver/physiology , Membrane Transport Proteins , Promoter Regions, Genetic/physiology , Animals , Base Sequence , COS Cells , Carrier Proteins/metabolism , Gene Expression Regulation/physiology , Genetic Complementation Test , Hepatoblastoma , Homeodomain Proteins/metabolism , Humans , Liver/cytology , Liver Neoplasms , Molecular Sequence Data , Mutagenesis/physiology , Oligonucleotide Probes , Organic Anion Transporters, Sodium-Dependent , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Symporters , Transcription Factors , Transcription, Genetic/physiology , Tumor Cells, Cultured
13.
Gene ; 246(1-2): 311-20, 2000 Apr 04.
Article in English | MEDLINE | ID: mdl-10767553

ABSTRACT

The orphan homeobox gene, Hex, has a limited domain of expression which includes the developing and adult mouse liver. Hex is expressed in the developing liver coincident with the forkhead/winged helix transcription factor, Hepatocyte Nuclear Factor 3beta (HNF3beta). Although preliminary characterization of the mouse Hex promoter has recently been reported, the identity of the molecular regulators that drive liver expression is not known. We hypothesized that putative HNF3beta and GATA-4 elements within the Hex promoter would confer liver-enriched expression. A series of Hex promoter-driven luciferase reporter constructs were transfected in liver-derived HepG2 and fibroblast-like Cos cells+/-HNF3beta or GATA expression plasmids. The Hex promoter region from nt -235/+22 conferred basal activity in both HepG2 and Cos cells, with the region from -103/+22 conferring liver-enriched activity. HNF3beta and GATA-4 transactivated the promoter via response elements located within nt -103/+22, whereas Sp1 activated the -235/+22 construct. Mutation of the HNF3 element significantly reduced promoter activity in HepG2 cells, whereas this element in isolation conferred HNF3beta responsiveness to a heterologous promoter. Electrophoretic mobility shift assays were performed to confirm transcription factor:DNA binding. We conclude that HNF3beta and GATA-4 contribute to liver-enriched expression of Hex.


Subject(s)
DNA-Binding Proteins/physiology , Homeodomain Proteins/genetics , Liver/metabolism , Nuclear Proteins/physiology , Transcription Factors/physiology , Animals , COS Cells , Cell Line , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , GATA4 Transcription Factor , Genes, Homeobox/genetics , Hepatocyte Nuclear Factor 3-beta , Humans , Liver/cytology , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Plasmids , Promoter Regions, Genetic/genetics , Protein Binding , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Analysis, DNA , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Sp1 Transcription Factor/physiology , Sp3 Transcription Factor , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation , Transfection , Tumor Cells, Cultured
14.
J Biol Chem ; 275(12): 8835-43, 2000 Mar 24.
Article in English | MEDLINE | ID: mdl-10722729

ABSTRACT

Cytokines have been implicated in the pathogenesis of inflammatory cholestasis. This is due to transcriptional down-regulation of hepatic transporters including the Na(+)/bile acid cotransporter, ntcp, and the multispecific organic anion exporter, mrp2. We have recently shown that ntcp suppression by lipopolysaccharide in vivo is caused by down-regulation of transactivators including the previously uncharacterized Footprint B-binding protein. Both the ntcp FpB element and the mrp2 promoter contain potential retinoid-response elements. We hypothesized that retinoic acid receptor (RAR) and retinoid X receptor (RXR) heterodimers would activate these two genes and that cytokines that reduce bile flow might do so by suppressing nuclear levels of these transactivators. Retinoid transactivation and interleukin-1beta down-regulation of the ntcp and mrp2 promoters were mapped to RXRalpha:RARalpha-response elements. Gel mobility shift assays demonstrated specific binding of RXRalpha:RARalpha heterodimers to the ntcp and mrp2 retinoid-response elements. The RXRalpha:RARalpha complex was down-regulated by IL-1beta in HepG2 cells. An unexpected finding was that an adjacent CAAT-enhancer-binding protein element was required for maximal transactivation of the ntcp promoter by RXRalpha:RARalpha. Taken together, these studies demonstrate regulation of two hepatobiliary transporter genes by RXRalpha:RARalpha and describe a mechanism which likely contributes to their down-regulation during inflammation.


Subject(s)
Bile/metabolism , Carrier Proteins/genetics , Interleukin-1/pharmacology , Liver/metabolism , Membrane Transport Proteins , Retinoids/pharmacology , Transcriptional Activation , Albumins/genetics , Animals , Anion Transport Proteins , Carrier Proteins/biosynthesis , Cholestasis/etiology , Cytokines/pharmacology , DNA-Binding Proteins/metabolism , Dimerization , Humans , Interleukin-6/genetics , Mutation , Nuclear Proteins/metabolism , Organic Anion Transporters, Sodium-Dependent , Promoter Regions, Genetic , Protein Binding , Protein Isoforms/metabolism , Rats , Receptors, Retinoic Acid/metabolism , Recombinant Fusion Proteins/biosynthesis , Response Elements , Retinoid X Receptors , Symporters , Transcription Factors/metabolism
15.
Hepatology ; 31(1): 124-30, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10613737

ABSTRACT

Endotoxemia leads to cytokine-mediated alterations of the hepatocellular sodium-taurocholate-cotransporting polypeptide (ntcp). We hypothesized that stimulated macrophages are essential transducers for down-regulating hepatocellular bile salt uptake in response to endotoxin (lipopolysaccharide [LPS]) exposure. Using an in vitro model, we exposed mouse macrophages (IC-21 cell line) to LPS for 24 hours. Concentrations of cytokines tumor necrosis factor-alpha (TNF-alpha), interleukin (IL)-1beta, and IL-6 increased 10.6-fold, 12.5-fold, and 444-fold, respectively, in LPS-conditioned IC-21 medium (CM) versus unconditioned IC-21 medium (UM). WIF-B rat hepatoma hybrid cells were incubated with either CM or UM or treated directly with medium containing recombinant TNF-alpha, IL-1beta, and IL-6. [(3)H]Taurocholate ([(3)H]TC) uptake decreased in WIF-B cells exposed to either TNF-alpha (54% of control), IL-1beta (78%), IL-6 (55%) as single additives, or in triple combination (TCC) (43%). A virtually identical decrease was observed after exposing WIF-B cells to CM (52%, P <.001). LPS had no direct effect on [(3)H]TC uptake. CM treatment did not decrease L-alanine transport in WIF-B cells. Blocking antibodies against TNF-alpha, IL-1beta, and IL-6 restored the diminished [(3)H]TC uptake in cells exposed to TCC and CM to 87% and 107% of controls, respectively. Northern blotting revealed that ntcp messenger RNA (mRNA) expression was significantly reduced in WIF-B cells after exposure to CM, and in primary rat hepatocytes exposed to CM or TNF-alpha (68%, 14%, and 29% of control, respectively). We conclude that macrophages and their ability to secrete the cytokines TNF-alpha, IL-1beta, and IL-6 may be essential in mediating the endotoxin-induced cholestatic effect of decreased hepatocellular bile salt uptake.


Subject(s)
Bile Acids and Salts/metabolism , Lipopolysaccharides/pharmacology , Liver Neoplasms, Experimental/metabolism , Macrophage Activation , Macrophages/physiology , Animals , Cell Line , Culture Media, Conditioned , Interleukin-1/biosynthesis , Interleukin-1/pharmacology , Interleukin-6/biosynthesis , Interleukin-6/pharmacology , Male , Mice , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Taurocholic Acid/metabolism , Tritium , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology
17.
Hepatology ; 30(1): 223-9, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10385660

ABSTRACT

The Na+-taurocholate cotransport polypeptide (ntcp) is the primary transporter for the uptake of bile acids in the liver. The second messenger adenosine 3':5'-cyclic monophosphate (cAMP) rapidly increases ntcp protein concentration in the plasma membrane, yet the mechanism is unknown. To investigate this, HepG2 cells were transiently transfected with a carboxy-terminal-tagged green fluorescence protein (GFP) conjugate of ntcp, and then examined by confocal video microscopy. Transporter activity was directly assayed with 3H-taurocholic acid (TC) scintigraphy. ntcp-GFP targeted to the plasma membrane in transfected cells, and the conjugate protein transported 3H-TC as effectively as unmodified rat ntcp. Stimulation of ntcp-GFP cells with cAMP increased GFP fluorescence in the plasma membrane by 40% (P <.0001) within 2.5 minutes and by 55% within 10 minutes. Similarly, cAMP increased transport of bile acids by 30%. Cytochalasin D, an inhibitor of microfilaments, did not prevent ntcp-GFP from targeting to the plasma membrane, but completely abolished the increase in GFP fluorescence seen in response to cAMP. In contrast, the microtubule inhibitor, nocodazole, prevented development of membrane fluorescence in 48 (96%) of 50 cells. Cells regained plasma membrane fluorescence within 2 hours after nocodazole removal. These findings suggest that targeting of ntcp to the plasma membrane consists of 2 steps: 1) delivery of ntcp to the region of the plasma membrane via microtubules; and 2) insertion of ntcp into the plasma membrane, in a microfilament- and cAMP-sensitive fashion.


Subject(s)
Actin Cytoskeleton/metabolism , Bile Acids and Salts/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Membrane/metabolism , Membrane Transport Proteins , Taurocholic Acid/metabolism , Actin Cytoskeleton/drug effects , Animals , Base Sequence , Biological Transport , Carcinoma, Hepatocellular , Carrier Proteins/chemistry , Cell Membrane/drug effects , Cyclic AMP/pharmacology , Cytochalasin D/pharmacology , Green Fluorescent Proteins , Humans , Kinetics , Liver Neoplasms , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Models, Molecular , Molecular Sequence Data , Open Reading Frames , Organic Anion Transporters, Sodium-Dependent , Protein Structure, Secondary , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Symporters , Transfection , Tritium , Tumor Cells, Cultured
19.
Curr Opin Gastroenterol ; 15(3): 184-91, 1999 May.
Article in English | MEDLINE | ID: mdl-17023944

ABSTRACT

This paper highlights several key issues, ideas, and findings that significantly contribute to our understanding of the organization, communication, and molecular machinery of the liver. The functional anatomy of the liver has been studied in several ways that have revealed the extent of the biliary tree within the hepatic parenchyma, including identification of the canals of Hering as their most distal ramification. The canals of Hering are also considered as the potential residence of hepatic progenitor cells. Hepatocytes can "communicate" with each other via gap junctions, but might also deliver hormones and nucleotides downstream to cholangiocytes. The interaction of inflammatory cells and inflammatory mediators with hepatocytes is of particular importance in transplant immunology, infection, inflammation, viral hepatitis, and fibrogenesis. The role of these mediators as well as certain "toxic" bile acids in apoptosis has become clearer with the discovery of the mitochondrial permeability transition. Moreover, ursodeoxycholic acid can reduce apoptosis by minimizing the mitochondrial permeability transition. Two new nuclear hormone receptors, PXR and SXR, have been identified. These are both activated by a variety of chemically distinct ligands, whose final common goal is the activation of cytochrome P450-containing drug-metabolizing enzymes. Thus, these two receptors are critical to the body's ability to metabolize a variety of compounds properly. Additional insight into the role of cytokines and cytokine receptors in liver regeneration is presented. Finally, in vivo gene therapy of liver-expressed genes by chimeric oligonucleotides appears quite promising as a means of correcting single nucleotide gene defects.

SELECTION OF CITATIONS
SEARCH DETAIL
...