Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Gastroenterology ; 138(7): 2410-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20197068

ABSTRACT

BACKGROUND & AIMS: Mechanisms for age restriction of rotavirus diarrhea are unclear. Because rotavirus primarily infects small intestine, colonic contribution has not been widely studied. Recent data suggest that colonic secretion postbacterial infection is mediated by galanin-1 receptors (Gal1-R). We evaluated age-dependent expression of Gal1-R in Rhesus rotavirus (RRV)-infected mice and its contribution to fluid secretion. METHODS: Twenty-four hours after infection of C57BL/6J mice (wild type or Gal1-R knockout) with RRV or vehicle, closed small intestinal and colon loops were constructed. Net fluid content of the loops was calculated (milligrams/centimeters) at 2 hours post-treatment with galanin, galanin antibody, or lidocaine. Gal1-R expression was quantified by automated chromogen analysis. RESULTS: Viral antigen was detected in small intestinal epithelial cells but not in colon. Developmental Gal1-R was widely expressed in the small intestine but minimally in the colon. Postinfection, markedly increased Gal1-R was seen in the colon but not after day 25. Galanin caused a significantly higher increase in the net fluid content of infected colon than small intestine. Treatment with lidocaine reduced net fluid secretion in the small intestine and the colon. Mean diarrheal scores were significantly reduced in Gal1-R knockout mice compared with wild type (1.19 +/- 0.31, n = 22 vs 3.36 +/- 0.50, n = 35, P = .0001). CONCLUSIONS: These data show that RRV infection of the small intestine increases colonic secretion through Gal1-R and provide a promising start toward understanding the age restriction of rotavirus diarrhea.


Subject(s)
Colon/metabolism , Intestine, Small/virology , Receptor, Galanin, Type 1/physiology , Rotavirus Infections/metabolism , Age Factors , Animals , Colon/virology , Diarrhea/etiology , Galanin/pharmacology , Lidocaine/pharmacology , Mice , Mice, Inbred C57BL , Rotavirus Infections/complications
2.
Am J Physiol Renal Physiol ; 295(5): F1463-71, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18753297

ABSTRACT

Progressive renal enlargement due to the growth of innumerable fluid-filled cysts is a central pathophysiological feature of autosomal dominant polycystic kidney disease (ADPKD). These epithelial neoplasms enlarge slowly and damage noncystic parenchyma by mechanisms that have not been clearly defined. In a microarray analysis of cultured human ADPKD cyst epithelial cells, periostin mRNA was overexpressed 15-fold compared with normal human kidney (NHK) cells. Periostin, initially identified in osteoblasts, is not expressed in normal adult kidneys but is expressed transiently during renal development. We found periostin in cyst-lining cells in situ in the extracellular matrix adjacent to the cysts and within cyst fluid. ADPKD cells secreted periostin across luminal and basolateral plasma membranes. Periostin increased proliferation of cyst epithelial cells 27.9 +/- 3.1% (P < 0.001) above baseline and augmented in vitro cyst growth but did not affect proliferation of normal renal cells. Expression of alphaV-integrin, a periostin receptor, was ninefold higher in ADPKD cells compared with NHK cells, and antibodies that block alphaV-integrin inhibited periostin-induced cell proliferation. We conclude that periostin is a novel autocrine mitogen secreted by mural epithelial cells with the potential to accelerate cyst growth and promote interstitial remodeling in ADPKD.


Subject(s)
Cell Adhesion Molecules/physiology , Cell Proliferation , Integrin alphaV/physiology , Antibodies/immunology , Antibodies/pharmacology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/pharmacology , Cell Cycle/drug effects , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/pharmacology , Cyst Fluid/metabolism , Cysts/metabolism , Cysts/pathology , Epidermal Growth Factor/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Extracellular Matrix/metabolism , Flow Cytometry , Gene Expression/drug effects , Humans , Immunohistochemistry , Integrin alphaV/genetics , Integrin alphaV/immunology , Kidney/metabolism , Kidney/pathology , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology
3.
J Am Soc Nephrol ; 17(1): 178-87, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16319189

ABSTRACT

Polycystic kidney disease (PKD) is a lethal disorder characterized by progressive expansion of renal cysts. Genetic mutations associated with PKD are thought to disrupt intracellular Ca2+ regulation, leading to abnormal proliferation of tubule epithelial cells. cAMP stimulates the B-Raf/MEK/extracellular signal-regulated kinase (B-Raf/MEK/ERK) pathway and accelerates the proliferation of cells that are cultured from PKD cysts. By contrast, cAMP inhibits the proliferation of cells from normal human kidneys (NHK) and M-1 mouse collecting duct cells. Previously, it was found that a sustained reduction of intracellular Ca2+ levels in NHK and M-1 cells that were treated with Ca2+ entry blockers allowed cAMP activation of the B-Raf/MEK/ERK pathway, switching the cells to a cAMP-growth stimulated phenotype. In this study, primary cultures of cyst epithelial cells from autosomal dominant (ADPKD) and recessive (ARPKD) PKD kidneys were used to determine whether controlled addition of Ca2+ could reverse the aberrant mitogenic response to cAMP. Steady-state intracellular Ca2+ levels were found to be 20 nM lower in cyst-derived ADPKD cells (57 +/- 2 nM) compared with NHK cells (77 +/- 2 nM). Treatment of ADPKD cells or ARPKD cells with either Bay K8644, a Ca2+ channel activator, or A23187, a Ca2+ ionophore, caused sustained increases in intracellular Ca2+ levels and completely reversed the mitogenic response to cAMP. Elevation of intracellular Ca2+ levels in ADPKD cells increased Akt activity and blocked cAMP-dependent B-Raf and ERK activation. Thus, increases in [Ca2+]i are able to restore the normal anti-mitogenic response to cAMP in cells that are derived from two genetically distinct forms of PKD.


Subject(s)
Calcium/metabolism , Polycystic Kidney Diseases/pathology , Cell Proliferation , Cells, Cultured , Cyclic AMP/pharmacology , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/physiology , Humans , Kidney/metabolism , MAP Kinase Kinase Kinases/physiology , Phenotype , Polycystic Kidney Diseases/metabolism , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Recessive/metabolism , Proto-Oncogene Proteins B-raf/physiology , Signal Transduction
4.
J Virol ; 79(6): 3509-16, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15731245

ABSTRACT

Osteopontin (OPN) is a sialated phosphoprotein found in tissues and secreted into body fluids. It is an integrin ligand with pleiotropic functions as an extracellular matrix protein in mineralized tissues and a cytokine that is active in cell signaling (A. B. Tuck, C. Hota, S. M. Wilson, and A. F. Chambers, Oncogene 22:1198-1205, 2003). To determine whether OPN may be important in mucosal defense against viral pathogens, we evaluated the OPN response to rotavirus infection and the extent of diarrhea manifested by infected opn null mutant (opn-/-) mice. Reverse transcription-PCR, Northern and Western blots, and immunohistochemical studies of the HT-29 intestinal epithelial cell line and murine intestine were used to evaluate OPN mRNA and product. Intestinal closed loops and diarrheal observations determined disease severity and duration. OPN mRNA levels increased after infection of HT-29 cells, peaking in 4 to 6 h. Infected cultures contained 925 microg of OPN/ml, while for controls the levels were below detection (50 microg/ml). Infection increased OPN mRNA levels in intestinal tissue between 2 and 24 h postinoculation and increased OPN protein in intestinal fluid. The cellular localization of OPN was supranuclear and apical, and responding cells were diffusely distributed on the villus surface. Three days after infection, closed intestinal loops from opn-/- mice contained more fluid than loops from controls, although secretion levels at the onset of illness were similar. Null mutant mice experienced more intense and prolonged diarrhea than controls. Rotavirus infection of intestinal epithelial cells and murine intestine caused marked increases in OPN mRNA levels and secreted OPN protein. OPN-deficient mice suffered prolonged disease.


Subject(s)
Cytokines/immunology , Diarrhea/physiopathology , Intestinal Mucosa/immunology , Intestinal Mucosa/virology , Intestines/immunology , Rotavirus Infections/physiopathology , Sialoglycoproteins/immunology , Animals , Blotting, Northern , Blotting, Western , Cytokines/metabolism , Diarrhea/immunology , Diarrhea/pathology , Diarrhea/virology , Disease Models, Animal , Female , HT29 Cells , Humans , Intestinal Mucosa/metabolism , Intestines/pathology , Intestines/virology , Mice , Mice, Knockout , Osteopontin , Pregnancy , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Rotavirus/growth & development , Rotavirus Infections/immunology , Rotavirus Infections/pathology , Rotavirus Infections/virology , Sialoglycoproteins/metabolism
5.
J Biol Chem ; 279(53): 55455-64, 2004 Dec 31.
Article in English | MEDLINE | ID: mdl-15466861

ABSTRACT

Regulation of intracellular Ca(2+) mobilization has been associated with the functions of polycystin-1 (PC1) and polycystin-2 (PC2), the protein products of the PKD1 and PKD2 genes. We have now demonstrated that PC1 can activate the calcineurin/NFAT (nuclear factor of activated T-cells) signaling pathway through Galpha(q) -mediated activation of phospholipase C (PLC). Transient transfection of HEK293T cells with an NFAT promoter-luciferase reporter demonstrated that membrane-targeted PC1 constructs containing the membrane proximal region of the C-terminal tail, which includes the heterotrimeric G protein binding and activation domain, can stimulate NFAT luciferase activity. Inhibition of glycogen synthase kinase-3beta by LiCl treatment further increased PC1-mediated NFAT activity. PC1-mediated activation of NFAT was completely inhibited by the calcineurin inhibitor, cyclosporin A. Cotransfection of a construct expressing the Galpha(q) subunit augmented PC1-mediated NFAT activity, whereas the inhibitors of PLC (U73122) and the inositol trisphosphate and ryanodine receptors (xestospongin and 2-aminophenylborate) and a nonspecific Ca(2+) channel blocker (gadolinium) diminished PC1-mediated NFAT activity. PC2 was not able to activate NFAT. An NFAT-green fluorescent protein nuclear localization assay demonstrated that PC1 constructs containing the C-tail only or the entire 11-transmembrane spanning region plus C-tail induced NFAT-green fluorescent protein nuclear translocation. NFAT expression was demonstrated in the M-1 mouse cortical collecting duct cell line and in embryonic and adult mouse kidneys by reverse transcriptase-PCR and immunolocalization. These data suggest a model in which PC1 signaling leads to a sustained elevation of intracellular Ca(2+) mediated by PC1 activation of Galpha(q) followed by PLC activation, release of Ca(2+) from intracellular stores, and activation of store-operated Ca(2+) entry, thus activating calcineurin and NFAT.


Subject(s)
Calcineurin/metabolism , Proteins/physiology , Signal Transduction , Active Transport, Cell Nucleus , Animals , Blotting, Western , Boronic Acids/pharmacology , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels , Cell Line , Cell Nucleus/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Estrenes/pharmacology , Gadolinium/pharmacology , Genes, Reporter , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Inositol 1,4,5-Trisphosphate Receptors , Kidney/embryology , Kidney/metabolism , Lithium Chloride/pharmacology , Luciferases/metabolism , Macrocyclic Compounds , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Microscopy, Fluorescence , NFATC Transcription Factors , Oxazoles/pharmacology , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Pyrrolidinones/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ryanodine Receptor Calcium Release Channel/metabolism , TRPP Cation Channels , Time Factors , Tissue Distribution , Transfection , Type C Phospholipases/metabolism
6.
J Biol Chem ; 279(39): 40419-30, 2004 Sep 24.
Article in English | MEDLINE | ID: mdl-15263001

ABSTRACT

cAMP can be either mitogenic or anti-mitogenic, depending on the cell type. We demonstrated previously that cAMP inhibited the proliferation of normal renal epithelial cells and stimulated the proliferation of cells derived from the cysts of polycystic kidney disease (PKD) patients. The protein products of the genes causing PKD, polycystin-1 and polycystin-2, are thought to regulate intracellular calcium levels, suggesting that abnormal polycystin function may affect calcium signaling and thus cause a switch to the cAMP growth-stimulated phenotype. To test this hypothesis, we disrupted intracellular calcium mobilization by treating immortalized mouse M-1 collecting duct cells and primary cultures of human kidney epithelial cells with calcium channel blockers and by lowering extracellular calcium with EGTA. Calcium restriction for 3-5 h converted both cell types from a normal cAMP growth-inhibited phenotype to an abnormal cAMP growth-stimulated phenotype, characteristic of PKD. In M-1 cells, we showed that calcium restriction was associated with an elevation in B-Raf protein levels and cAMP-stimulated, Ras-dependent activation of B-Raf and ERK. Moreover, the activity of Akt, a negative regulator of B-Raf, was decreased by calcium restriction. Inhibition of Akt or phosphatidylinositol 3-kinase also allowed cAMP-dependent activation of B-Raf and ERK in normal calcium. These results suggest that calcium restriction causes an inhibition of the phosphatidylinositol 3-kinase/Akt pathway, which relieves the inhibition of B-Raf to allow the cAMP growth-stimulated phenotypic switch. Finally, M-1 cells stably overexpressing an inducible polycystin-1 C-terminal cytosolic tail construct were shown to exhibit a cAMP growth-stimulated phenotype involving B-Raf and ERK activation, which was reversed by the calcium ionophore A23187. We conclude that disruption of calcium mobilization in cells that are normally growth-inhibited by cAMP can derepress the B-Raf/ERK pathway, thus converting these cells to a phenotype that is growth-stimulated by cAMP.


Subject(s)
Calcium/metabolism , Cyclic AMP/metabolism , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Animals , Blotting, Western , Calcimycin/pharmacology , Calcium Channel Blockers/pharmacology , Cell Division , Cell Line , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Egtazic Acid/pharmacology , Enzyme Activation , Epithelial Cells , Humans , Ionophores/pharmacology , Kidney/cytology , Kidney/metabolism , Membrane Proteins/metabolism , Mice , Models, Biological , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Polycystic Kidney Diseases/metabolism , Proteins/metabolism , Proto-Oncogene Proteins B-raf , TRPP Cation Channels , Time Factors , Transfection , Verapamil/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL