Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Crohns Colitis ; 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38366839

ABSTRACT

OBJECTIVE: Patients with mutations in ATP8B1 develop Progressive Familial Intrahepatic Cholestasis type 1 (PFIC1), a severe liver disease that requires life-saving liver transplantation. PFIC1 patients also present with gastrointestinal problems, including intestinal inflammation and diarrhea, which are aggravated after liver transplantation. Here we investigate the intestinal function of ATP8B1 in relation to inflammatory bowel diseases. DESIGN: ATP8B1 expression was investigated in intestinal samples of patients with Crohn's Disease (CD) or Ulcerative Colitis (UC) as well as in murine models of intestinal inflammation. Colitis was induced in ATP8B1-deficient mice with Dextran Sodium Sulphate (DSS) and intestinal permeability was investigated. Epithelial barrier function was assessed in ATP8B1 knock-down Caco2-BBE cells. Co-immunoprecipitation experiments were performed in Caco2-BBE cells overexpressing ATP8B1-eGFP. Expression and localization of ATP8B1 and tight junction proteins were investigated in cells and in biopsies of UC and PFIC1 patients. RESULTS: ATP8B1 expression was decreased in UC and DSS-treated mice, and associated with a decreased Tight Junctional pathway transcriptional program. ATP8B1-deficient mice were extremely sensisitve to DSS-induced colitis, evidenced by increased intestinal barrier leakage. ATP8B1 knockdown cells showed delayed barrier establishment that associated with affected Claudin-4 (CLDN4) levels and localization.. CLDN4 immunohistochemistry showed a tight-junctional staining in control tissue, whereas in UC and intestinal PFIC1 samples, CLDN4 was not properly localized. CONCLUSION: ATP8B1 is important in the establishment of the intestinal barrier Downregulation of ATP8B1 levels in UC, and subsequent altered localization of tight junctional proteins, including CLDN4, might therefore be an important mechanism in UC pathophysiology.

2.
Int J Mol Sci ; 23(20)2022 Oct 15.
Article in English | MEDLINE | ID: mdl-36293199

ABSTRACT

ATP8B1 is a phospholipid flippase that is deficient in patients with progressive familial intrahepatic cholestasis type 1 (PFIC1). PFIC1 patients suffer from severe liver disease but also present with dyslipidemia, including low plasma cholesterol, of yet unknown etiology. Here we show that ATP8B1 knockdown in HepG2 cells leads to a strong increase in the mitochondrial oxidative phosphorylation (OXPHOS) without a change in glycolysis. The enhanced OXPHOS coincides with elevated low-density lipoprotein receptor protein and increased mitochondrial fragmentation and phosphatidylethanolamine levels. Furthermore, expression of phosphatidylethanolamine N-methyltransferase, an enzyme that catalyzes the conversion of mitochondrial-derived phosphatidylethanolamine to phosphatidylcholine, was reduced in ATP8B1 knockdown cells. We conclude that ATP8B1 deficiency results in elevated mitochondrial PE levels that stimulate mitochondrial OXPHOS. The increased OXPHOS leads to elevated LDLR levels, which provides a possible explanation for the reduced plasma cholesterol levels in PFIC1 disease.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Phosphatidylethanolamine N-Methyltransferase/metabolism , Adenosine Triphosphatases/metabolism , Phosphatidylethanolamines , Carcinoma, Hepatocellular/genetics , Oxidative Phosphorylation , Phospholipids/metabolism , Liver Neoplasms/genetics , Cholesterol , Phosphatidylcholines , Lipoproteins, LDL/metabolism
3.
Cell Biochem Funct ; 40(8): 914-925, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36169099

ABSTRACT

ATP8B1 is a phospholipid flippase and member of the type 4 subfamily of P-type ATPases (P4-ATPase) subfamily. P4-ATPases catalyze the translocation of phospholipids across biological membranes, ensuring proper membrane asymmetry, which is crucial for membrane protein targeting and activity, vesicle biogenesis, and barrier function. Here we have investigated the role of ATP8B1 in the endolysosomal pathway in macrophages. Depletion of ATP8B1 led to delayed degradation of content in the phagocytic pathway and in overacidification of the endolysosomal system. Furthermore, ATP8B1 knockdown cells exhibited large multivesicular bodies filled with intraluminal vesicles. Similar phenotypes were observed in CRISPR-generated ATP8B1 knockout cells. Importantly, induction of autophagy led to accumulation of autophagosomes in ATP8B1 knockdown cells. Collectively, our results support a novel role for ATP8B1 in lysosomal fusion in macrophages, a process crucial in the terminal phase of endolysosomal degradation.


Subject(s)
Adenosine Triphosphatases , Phospholipids , Phospholipids/metabolism , Cell Membrane/metabolism , Adenosine Triphosphatases/metabolism , Membrane Proteins/metabolism , Lysosomes
4.
PLoS One ; 14(4): e0213069, 2019.
Article in English | MEDLINE | ID: mdl-30947313

ABSTRACT

Extracellular vesicles (EVs) released by cells have a role in intercellular communication to regulate a wide range of biological processes. Two types of EVs can be recognized. Exosomes, which are released from multi-vesicular bodies upon fusion with the plasma membrane, and ectosomes, which directly bud from the plasma membrane. How cells regulate the quantity of EV release is largely unknown. One of the initiating events in vesicle biogenesis is the regulated transport of phospholipids from the exoplasmic to the cytosolic leaflet of biological membranes. This process is catalyzed by P4-ATPases. The role of these phospholipid transporters in intracellular vesicle transport has been established in lower eukaryotes and is slowly emerging in mammalian cells. In Caenorhabditis elegans (C. elegans), deficiency of the P4-ATPase member TAT-5 resulted in enhanced EV shedding, indicating a role in the regulation of EV release. In this study, we investigated whether the mammalian ortholog of TAT-5, ATP9A, has a similar function in mammalian cells. We show that knockdown of ATP9A expression in human hepatoma cells resulted in a significant increase in EV release that was independent of caspase-3 activation. Pharmacological blocking of exosome release in ATP9A knockdown cells did significantly reduce the total number of EVs. Our data support a role for ATP9A in the regulation of exosome release from human cells.


Subject(s)
Adenosine Triphosphatases/genetics , Exosomes/genetics , Extracellular Vesicles/genetics , Membrane Transport Proteins/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caspase 3/genetics , Cell Communication/genetics , Cell Membrane/genetics , Cell-Derived Microparticles/genetics , Endocytosis/genetics , Extracellular Vesicles/metabolism , Gene Expression Regulation , Hep G2 Cells , Humans , Phospholipids/metabolism , Protein Transport/genetics
6.
Cell Mol Life Sci ; 74(4): 715-730, 2017 02.
Article in English | MEDLINE | ID: mdl-27628304

ABSTRACT

P4-ATPases are lipid flippases that catalyze the transport of phospholipids to create membrane phospholipid asymmetry and to initiate the biogenesis of transport vesicles. Here we show, for the first time, that lipid flippases are essential to dampen the inflammatory response and to mediate the endotoxin-induced endocytic retrieval of Toll-like receptor 4 (TLR4) in human macrophages. Depletion of CDC50A, the ß-subunit that is crucial for the activity of multiple P4-ATPases, resulted in endotoxin-induced hypersecretion of proinflammatory cytokines, enhanced MAP kinase signaling and constitutive NF-κB activation. In addition, CDC50A-depleted THP-1 macrophages displayed reduced tolerance to endotoxin. Moreover, endotoxin-induced internalization of TLR4 was strongly reduced and coincided with impaired endosomal MyD88-independent signaling. The phenotype of CDC50A-depleted cells was also induced by separate knockdown of two P4-ATPases, namely ATP8B1 and ATP11A. We conclude that lipid flippases are novel elements of the innate immune response that are essential to attenuate the inflammatory response, possibly by mediating endotoxin-induced internalization of TLR4.


Subject(s)
ATP-Binding Cassette Transporters/immunology , Adenosine Triphosphatases/immunology , Endocytosis , Lipopolysaccharides/immunology , Macrophages/immunology , Toll-Like Receptor 4/immunology , ATP-Binding Cassette Transporters/genetics , Adenosine Triphosphatases/genetics , Cell Line , Gene Knockdown Techniques , Humans , Immunity, Innate , Macrophages/cytology , Macrophages/metabolism , Membrane Proteins/genetics , Membrane Proteins/immunology , Myeloid Differentiation Factor 88/immunology , NF-kappa B/immunology , Signal Transduction
7.
Biochim Biophys Acta ; 1863(9): 2280-8, 2016 09.
Article in English | MEDLINE | ID: mdl-27301931

ABSTRACT

Progressive familial intrahepatic cholestasis type 1 (PFIC1) is caused by mutations in the gene encoding the phospholipid flippase ATP8B1. Apart from severe cholestatic liver disease, many PFIC1 patients develop extrahepatic symptoms characteristic of cystic fibrosis (CF), such as pulmonary infection, sweat gland dysfunction and failure to thrive. CF is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR), a chloride channel essential for epithelial fluid transport. Previously it was shown that CFTR transcript levels were strongly reduced in livers of PFIC1 patients. Here we have investigated the hypothesis that ATP8B1 is important for proper CFTR expression and function. We analyzed CFTR expression in ATP8B1-depleted intestinal and pulmonary epithelial cell lines and assessed CFTR function by measuring short-circuit currents across transwell-grown ATP8B1-depleted intestinal T84 cells and by a genetically-encoded fluorescent chloride sensor. In addition, we studied CFTR surface expression upon induction of CFTR transcription. We show that CFTR protein levels are strongly reduced in the apical membrane of human ATP8B1-depleted intestinal and pulmonary epithelial cell lines, a phenotype that coincided with reduced CFTR activity. Apical membrane insertion upon induction of ectopically-expressed CFTR was strongly impaired in ATP8B1-depleted cells. We conclude that ATP8B1 is essential for correct apical localization of CFTR in human intestinal and pulmonary epithelial cells, and that impaired CFTR localization underlies some of the extrahepatic phenotypes observed in ATP8B1 deficiency.


Subject(s)
Adenosine Triphosphatases/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Phospholipid Transfer Proteins/metabolism , Phospholipids/metabolism , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Chlorides/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epithelial Cells/metabolism , Humans , Intestinal Mucosa/metabolism , Ion Channel Gating , Lung/cytology , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
Biochim Biophys Acta ; 1842(12 Pt A): 2378-86, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25239307

ABSTRACT

Deficiency of the phospholipid flippase ATPase, aminophospholipid transporter, class I, type 8B, member 1 (ATP8B1) causes progressive familial intrahepatic cholestasis type 1 (PFIC1) and benign recurrent intrahepatic cholestasis type 1 (BRIC1). Apart from cholestasis, many patients also suffer from diarrhea of yet unknown etiology. Here we have studied the hypothesis that intestinal ATP8B1 deficiency results in bile salt malabsorption as a possible cause of PFIC1/BRIC1 diarrhea. Bile salt transport was studied in ATP8B1-depleted intestinal Caco-2 cells. Apical membrane localization was studied by a biotinylation approach. Fecal bile salt and electrolyte contents were analyzed in stool samples of PFIC1 patients, of whom some had undergone biliary diversion or liver transplantation. Bile salt uptake by the apical sodium-dependent bile salt transporter solute carrier family 10 (sodium/bile acid cotransporter), member 2 (SLC10A2) was strongly impaired in ATP8B1-depleted Caco-2 cells. The reduced SLC10A2 activity coincided with strongly reduced apical membrane localization, which was caused by impaired apical membrane insertion of SLC10A2. Moreover, we show that endogenous ATP8B1 exists in a functional heterodimer with transmembrane protein 30A (CDC50A) in Caco-2 cells. Analyses of stool samples of post-transplant PFIC1 patients demonstrated that bile salt content was not changed, whereas sodium and chloride concentrations were elevated and potassium levels were decreased. The ATP8B1-CDC50A heterodimer is essential for the apical localization of SLC10A2 in Caco-2 cells. Diarrhea in PFIC1/BRIC1 patients has a secretory origin to which SLC10A2 deficiency may contribute. This results in elevated luminal bile salt concentrations and consequent enhanced electrolyte secretion and/or reduced electrolyte resorption.


Subject(s)
Adenosine Triphosphatases/metabolism , Membrane Proteins/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Symporters/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Bile Acids and Salts/metabolism , Biological Transport/genetics , Blotting, Western , Caco-2 Cells , Cell Membrane/metabolism , Cholestasis, Intrahepatic/genetics , Cholestasis, Intrahepatic/metabolism , Feces/chemistry , Humans , Intestinal Mucosa/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mutation , Protein Multimerization , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Taurocholic Acid/metabolism , Taurocholic Acid/pharmacokinetics
9.
J Histochem Cytochem ; 60(3): 205-18, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22253360

ABSTRACT

CDC50 proteins are ß-subunits for P4 ATPases, which upon heterodimerization form a functional phospholipid translocation complex. Emerging evidence in mouse models and men links mutations in P4 ATPase genes with human disease. This study analyzed the tissue distribution and cellular localization of CDC50A, the most abundant and ubiquitously expressed CDC50 homologue in the mouse. The authors have raised antibodies that detect mouse and human CDC50A and studied CDC50A localization and glycosylation status in mouse liver cells. CDC50A is a terminal-glycosylated glycoprotein and is expressed in hepatocytes and liver sinusoidal endothelial cells, where it resides in detergent-resistant membranes. In pancreas and stomach, CDC50A localized to secretory vesicles, whereas in the kidney, CDC50A localized to the apical region of proximal convoluted tubules of the cortex. In WIF-B9 cells, CDC50A partially costains with the trans-Golgi network. Data suggest that CDC50A is present as a fully glycosylated protein in vivo, which presumes interaction with distinct P4 ATPases.


Subject(s)
Adenosine Triphosphatases/metabolism , Liver/metabolism , Membrane Proteins/analysis , Animals , Blotting, Western , Cell Line , Cell Membrane/metabolism , Cricetinae , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gastric Mucosa/metabolism , Glycosylation , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Immunohistochemistry , Isoenzymes/analysis , Isoenzymes/biosynthesis , Kidney/cytology , Kidney/metabolism , Liver/cytology , Membrane Proteins/biosynthesis , Mice , Organ Specificity , Pancreas/cytology , Pancreas/metabolism , Rats , Stomach/cytology
10.
Gastroenterology ; 141(5): 1927-37.e1-4, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21820390

ABSTRACT

BACKGROUND & AIMS: Progressive familial intrahepatic cholestasis can be caused by mutations in ABCB4 or ATP8B1; each encodes a protein that translocates phospholipids, but in opposite directions. ABCB4 flops phosphatidylcholine from the inner to the outer leaflet, where it is extracted by bile salts. ATP8B1, in complex with the accessory protein CDC50A, flips phosphatidylserine in the reverse direction. Abcb4(-/-) mice lack biliary secretion of phosphatidylcholine, whereas Atp8b1-deficient mice have increased excretion of phosphatidylserine into bile. Each system is thought to have a role protecting the canalicular membrane from bile salts. METHODS: To investigate the relationship between the mechanisms of ABCB4 and ATP8B1, we expressed the transporters separately and together in cultured cells and studied viability and phospholipid transport. We also created mice with disruptions in ABCB4 and ATP8B1 (double knockouts) and studied bile formation and hepatic damage in mice fed bile salts. RESULTS: Overexpression of ABCB4 was toxic to HEK293T cells; the toxicity was counteracted by coexpression of the ATP8B1-CDC50A complex. In Atp8b1-deficient mice, bile salts induced extraction of phosphatidylserine and ectoenzymes from the canalicular membrane; this process was not observed in the double-knockout mice. CONCLUSIONS: ATP8B1 is required for hepatocyte function, particularly in the presence of ABCB4. This is most likely because the phosphatidylserine flippase complex of ATP8B1-CDC50A counteracts the destabilization of the membrane that occurs when ABCB4 flops phosphatidylcholine. Lipid asymmetry is therefore important for the integrity of the canalicular membrane; ABCB4 and ATP8B1 cooperate to protect hepatocytes from bile salts.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/physiology , Adenosine Triphosphatases/physiology , Bile Canaliculi/cytology , Cell Membrane/physiology , ATP Binding Cassette Transporter, Subfamily B/deficiency , ATP Binding Cassette Transporter, Subfamily B/genetics , Adenosine Triphosphatases/deficiency , Adenosine Triphosphatases/genetics , Animals , Bile Acids and Salts/pharmacology , Bile Canaliculi/physiology , Cells, Cultured , HEK293 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Male , Mice , Mice, Knockout , Models, Animal , Phosphatidylcholines/metabolism , Phospholipid Transfer Proteins , ATP-Binding Cassette Sub-Family B Member 4
11.
Hepatology ; 50(5): 1597-605, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19731236

ABSTRACT

UNLABELLED: Mutations in ATP8B1 cause progressive familial intrahepatic cholestasis type 1 (PFIC1) and benign recurrent intrahepatic cholestasis type 1 (BRIC1), forming a spectrum of cholestatic disease. Whereas PFIC1 is a progressive, endstage liver disease, BRIC1 patients suffer from episodic periods of cholestasis that resolve spontaneously. At present it is not clear how the type and location of the mutations relate to the clinical manifestations of PFIC1 and BRIC1. ATP8B1 localizes to the canalicular membrane of hepatocytes where it mediates the inward translocation of phosphatidylserine. ATP8B1 interacts with CDC50A, which is required for endoplasmic reticulum exit and plasma membrane localization. In this study we analyzed a panel of missense mutations causing PFIC1 (G308V, D554N, G1040R) or BRIC1 (D70N, I661T). In addition, we included two mutations that have been associated with intrahepatic cholestasis of pregnancy (ICP) (D70N, R867C). We examined the effect of these mutations on protein stability and interaction with CDC50A in Chinese hamster ovary cells, and studied the subcellular localization in WIF-B9 cells. Protein stability was reduced for three out of six mutations studied. Two out of three PFIC1 mutant proteins did not interact with CDC50A, whereas BRIC1/ICP mutants displayed reduced interaction. Importantly, none of the PFIC1 mutants were detectable in the canalicular membrane of WIF-B9 cells, whereas all BRIC1/ICP mutants displayed the same cellular staining pattern as wild-type ATP8B1. Our data indicate that PFIC1 mutations lead to the complete absence of canalicular expression, whereas in BRIC1/ICP residual protein is expressed in the canalicular membrane. CONCLUSION: These data provide an explanation for the difference in severity between the phenotypes of PFIC1 and BRIC1.


Subject(s)
Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Hepatocytes/metabolism , Mutation, Missense/genetics , Animals , Cell Line , Cholestasis, Intrahepatic/metabolism , Cholestasis, Intrahepatic/pathology , Cricetinae , Cricetulus , Disease Models, Animal , Female , Hepatocytes/pathology , Humans , Membrane Proteins/metabolism , Phospholipid Transfer Proteins , Rats
12.
J Biol Chem ; 284(15): 9947-54, 2009 Apr 10.
Article in English | MEDLINE | ID: mdl-19228692

ABSTRACT

Mutations in ATP8B1 cause severe inherited liver disease. The disease is characterized by impaired biliary bile salt excretion (cholestasis), but the mechanism whereby impaired ATP8B1 function results in cholestasis is poorly understood. ATP8B1 is a type 4 P-type ATPase and is a flippase for phosphatidylserine. Atp8b1-deficient mice display a dramatic increase in the biliary extraction of cholesterol from the canalicular (apical) membrane of the hepatocyte. Here we studied the hypothesis that disproportionate cholesterol extraction from the canalicular membrane impairs the activity of the bile salt transporter, ABCB11, and as a consequence causes cholestasis. Using single pass liver perfusions, we show that not only ABCB11-mediated transport but also Abcc2-mediated transport were reduced at least 4-fold in Atp8b1 deficiency. We show that canalicular membranes of cholestatic Atp8b1-deficient mice have a dramatically reduced cholesterol to phospholipid ratio, i.e. 0.75 +/- 0.24 versus 2.03 +/- 0.71 for wild type. In vitro depletion of cholesterol from mouse liver plasma membranes using methyl-beta-cyclodextrin demonstrated a near linear relation between cholesterol content of the membranes and ATP-dependent taurocholate transport. Abcc2-mediated transport activity was not affected up to 30% of membrane cholesterol depletion but declined to negligible levels at 70% of membrane cholesterol depletion. These effects were reversible as cholesterol repletion of the liver membranes completely restored Abcb11- and Abcc2-mediated transport. Our data demonstrate that membrane cholesterol content is a critical determinant of ABCB11/ABCC2 transport activity, provide an explanation for the etiology of ATP8B1 disease, and suggest a novel mechanism protecting the canalicular membrane against luminal bile salt overload.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/physiology , Bile Canaliculi/metabolism , Cell Membrane/metabolism , Cholesterol/metabolism , Liver/metabolism , Mutation , ATP Binding Cassette Transporter, Subfamily B, Member 11 , Animals , Biological Transport , Mice , Mice, Inbred C57BL , Models, Biological , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/physiology , Perfusion , Phospholipid Transfer Proteins , Recombinant Proteins/metabolism
13.
Gastroenterology ; 134(7): 2091-100, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18466903

ABSTRACT

BACKGROUNDS & AIMS: ATP8B1 is a phosphatidylserine flippase in the canalicular membrane; patients with mutations in ATP8B1 develop severe chronic (PFIC1) or periodic (BRIC1) cholestatic liver disease. We have observed that Atp8b1 deficiency leads to enhanced biliary cholesterol excretion. It has been established that biliary cholesterol excretion depends on transport by the heterodimer Abcg5/Abcg8. We hypothesized that the increased cholesterol output was due to enhanced extraction from the altered canalicular membrane rather than to higher Abcg5/Abcg8 activity. We therefore studied the relation between Abcg5/Abcg8 expression and biliary cholesterol excretion in mice lacking Atp8b1, Abcg8, or both (GF mice). METHODS: Bile formation was studied in LXR agonist-fed wild-type mice as well as mice lacking Atp8b1 or Abcg8, or in GF mice upon infusion of taurocholate. Bile samples were analyzed for cholesterol, bile salt, phospholipids, and ectoenzyme content. RESULTS: LXR agonist increased Abcg5/8 expression, and this was accompanied by increased biliary cholesterol output in both wild-type and Atp8b1(G308V/G308V) mice. However, Atp8b1(G308V/G308V) mice maintained higher cholesterol output. Although in Abcg8(-/-) mice biliary cholesterol output was severely reduced, GF mice displayed high biliary cholesterol output, which was comparable with wild-type mice. Bile of both Atp8b1(G308V/G308V) and GF mice displayed elevated levels of phosphatidylserine and sphingomyelin, indicating membrane stress. CONCLUSIONS: Our data demonstrate that the increased biliary cholesterol excretion in Atp8b1-deficient mice is independent of Abcg5/8 activity. This implicates that Atp8b1 deficiency leads to a decrease in the detergent resistance and subsequent nonspecific extraction of cholesterol from the canalicular membrane by bile salts.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphatases/metabolism , Bile/metabolism , Cholesterol/metabolism , Lipoproteins/metabolism , Liver/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , ATP-Binding Cassette Transporters/genetics , Adenosine Triphosphatases/deficiency , Adenosine Triphosphatases/genetics , Animals , Bile/enzymology , Bile Acids and Salts/blood , Bile Canaliculi/metabolism , Cell Membrane/metabolism , DNA-Binding Proteins/agonists , DNA-Binding Proteins/metabolism , Hydrocarbons, Fluorinated , Lipoproteins/deficiency , Lipoproteins/genetics , Liver/drug effects , Liver/enzymology , Liver X Receptors , Male , Membrane Fluidity , Mice , Mice, Inbred C57BL , Mice, Knockout , Orphan Nuclear Receptors , Phospholipid Transfer Proteins , Phospholipids/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Sulfonamides/pharmacology , Time Factors , Up-Regulation
14.
Hepatology ; 47(1): 268-78, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17948906

ABSTRACT

UNLABELLED: Mutations in ATP8B1 cause progressive familial intrahepatic cholestasis type 1 and benign recurrent intrahepatic cholestasis type 1. Previously, we have shown in mice that Atp8b1 deficiency leads to enhanced biliary excretion of phosphatidylserine, and we hypothesized that ATP8B1 is a flippase for phosphatidylserine. However, direct evidence for this function is still lacking. In Saccharomyces cerevisiae, members of the Cdc50p/Lem3p family are essential for proper function of the ATP8B1 homologs. We have studied the role of two human members of this family, CDC50A and CDC50B, in the routing and activity of ATP8B1. When only ATP8B1 was expressed in Chinese hamster ovary cells, the protein localized to the endoplasmic reticulum. Coexpression with CDC50 proteins resulted in relocalization of ATP8B1 from the endoplasmic reticulum to the plasma membrane. Only when ATP8B1 was coexpressed with CDC50 proteins was a 250%-500% increase in the translocation of fluorescently labeled phosphatidylserine observed. Importantly, natural phosphatidylserine exposure in the outer leaflet of the plasma membrane was reduced by 17%-25% in cells coexpressing ATP8B1 and CDC50 proteins in comparison with cells expressing ATP8B1 alone. The coexpression of ATP8B1 and CDC50A in WIF-B9 cells resulted in colocalization of both proteins in the canalicular membrane. CONCLUSION: Our data indicate that CDC50 proteins are pivotal factors in the trafficking of ATP8B1 to the plasma membrane and thus may be essential determinants of ATP8B1-related disease. In the plasma membrane, ATP8B1 functions as a flippase for phosphatidylserine. Finally, CDC50A may be the potential beta-subunit or chaperone for ATP8B1 in hepatocytes.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , 4-Chloro-7-nitrobenzofurazan/analogs & derivatives , 4-Chloro-7-nitrobenzofurazan/metabolism , Amino Acid Sequence , Animals , Antigens/metabolism , CHO Cells , Cricetinae , Cricetulus , DNA, Complementary/isolation & purification , Gene Expression , Green Fluorescent Proteins/metabolism , HeLa Cells , Hemagglutinins/immunology , Humans , Liver/metabolism , Lysosomes/metabolism , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Phosphatidylserines/metabolism , RNA, Messenger/metabolism
15.
Hepatology ; 44(1): 195-204, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16799980

ABSTRACT

Progressive familial intrahepatic cholestasis type 1 (PFIC1, Byler disease, OMIM 211600) is a severe inherited liver disease caused by mutations in ATP8B1. ATP8B1 is a member of the type 4 subfamily of P-type ATPases, which are phospholipid flippases. PFIC1 patients generally develop end-stage liver disease before the second decade of life. The disease is characterized by impaired biliary bile salt excretion, but the mechanism whereby impaired ATP8B1 function results in cholestasis is unclear. In a mouse model for PFIC1, we observed decreased resistance of the hepatocanalicular membrane to hydrophobic bile salts as evidenced by enhanced biliary recovery of phosphatidylserine, cholesterol, and ectoenzymes. In liver specimens from PFIC1 patients, but not in those from control subjects, ectoenzyme expression at the canalicular membrane was markedly deficient. In isolated mouse livers Atp8b1 deficiency impaired the transport of hydrophobic bile salts into bile. In conclusion, our study shows that Atp8b1 deficiency causes loss of canalicular phospholipid membrane asymmetry that in turn renders the canalicular membrane less resistant toward hydrophobic bile salts. The loss of phospholipid asymmetry may subsequently impair bile salt transport and cause cholestasis.


Subject(s)
Adenosine Triphosphatases/deficiency , Bile Acids and Salts/metabolism , Bile Canaliculi/metabolism , Cell Membrane/metabolism , Cholestasis, Intrahepatic/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Bile Canaliculi/ultrastructure , Biological Transport , Blotting, Western , Cell Membrane/ultrastructure , Cholestasis, Intrahepatic/pathology , Chromatography, Thin Layer , Disease Models, Animal , Hydrophobic and Hydrophilic Interactions , Immunohistochemistry , In Vitro Techniques , Male , Mice , Microscopy, Electron , Phospholipid Transfer Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...