Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Hepatol ; 73(5): 1013-1022, 2020 11.
Article in English | MEDLINE | ID: mdl-32540177

ABSTRACT

BACKGROUND & AIMS: The heterodimeric integrin receptor α4ß7 regulates CD4 T cell recruitment to inflamed tissues, but its role in the pathogenesis of non-alcoholic steatohepatitis (NASH) is unknown. Herein, we examined the role of α4ß7-mediated recruitment of CD4 T cells to the intestine and liver in NASH. METHODS: Male littermate F11r+/+ (control) and junctional adhesion molecule A knockout F11r-/- mice were fed a normal diet or a western diet (WD) for 8 weeks. Liver and intestinal tissues were analyzed by histology, quantitative reverse transcription PCR (qRT-PCR), 16s rRNA sequencing and flow cytometry. Colonic mucosa-associated microbiota were analyzed using 16s rRNA sequencing. Liver biopsies from patients with NASH were analyzed by confocal imaging and qRT-PCR. RESULTS: WD-fed knockout mice developed NASH and had increased hepatic and intestinal α4ß7+ CD4 T cells relative to control mice who developed mild hepatic steatosis. The increase in α4ß7+ CD4 T cells was associated with markedly higher expression of the α4ß7 ligand mucosal addressin cell adhesion molecule 1 (MAdCAM-1) in the colonic mucosa and livers of WD-fed knockout mice. Elevated MAdCAM-1 expression correlated with increased mucosa-associated Proteobacteria in the WD-fed knockout mice. Antibiotics reduced MAdCAM-1 expression indicating that the diet-altered microbiota promoted colonic and hepatic MAdCAM-1 expression. α4ß7 blockade in WD-fed knockout mice significantly decreased α4ß7+ CD4 T cell recruitment to the intestine and liver, attenuated hepatic inflammation and fibrosis, and improved metabolic indices. MAdCAM-1 blockade also reduced hepatic inflammation and fibrosis in WD-fed knockout mice. Hepatic MAdCAM-1 expression was elevated in patients with NASH and correlated with higher expression of α4 and ß7 integrins. CONCLUSIONS: These findings establish α4ß7/MAdCAM-1 as a critical axis regulating NASH development through colonic and hepatic CD4 T cell recruitment. LAY SUMMARY: Non-alcoholic steatohepatitis (NASH) is an advanced and progressive form of non-alcoholic fatty liver disease (NAFLD), and despite its growing incidence no therapies currently exist to halt NAFLD progression. Herein, we show that blocking integrin receptor α4ß7-mediated recruitment of CD4 T cells to the intestine and liver not only attenuates hepatic inflammation and fibrosis, but also improves metabolic derangements associated with NASH. These findings provide evidence for the potential therapeutic application of α4ß7 antibody in the treatment of human NASH.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Diet, Western/adverse effects , Integrins/metabolism , Intestinal Mucosa/immunology , Liver/immunology , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , CD4-Positive T-Lymphocytes/metabolism , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Gastrointestinal Microbiome/genetics , Humans , Integrins/antagonists & inhibitors , Integrins/immunology , Liver/pathology , Male , Mice , Mice, Knockout , Mucoproteins/antagonists & inhibitors , Mucoproteins/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology , RNA, Ribosomal, 16S/genetics , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics
2.
Biochim Biophys Acta Mol Basis Dis ; 1864(10): 3537-3545, 2018 10.
Article in English | MEDLINE | ID: mdl-30293572

ABSTRACT

Adiponectin inhibits hepatic stellate cell (HSC) activation and subsequent development of liver fibrosis via multiple mechanisms. Phosphatase and tensin homolog deletion 10 (PTEN) plays a crucial role in suppression of HSC activation, but its regulation by adiponectin is not fully understood. Here, we investigated the effect of adiponectin on PTEN in LX-2 cells, a human cell line and examined the underlying molecular mechanisms involved in adiponectin-mediated upregulation of PTEN activity during fibrosis. PTEN expression was found to be significantly reduced in the livers of mice treated with CCl4, whereas its expression was rescued by adiponectin treatment. The DNA methylation proteins DNMT1, DNMT3A, and DNMT3B are all highly expressed in activated primary HSCs compared to quiescent HSCs, and thus represent additional regulatory targets during liver fibrogenesis. Expression of DNMT proteins was significantly induced in the presence of fibrotic stimuli; however, only DNMT3B expression was reduced in the presence of adiponectin. Adiponectin-induced suppression of DNMT3B was found to be mediated by enhanced miR-29b expression. Furthermore, PTEN expression was significantly increased by overexpression of miR-29b, whereas its expression was markedly reduced by a miR-29b inhibitor in LX-2 cells. These findings suggest that adiponectin-induced upregulation of miR-29b can suppress DNMT3B transcription in LX-2 cells, thus resulting in reduced methylation of PTEN CpG islands and ultimately suppressing the PI3K/AKT pathway. Together, these data suggest a possible new explanation for the inhibitory effect of adiponectin on HSC activation and liver fibrogenesis.


Subject(s)
Adiponectin/metabolism , Carbon Tetrachloride/adverse effects , DNA (Cytosine-5-)-Methyltransferases/genetics , Hepatic Stellate Cells/cytology , Liver Cirrhosis/metabolism , MicroRNAs/genetics , PTEN Phosphohydrolase/genetics , Animals , Cell Line , Cell Proliferation , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA Methyltransferase 3A , Epigenesis, Genetic , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Humans , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Mice , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction , Up-Regulation , DNA Methyltransferase 3B
3.
J Biol Chem ; 293(33): 12781-12792, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29941453

ABSTRACT

Liver fibrosis arises from dysregulated wound healing due to persistent inflammatory hepatic injury. Periostin is a nonstructural extracellular matrix protein that promotes organ fibrosis in adults. Here, we sought to identify the molecular mechanisms in periostin-mediated hepatic fibrosis. Hepatic fibrosis in periostin-/- mice was attenuated as evidenced by significantly reduced collagen fibril density and liver stiffness compared with those in WT controls. A single dose of carbon tetrachloride caused similar acute liver injury in periostin-/- and WT littermates, and we did not detect significant differences in transaminases and major fibrosis-related hepatic gene expression between these two genotypes. Activated hepatic stellate cells (HSCs) are the major periostin-producing liver cell type. We found that in primary rat HSCs in vitro, periostin significantly increases the expression levels and activities of lysyl oxidase (LOX) and lysyl oxidase-like (LOXL) isoforms 1-3. Periostin also induced expression of intra- and extracellular collagen type 1 and fibronectin in HSCs. Interestingly, periostin stimulated phosphorylation of SMAD2/3, which was sustained despite short hairpin RNA-mediated knockdown of transforming growth factor ß (TGFß) receptor I and II, indicating that periostin-mediated SMAD2/3 phosphorylation is independent of TGFß receptors. Moreover, periostin induced the phosphorylation of focal adhesion kinase (FAK) and AKT in HSCs. Notably, siRNA-mediated FAK knockdown failed to block periostin-induced SMAD2/3 phosphorylation. These results suggest that periostin promotes enhanced matrix stiffness in chronic liver disease by activating LOX and LOXL, independently of TGFß receptors. Hence, targeting periostin may be of therapeutic benefit in combating hepatic fibrosis.


Subject(s)
Cell Adhesion Molecules/physiology , Chemical and Drug Induced Liver Injury/pathology , Hepatic Stellate Cells/pathology , Liver Cirrhosis/pathology , Protein-Lysine 6-Oxidase/metabolism , Animals , Carbon Tetrachloride/toxicity , Cells, Cultured , Chemical and Drug Induced Liver Injury/enzymology , Hepatic Stellate Cells/enzymology , Liver Cirrhosis/enzymology , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Rats , Rats, Sprague-Dawley , Signal Transduction
4.
Physiol Rep ; 5(23)2017 Dec.
Article in English | MEDLINE | ID: mdl-29208693

ABSTRACT

Alcohol consumption promotes loss of intestinal barrier function. However, mechanisms by which ethanol affects the tight junction (TJ), the cellular structure responsible for maintaining the gut epithelial barrier, are not well understood. Three classes of transmembrane proteins comprise TJs: occludin, claudins, and junctional adhesion molecules (JAMs). It has recently been postulated that JAM-A (F11R), the most abundant JAM expressed in intestinal epithelium, regulates "leak" pathway flux, a paracellular route for the nonselective permeation of large solutes. Since transluminal flux of many gut-derived antigens occurs through this pathway, we investigated the role of JAM-A in ethanol-induced disruption of the intestinal epithelial barrier. Using Caco-2 and SK-CO15 monolayers, we found that ethanol induced a dose- and time-dependent decrease in JAM-A protein expression to about 70% of baseline levels. Alcohol also reduced Ras-related protein 2 (Rap2) activity, and enhanced myosin light chain kinase (MLCK) activity, changes consistent with impaired JAM-A signaling. Stable overexpression and shRNA-mediated knockdown of JAM-A were employed to investigate the role of JAM-A in paracellular-mediated flux following alcohol exposure. The paracellular flux of 40-kDa fluorescein isothiocynate (FITC)-dextran following ethanol treatment was decreased by the overexpression of JAM-A; conversely, flux was enhanced by JAM-A knockdown. Thus, we conclude that ethanol-mediated control of JAM-A expression and function contributes to mechanisms by which this chemical induces intestinal epithelial leakiness.


Subject(s)
Cell Adhesion Molecules/metabolism , Ethanol/toxicity , Intestinal Absorption/drug effects , Intestinal Mucosa/metabolism , Receptors, Cell Surface/metabolism , Animals , Caco-2 Cells , Cell Adhesion Molecules/genetics , HEK293 Cells , Humans , Intestinal Mucosa/drug effects , Male , Mice , Mice, Inbred C57BL , Myosin Light Chains/metabolism , Receptors, Cell Surface/genetics , rap GTP-Binding Proteins/metabolism
5.
Gastroenterology ; 151(4): 733-746.e12, 2016 10.
Article in English | MEDLINE | ID: mdl-27342212

ABSTRACT

BACKGROUND & AIMS: There is evidence from clinical studies that compromised intestinal epithelial permeability contributes to the development of nonalcoholic steatohepatitis (NASH), but the exact mechanisms are not clear. Mice with disruption of the gene (F11r) encoding junctional adhesion molecule A (JAM-A) have defects in intestinal epithelial permeability. We used these mice to study how disruption of the intestinal epithelial barrier contributes to NASH. METHODS: Male C57BL/6 (control) or F11r(-/-) mice were fed a normal diet or a diet high in saturated fat, fructose, and cholesterol (HFCD) for 8 weeks. Liver and intestinal tissues were collected and analyzed by histology, quantitative reverse-transcription polymerase chain reaction, and flow cytometry. Intestinal epithelial permeability was assessed in mice by measuring permeability to fluorescently labeled dextran. The intestinal microbiota were analyzed using 16S ribosomal RNA sequencing. We also analyzed biopsy specimens from proximal colons of 30 patients with nonalcoholic fatty liver disease (NAFLD) and 19 subjects without NAFLD (controls) undergoing surveillance colonoscopy. RESULTS: F11r(-/-) mice fed a HFCD, but not a normal diet, developed histologic and pathologic features of severe NASH including steatosis, lobular inflammation, hepatocellular ballooning, and fibrosis, whereas control mice fed a HFCD developed only modest steatosis. Interestingly, there were no differences in body weight, ratio of liver weight:body weight, or glucose homeostasis between control and F11r(-/-) mice fed a HFCD. In these mice, liver injury was associated with significant increases in mucosal inflammation, tight junction disruption, and intestinal epithelial permeability to bacterial endotoxins, compared with control mice or F11r(-/-) mice fed a normal diet. The HFCD led to a significant increase in inflammatory microbial taxa in F11r(-/-) mice, compared with control mice. Administration of oral antibiotics or sequestration of bacterial endotoxins with sevelamer hydrochloride reduced mucosal inflammation and restored normal liver histology in F11r(-/-) mice fed a HFCD. Protein and transcript levels of JAM-A were significantly lower in the intestinal mucosa of patients with NAFLD than without NAFLD; decreased expression of JAM-A correlated with increased mucosal inflammation. CONCLUSIONS: Mice with defects in intestinal epithelial permeability develop more severe steatohepatitis after a HFCD than control mice, and colon tissues from patients with NAFLD have lower levels of JAM-A and higher levels of inflammation than subjects without NAFLD. These findings indicate that intestinal epithelial barrier function and microbial dysbiosis contribute to the development of NASH. Restoration of intestinal barrier integrity and manipulation of gut microbiota might be developed as therapeutic strategies for patients with NASH.


Subject(s)
Cell Adhesion Molecules/deficiency , Diet, High-Fat/adverse effects , Non-alcoholic Fatty Liver Disease/genetics , Receptors, Cell Surface/deficiency , Animals , Cholesterol , Diet, High-Fat/methods , Dietary Carbohydrates , Disease Models, Animal , Dysbiosis/complications , Dysbiosis/genetics , Fructose , Gastrointestinal Microbiome/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/microbiology , Non-alcoholic Fatty Liver Disease/pathology , Permeability , Reverse Transcriptase Polymerase Chain Reaction
6.
Lab Invest ; 96(8): 895-908, 2016 08.
Article in English | MEDLINE | ID: mdl-27239734

ABSTRACT

The CCAAT/enhancer-binding protein (C/EBP) homologous protein (CHOP), a major transcriptional regulator of endoplasmic reticulum (ER) stress-mediated apoptosis, is implicated in lipotoxicity-induced ER stress and hepatocyte apoptosis in non-alcoholic fatty liver disease (NAFLD). We have previously demonstrated that the glucagon-like peptide-1 (GLP-1) agonist, liraglutide, protects steatotic hepatocytes from lipotoxicity-induced apoptosis by improved handling of free fatty acid (FFA)-induced ER stress. In the present study, we investigated whether CHOP is critical for GLP-1-mediated restoration of ER homeostasis and mitigation of hepatocyte apoptosis in a murine model of NASH (non-alcoholic steatohepatitis). Our data show that despite similar caloric intake, CHOP KO (CHOP(-/-)) mice fed a diet high in fat, fructose, and cholesterol (HFCD) for 16 weeks developed more severe histological features of NASH compared with wild-type (WT) controls. Severity of NASH in HFCD-fed CHOP(-/-) mice correlated with significant decrease in peroxisomal ß-oxidation, and increased de novo lipogenesis and ER stress-mediated hepatocyte apoptosis. Four weeks of liraglutide treatment markedly attenuated steatohepatitis in HFCD-fed WT mice by improving insulin sensitivity, and suppressing de novo lipogenesis and ER stress-mediated hepatocyte apoptosis. However, in the absence of CHOP, liraglutide did not improve insulin sensitivity, nor suppress peroxisomal ß-oxidation or ER stress-mediated hepatocyte apoptosis. Taken together, these data indicate that CHOP protects hepatocytes from HFCD-induced ER stress, and has a significant role in the mechanism of liraglutide-mediated protection against NASH pathogenesis.


Subject(s)
Liraglutide/pharmacology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/prevention & control , Transcription Factor CHOP/metabolism , Animals , Apoptosis/drug effects , Blood Glucose/metabolism , Cells, Cultured , Cholesterol/metabolism , Diet, High-Fat/adverse effects , Dietary Carbohydrates/administration & dosage , Dietary Carbohydrates/adverse effects , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Exenatide , Hepatocytes/drug effects , Hepatocytes/metabolism , Insulin Resistance , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/pathology , Peptides/pharmacology , Protective Agents/pharmacology , Transcription Factor CHOP/deficiency , Transcription Factor CHOP/genetics , Venoms/pharmacology
7.
J Nutr Biochem ; 26(3): 285-92, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25577467

ABSTRACT

UNLABELLED: Nonalcoholic fatty liver disease (NAFLD) is the hepatic manifestation of the metabolic syndrome (MetS). Up to a third of NAFLD subjects are at risk for developing nonalcoholic steatohepatitis (NASH). Many rodent models fail to replicate both MetS and NASH. The purpose of this study was to develop a reliable mouse model of NASH and MetS using a diet containing cholesterol, saturated fat and carbohydrate that is reflective of Western diets of North Americans. EXPERIMENTAL DESIGN: We used adult male C57BL/6 J 4- to 5-week-old mice and administered a solid diet containing 0.2% cholesterol, 45% of its calories from fat, with 30% of the fat in the form of partially hydrogenated vegetable oil. We also provided carbohydrate largely as high-fructose corn syrup equivalent in water. In a separate cohort, we gave the identical diet in the absence of cholesterol. Glucose and insulin tolerance testing was conducted throughout the feeding period. The feeding was conducted for 16 weeks, and the mice were sacrificed for histological analysis, markers of MetS, liver inflammation, circulating lipids, as well as liver staining for fibrosis and alpha smooth muscle actin (α-SMA). RESULTS: We found that cholesterol significantly increased serum leptin, interleukin-6, liver weight and liver weight/body weight ratio, fibrosis and liver α-SMA. CONCLUSIONS: Mice administered a diet accurately reflecting patterns associated with humans afflicted with MetS can reliably replicate features of MetS, NASH and significant liver fibrosis. The model we describe significantly reduces the time by several months for development of stage 3 hepatic fibrosis.


Subject(s)
Cholesterol, Dietary/adverse effects , Diet, High-Fat/adverse effects , Diet, Western/adverse effects , Disease Models, Animal , Liver/physiopathology , Metabolic Syndrome/etiology , Non-alcoholic Fatty Liver Disease/etiology , Actins/metabolism , Adipokines/blood , Adipokines/metabolism , Animals , Biomarkers/blood , Biomarkers/metabolism , Glucose Intolerance/etiology , High Fructose Corn Syrup/adverse effects , Hydrogenation , Insulin Resistance , Liver/metabolism , Liver/pathology , Liver Cirrhosis/etiology , Male , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Metabolic Syndrome/physiopathology , Mice, Inbred C57BL , Organ Size , Time Factors
8.
PLoS One ; 9(10): e110405, 2014.
Article in English | MEDLINE | ID: mdl-25310107

ABSTRACT

Liver fibrosis is a growing global health problem characterized by excess deposition of fibrillar collagen, and activation of hepatic stellate cells (HSCs). Adiponectin is known to possess anti-fibrotic properties; however a high physiological concentration and multiple forms circulating in blood prohibit clinical use. Recently, an adiponectin-like small synthetic peptide agonist (ADP355: H-DAsn-Ile-Pro-Nva-Leu-Tyr-DSer-Phe-Ala-DSer-NH2) was synthesized for the treatment of murine breast cancer. The present study was designed to evaluate the efficacy of ADP355 as an anti-fibrotic agent in the in vivo carbon tetrachloride (CCl4)-induced liver fibrosis model. Liver fibrosis was induced in eight-week old male C57BL/6J mice by CCl4-gavage every other day for four weeks before injection of a nanoparticle-conjugated with ADP355 (nano-ADP355). Control gold nanoparticles and nano-ADP355 were administered by intraperitoneal injection for two weeks along with CCl4-gavage. All mice were sacrificed after 6 weeks, and serum and liver tissue were collected for biochemical, histopathologic and molecular analyses. Biochemical studies suggested ADP355 treatment attenuates liver fibrosis, determined by reduction of serum aspartate aminotransferase (AST), alanine aminotransferase ALT) and hydroxyproline. Histopathology revealed chronic CCl4-treatment results in significant fibrosis, while ADP355 treatment induced significantly reversed fibrosis. Key markers for fibrogenesis-α-smooth muscle actin (α-SMA), transforming growth factor-beta1 (TGF-ß1), connective tissue growth factor (CTGF), and the tissue inhibitor of metalloproteinase I (TIMP1) were also markedly attenuated. Conversely, liver lysates from ADP355 treated mice increased phosphorylation of both endothelial nitric oxide synthase (eNOS) and AMPK while AKT phosphorylation was diminished. These findings suggest ADP355 is a potent anti-fibrotic agent that can be an effective intervention against liver fibrosis.


Subject(s)
Adiponectin/agonists , Liver Cirrhosis, Experimental/pathology , Oligopeptides/pharmacology , AMP-Activated Protein Kinases/metabolism , Actins/metabolism , Adiponectin/metabolism , Animals , Carbon Tetrachloride/adverse effects , Collagen/metabolism , Disease Models, Animal , Gene Expression , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/drug therapy , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/metabolism , Male , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Mice , Nitric Oxide Synthase Type III/metabolism , Oligopeptides/administration & dosage , Phosphorylation , Proteolysis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
9.
FASEB J ; 28(12): 5172-83, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25154876

ABSTRACT

Previous evidence indicates that adiponectin possesses antifibrogenic activity in inhibiting liver fibrosis. Therapeutic strategies, however, are limited by adiponectin quaternary structure and effective concentrations in circulation. Here we postulate a novel molecular mechanism, whereby adiponectin targets focal adhesion kinase (FAK) activity and disrupts key features of the fibrogenic response. Adiponectin-null (Ad(-/-)) mice and wild-type littermates were exposed to either saline or carbon tetrachloride (CCl4) for 6 wk. CCl4-gavaged mice were also injected with attenuated adenoviral adiponectin (Ad-Adn) or Ad-LacZ for 2 wk. Hepatic stellate cells (HSCs) were treated with or without adiponectin to elucidate signal transduction mechanisms. In vivo delivery of Ad-Adn markedly attenuates CCl4-induced expression of key integrin proteins and markers of HSC activation: αv, ß3, ß1, α2(I) collagen, and α-smooth muscle actin. Confocal experiments of liver tissues demonstrated that adiponectin delivery also suppressed vinculin and p-FAK activity in activated HSCs. In vitro, adiponectin induced dephosphorylation of FAK, mediated by a physical association with activated tyrosine phosphatase, Shp2. Conversely, Shp2 knockdown by siRNA significantly attenuated adiponectin-induced FAK deactivation, and expression of TIMP1 and α2(I) collagen was abolished in the presence of adiponectin and si-FAK. Finally, we documented that either adiponectin or the synthetic peptide with adiponectin properties, ADP355, suppressed p-FAK in synthetic matrices with stiffness measurements of 9 and 15 kPa, assessed by immunofluorescent imaging and quantitation. The in vivo and in vitro data presented indicate that disassembly of focal adhesion complexes in HSCs is pivotal for hepatic fibrosis therapy, now that small adiponectin-like peptides are available.


Subject(s)
Adiponectin/physiology , Focal Adhesions , Hepatic Stellate Cells/cytology , Liver Cirrhosis/therapy , Animals , Base Sequence , DNA Primers , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
10.
J Membr Biol ; 245(5-6): 221-30, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22722762

ABSTRACT

Gap junction channels composed of connexins connect cells, allowing intercellular communication. Their cellular assembly involves a unique quality-control pathway. Some connexins [including connexin43 (Cx43) and Cx46] oligomerize in the trans-Golgi network following export of stabilized monomers from the endoplasmic reticulum (ER). In contrast, other connexins (e.g., Cx32) oligomerize early in the secretory pathway. Amino acids near the cytoplasmic aspect of the third transmembrane domain have previously been shown to determine this difference in assembly sites. Here, we characterized the oligomerization of two connexins expressed prominently in the vasculature, Cx37 and Cx40, using constructs containing a C-terminal dilysine-based ER retention/retrieval signal (HKKSL) or treatment with brefeldin A to block ER vesicle trafficking. Both methods led to intracellular retention of connexins, since the cells lacked gap junction plaques. Retention of Cx40 in the ER prevented it from oligomerizing, comparable to Cx43. By contrast, ER-retained Cx37 was partially oligomerized. Replacement of two amino acids near the third transmembrane domain of Cx43 (L152 and R153) with the corresponding amino acids from Cx37 (M152 and G153) resulted in early oligomerization in the ER. Thus, residues that allow Cx37 to oligomerize early in the secretory pathway could restrict its interactions with coexpressed Cx40 or Cx43 by favoring homomeric oligomerization, providing a structural basis for cells to produce gap junction channels with different connexin composition.


Subject(s)
Amino Acids/metabolism , Cell Membrane/metabolism , Connexins/metabolism , Cytosol/metabolism , Amino Acids/chemistry , Connexin 43/chemistry , Connexin 43/metabolism , Connexins/chemistry , Fluorescent Antibody Technique , Gap Junctions/metabolism , Humans , Protein Multimerization , Structure-Activity Relationship , Gap Junction alpha-4 Protein
11.
Mol Biol Cell ; 20(10): 2593-604, 2009 May.
Article in English | MEDLINE | ID: mdl-19321666

ABSTRACT

Connexin43 (Cx43) is a gap junction protein that forms multimeric channels that enable intercellular communication through the direct transfer of signals and metabolites. Although most multimeric protein complexes form in the endoplasmic reticulum (ER), Cx43 seems to exit from the ER as monomers and subsequently oligomerizes in the Golgi complex. This suggests that one or more protein chaperones inhibit premature Cx43 oligomerization in the ER. Here, we provide evidence that an ER-localized, 29-kDa thioredoxin-family protein (ERp29) regulates Cx43 trafficking and function. Interfering with ERp29 function destabilized monomeric Cx43 oligomerization in the ER, caused increased Cx43 accumulation in the Golgi apparatus, reduced transport of Cx43 to the plasma membrane, and inhibited gap junctional communication. ERp29 also formed a specific complex with monomeric Cx43. Together, this supports a new role for ERp29 as a chaperone that helps stabilize monomeric Cx43 to enable oligomerization to occur in the Golgi apparatus.


Subject(s)
Connexin 43/chemistry , Connexin 43/metabolism , Endoplasmic Reticulum/metabolism , Heat-Shock Proteins/metabolism , Animals , Cell Communication/drug effects , Cell Line , Endoplasmic Reticulum/drug effects , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Hexachlorocyclohexane/pharmacology , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Mice , Models, Biological , Mutant Proteins/metabolism , Protein Binding/drug effects , Protein Structure, Quaternary , Protein Transport/drug effects , RNA, Small Interfering/metabolism , Rats
12.
Am J Physiol Renal Physiol ; 294(6): F1448-52, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18417538

ABSTRACT

Volume depletion due to persistent glucosuria-induced osmotic diuresis is a significant problem in uncontrolled diabetes mellitus (DM). Angiotensin II receptor blockers (ARBs), such as candesartan, slow the progression of chronic kidney disease in patients with DM. However, mice with genetic knockout of components of the renin-angiotensin system have urine concentrating defects, suggesting that ARBs may exacerbate the volume depletion. Therefore, the effect of candesartan on UT-A1, UT-A3, NKCC2, and aquaporin-2 (AQP2) protein abundances was determined in control and 3-wk DM rats. Aldosterone levels in control rats (0.36 +/- 0.06 nM) and candesartan-treated rats (0.34 +/- 0.14 nM) were the same. DM rats had higher aldosterone levels (1.48 +/- 0.37 nM) that were decreased by candesartan (0.97 +/- 0.26 nM). Western analysis showed that UT-A1 expression was increased in DM rats compared with controls in inner medullary (IM) tip (158 +/- 13%) and base (120 +/- 25%). UT-A3 abundance was increased in IM tip (123 +/- 11%) and base (146 +/- 17%) of DM rats vs. controls. UT-A3 was unchanged in candesartan-treated control rats. In candesartan-treated DM rats, UT-A3 increased in IM tip (160 +/- 14%) and base (210 +/- 19%). Candesartan-treated DM rats had slightly higher AQP2 in IM (46%, P < 0.05) vs. control rats. NKCC2/BSC1 was increased 145 +/- 10% in outer medulla of DM vs. control rats. We conclude that candesartan augments compensatory changes in medullary transport proteins, reducing the losses of solute and water during uncontrolled DM. These changes may represent a previously unrecognized beneficial effect of type 1 ARBs in DM.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Benzimidazoles/pharmacology , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Kidney Medulla/drug effects , Kidney Medulla/metabolism , Tetrazoles/pharmacology , Animals , Aquaporin 2/metabolism , Biphenyl Compounds , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Hyperglycemia/complications , Hyperglycemia/metabolism , Kidney Concentrating Ability/drug effects , Male , Membrane Transport Proteins/metabolism , Rats , Rats, Sprague-Dawley , Renin-Angiotensin System/drug effects , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 1 , Water-Electrolyte Balance/drug effects , Urea Transporters
13.
J Biol Chem ; 282(41): 30005-13, 2007 Oct 12.
Article in English | MEDLINE | ID: mdl-17699514

ABSTRACT

Tissue barrier function is directly mediated by tight junction transmembrane proteins known as claudins. Cells that form tight junctions typically express multiple claudin isoforms which suggests that heterotypic (head-to-head) binding between different claudin isoforms may play a role in regulating paracellular permeability. However, little is known about motifs that control heterotypic claudin compatibility. We found that although claudin-3 and claudin-4 were heteromerically compatible when expressed in the same cell, they did not heterotypically interact despite having extracellular loop (EL) domains that are highly conserved at the amino acid level. Claudin-1 and -5, which were heterotypically compatible with claudin-3, did not heterotypically bind to claudin-4. In contrast, claudin-4 chimeras containing either the first EL domain or the second EL domain of claudin-3 were able to heterotypically bind to claudin-1, claudin-3, and claudin-5. Moreover, a single point mutation in the first extracellular loop domain of claudin-3 to convert Asn(44) to the corresponding amino acid in claudin-4 (Thr) produced a claudin capable of heterotypic binding to claudin-4 while still retaining the ability to bind to claudin-1 and -5. Thus, control of heterotypic claudin-claudin interactions is sensitive to small changes in the EL domains.


Subject(s)
Membrane Proteins/chemistry , Amino Acid Motifs , Amino Acid Sequence , Claudin-1 , Claudin-3 , Claudin-4 , Claudin-5 , Epithelial Cells/metabolism , HeLa Cells , Humans , Molecular Sequence Data , Protein Isoforms , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid
14.
J Am Soc Nephrol ; 17(10): 2680-6, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16959825

ABSTRACT

Urea transport, mediated by the urea transporter A1 (UT-A1) and/or UT-A3, is important for the production of concentrated urine. Vasopressin rapidly increases urea transport in rat terminal inner medullary collecting ducts (IMCD). A previous study showed that one mechanism for rapid regulation of urea transport is a vasopressin-induced increase in UT-A1 phosphorylation. This study tests whether vasopressin or directly activating adenylyl cyclase with forskolin also increases UT-A1 accumulation in the plasma membrane of rat IMCD. Inner medullas were harvested from rats 45 min after injection with vasopressin or vehicle. UT-A1 abundance in the plasma membrane was significantly increased in the membrane fraction after differential centrifugation and in the biotinylated protein population. Vasopressin and forskolin each increased the amount of biotinylated UT-A1 in rat IMCD suspensions that were treated ex vivo. The observed changes in the plasma membrane are specific, as the amount of biotinylated UT-A1 but not the calcium-sensing receptor was increased by forskolin. Next, whether forskolin or the V(2)-selective agonist dDAVP would increase apical membrane expression of UT-A1 in MDCK cells that were stably transfected with UT-A1 (UT-A1-MDCK cells) was tested. Forskolin and dDAVP significantly increased UT-A1 abundance in the apical membrane in UT-A1-MDCK cells. It is concluded that vasopressin and forskolin increase UT-A1 accumulation in the plasma membrane in rat IMCD and in the apical plasma membrane of UT-A1-MDCK cells. These findings suggest that vasopressin regulates urea transport by increasing UT-A1 accumulation in the plasma membrane and/or UT-A1 phosphorylation.


Subject(s)
Antidiuretic Agents/pharmacology , Cell Membrane/drug effects , Kidney Medulla/metabolism , Kidney Tubules, Collecting/metabolism , Membrane Transport Proteins/metabolism , Vasopressins/pharmacology , Animals , Biological Transport/drug effects , Cell Membrane/metabolism , Cells, Cultured , Colforsin/pharmacology , Deamino Arginine Vasopressin/pharmacology , Kidney Concentrating Ability/drug effects , Kidney Medulla/drug effects , Kidney Tubules, Collecting/drug effects , Male , Rats , Rats, Sprague-Dawley , Urea Transporters
15.
Am J Physiol Renal Physiol ; 290(6): F1443-52, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16434569

ABSTRACT

The primary mechanism by which the kidneys mediate net acid excretion is through ammonia metabolism. In the current study, we examined whether chronic metabolic acidosis, which increases ammonia metabolism, alters the cell-specific and/or the subcellular expression of the ammonia transporter family member, Rhcg, in the outer medullary collecting duct in the inner stripe (OMCDi). Chronic metabolic acidosis was induced in normal SD rats by HCl ingestion for 7 days; controls were pair-fed. The subcellular distribution of Rhcg was determined using immunogold electron microscopy and morphometric analyses. In intercalated cells, acidosis increased total Rhcg, apical plasma membrane Rhcg, and the proportion of total cellular Rhcg in the apical plasma membrane. Intracellular Rhcg decreased significantly, and basolateral Rhcg was unchanged. Because apical plasma membrane length increased in parallel with apical Rhcg immunolabel, apical plasma membrane Rhcg density was unchanged. In principal cells, acidosis increased total Rhcg, apical plasma membrane Rhcg, and the proportion of total cellular Rhcg in the apical plasma membrane while decreasing the intracellular proportion. In contrast to the intercalated cell, chronic metabolic acidosis did not significantly alter apical boundary length; accordingly, apical plasma membrane Rhcg density increased. In addition, basolateral Rhcg immunolabel increased in response to chronic metabolic acidosis. These results indicate that in the rat OMCDi 1) chronic metabolic acidosis increases apical plasma membrane Rhcg in both the intercalated cell and principal cell where it may contribute to enhanced apical ammonia secretion; 2) increased apical plasma membrane Rhcg results from both increased total protein and changes in the subcellular distribution of Rhcg; 3) the mechanism of Rhcg subcellular redistribution differs in intercalated and principal cells; and 4) Rhcg may contribute to regulated basolateral ammonia transport in the principal cell.


Subject(s)
Acidosis/metabolism , Cation Transport Proteins/analysis , Kidney/ultrastructure , Membrane Glycoproteins/analysis , Subcellular Fractions/chemistry , Acidosis/chemically induced , Ammonia/metabolism , Animals , Cell Membrane/chemistry , Chronic Disease , Cytoplasm/chemistry , Hydrochloric Acid , Immunohistochemistry , Kidney Medulla/chemistry , Kidney Medulla/ultrastructure , Loop of Henle/chemistry , Loop of Henle/ultrastructure , Male , Microscopy, Electron , Rats , Rats, Sprague-Dawley
16.
Am J Physiol Regul Integr Comp Physiol ; 290(5): R1446-59, 2006 May.
Article in English | MEDLINE | ID: mdl-16373440

ABSTRACT

Mammalian urea transporters are facilitated membrane transport proteins belonging to two families, UT-A and UT-B. They are best known for their role of maintaining the renal inner medullary urinary concentrating gradient. Urea transporters have also been identified in tissues not typically associated with urea metabolism. The purpose of this study was to survey the major organs in rat to determine the distribution of UT-A and UT-B mRNA transcripts and protein forms and determine their cellular localization. Five kidney subregions and 17 extrarenal tissues were screened by Northern blot analysis using two UT-A and three UT-B probes and by Western blot analysis using polyclonal COOH-terminal UT-A and UT-B antibodies. Immunohistochemistry was performed on 16 extrarenal tissues using the same antibodies. In kidney, we detected mRNA transcripts and protein bands consistent with previously-identified UT-A and UT-B isoforms, as well as novel forms. We found that UT-A mRNA and protein are widely expressed in extrarenal tissues in various forms that are different from the known isoforms. We determined the cellular localization of UT-A and UT-B in these tissues. We found that both UT-A and UT-B are ubiquitously expressed as numerous tissue-specific mRNA transcripts and protein forms that are localized to cell membranes, cytoplasm, or nuclei.


Subject(s)
Membrane Transport Proteins/biosynthesis , RNA, Messenger/biosynthesis , Animals , Blotting, Northern , Blotting, Western , Brain Chemistry/physiology , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Immunohistochemistry , In Vitro Techniques , Kidney/metabolism , Male , Membrane Transport Proteins/genetics , Rats , Rats, Sprague-Dawley , Reticulocytes/drug effects , Reticulocytes/metabolism , Testis/metabolism , Tissue Distribution , Urea Transporters
SELECTION OF CITATIONS
SEARCH DETAIL
...