Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Bioengineered ; 12(1): 4946-4961, 2021 12.
Article in English | MEDLINE | ID: mdl-34365894

ABSTRACT

The solute carrier family has been reported to play critical roles in the progression of several cancers; however, the relationship between solute carrier family 12 member 8 (SLC12A8) and bladder cancer (BC) has not been clearly confirmed. This study explores the prognostic value of SLC12A8 for BC and its correlation with immune cell infiltration. We found that the expression of SLC12A8 mRNA was significantly overexpressed in BC tissues compared with noncancerous tissues in multiple public databases, and the result was validated using real-time PCR and immunohistochemistry (IHC). The Kaplan-Meier method and Cox proportional hazards models were used to evaluate the prognostic value of SLC12A8 for BC. The high expression of SLC12A8 led to a shorter overall survival time and was an unfavorable prognostic biomarker for BC. The mechanisms of SLC12A8 promoting tumorigenesis were investigated by Gene Set Enrichment Analysis (GSEA). Moreover, the correlations of SLC12A8 expression with the tumor-infiltrating immune cells (TICs) in BC were explored using TIMER 2.0 and CIBERSORT. SLC12A8 was associated with CD4+ T cells, dendritic cells, neutrophils, and macrophages infiltration. The expression of SLC12A8 was positively correlated with crucial immune checkpoint molecules. In conclusion, SLC12A8 might be an unfavorable prognostic biomarker in BC related to tumor immune cell infiltration.


Subject(s)
Sodium-Potassium-Chloride Symporters , Urinary Bladder Neoplasms , Aged , Aged, 80 and over , Biomarkers, Tumor , Cell Line, Tumor , Databases, Genetic , Female , Humans , Male , Middle Aged , Prognosis , Sodium-Potassium-Chloride Symporters/genetics , Sodium-Potassium-Chloride Symporters/immunology , Sodium-Potassium-Chloride Symporters/metabolism , Urinary Bladder/pathology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology
2.
Biol Cell ; 104(11): 658-76, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22708623

ABSTRACT

BACKGROUND INFORMATION: The renal Na(+) -K(+) -2Cl(-) co-transporter (NKCC2) is expressed in kidney thick ascending limb cells, where it mediates NaCl re-absorption regulating body salt levels and blood pressure. RESULTS: In this study, we used a well-characterised NKCC2 construct (c-NKCC2) to identify NKCC2-interacting proteins by an antibody shift assay coupled with blue native/SDS-PAGE and mass spectrometry. Among the interacting proteins, we identified moesin, a protein belonging to ezrin, eadixin and moesin family. Co-immunoprecipitation experiments confirmed that c-NKCC2 interacts with the N-terminal domain of moesin in LLC-PK1 cells. Moreover, c-NKCC2 accumulates in intracellular and sub-apical vesicles in cells transfected with a moesin dominant negative green fluorescent protien (GFP)-tagged construct. In addition, moesin knock-down by short interfering RNA decreases by about 50% c-NKCC2 surface expression. Specifically, endocytosis and exocytosis assays showed that moesin knock-down does not affect c-NKCC2 internalisation but strongly reduces exocytosis of the co-transporter. CONCLUSIONS: Our data clearly demonstrate that moesin plays a critical role in apical membrane insertion of NKCC2, suggesting a possible involvement of moesin in regulation of Na(+) and Cl(-) absorption in the kidney.


Subject(s)
Microfilament Proteins/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Cell Membrane/metabolism , Cell Movement , Cells, Cultured , Endocytosis/physiology , Epithelial Cells/metabolism , Exocytosis/physiology , Gene Knockdown Techniques , Kidney/metabolism , Microfilament Proteins/genetics , Protein Binding , Protein Transport/physiology , Rats , Sodium-Potassium-Chloride Symporters/immunology , Solute Carrier Family 12, Member 1 , Swine/metabolism
3.
Article in English | MEDLINE | ID: mdl-19751844

ABSTRACT

The sublingual salt gland is the primary site of salt excretion in sea snakes; however, little is known about the mechanisms mediating ion excretion. Na(+)/K(+)-ATPase (NKA) and Na(+)/K(+)/2Cl(-) cotransporter (NKCC) are two proteins known to regulate membrane potential and drive salt secretion in most vertebrate secretory cells. We hypothesized that NKA and NKCC would localize to the basolateral membranes of the principal cells comprising the tubular epithelia of sea snake salt glands. Although there is evidence of NKA activity in salt glands from several species of sea snake, the localization of NKA and NKCC and other potential ion transporters remains unstudied. Using histology and immunohistochemistry, we localized NKA and NKCC in salt glands from three species of laticaudine sea snake: Laticauda semifasciata, L. laticaudata, and L. colubrina. Antibody specificity was confirmed using Western blots. The compound tubular glands of all three species were found to be composed of serous secretory epithelia, and NKA and NKCC were abundant in the basolateral membranes. These results are consistent with the morphology of secretory epithelia found in the rectal salt glands of marine elasmobranchs, the nasal glands of marine birds and the gills of teleost fishes, suggesting a similar function in regulating ion secretion.


Subject(s)
Elapidae/metabolism , Salt Gland/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Antibody Specificity , Blotting, Western , Epithelium/enzymology , Epithelium/metabolism , Salt Gland/enzymology , Sodium-Potassium-Chloride Symporters/immunology , Sodium-Potassium-Exchanging ATPase/immunology
4.
Am J Physiol Renal Physiol ; 292(2): F723-35, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17062845

ABSTRACT

Growth hormone (GH) has antidiuretic and antinatriuretic effects in rats and humans, but the molecular mechanisms responsible for these effects are unknown. The aim of this study was to investigate the mechanisms behind the acute renal effects of GH in rats. Female rats received rat (r)GH (2.8 mg/kg sc) or saline and were placed in metabolic cages for 5 h. Urinary excretion of electrolytes and urinary volume were reduced after rGH injection, while urine osmolality was increased. Creatinine and lithium clearance remained unchanged, suggesting that rGH increases reabsorption in segments distal to the proximal tubule. Total plasma insulin-like growth factor I (IGF-I) levels did not change, while cortical IGF-I mRNA abundance was increased. The relative abundance of total and Ser(256)-phosphorylated aquaporin 2 was found to be unchanged by immunoblotting, whereas a significant increase of Thr(96) and Thr(101)-phosphorylated NKCC2 (renal Na(+), K(+), 2Cl(-) cotransporter) was found in the inner stripe of outer medulla thick ascending limbs (mTAL). Additionally, an increased NKCC2 expression was observed in the cortical region. Immunohistochemistry confirmed these findings. The density of NKCC2 molecules in the apical membrane of mTAL cells appeared to be unchanged after rGH injection evaluated by immunoelectron microscopy. Basolateral addition of rGH or IGF-I to microperfused rat mTAL segments did not change transepithelial voltage. In conclusion, GH appears to exert its acute antinatriuretic and antidiuretic effects through indirect activation of NKCC2 in the mTAL.


Subject(s)
Diuresis/drug effects , Growth Hormone/administration & dosage , Kidney/drug effects , Natriuresis/drug effects , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Aquaporin 2/metabolism , Cell Membrane/chemistry , Female , Insulin/pharmacology , Insulin-Like Growth Factor I/biosynthesis , Insulin-Like Growth Factor I/metabolism , Kidney/ultrastructure , Phosphorylation , Rats , Rats, Wistar , Sodium-Potassium-Chloride Symporters/immunology , Solute Carrier Family 12, Member 1
5.
J Neurocytol ; 34(1-2): 11-36, 2005 Mar.
Article in English | MEDLINE | ID: mdl-16374707

ABSTRACT

The sequentially activated molecules of olfactory signal-onset are mostly concentrated in the long, thin distal parts of olfactory epithelial receptor cell cilia. Is this also true for molecules of olfactory signal-termination and -regulation? G-protein receptor kinase 3 (GRK3) supposedly aids in signal desensitization at the level of odor receptors, whereas beta-arrestin-2, Ca2+/calmodulin-dependent protein kinase II (CaMKII) and phosphodiesterase (PDE) PDE1C2 are thought to do so at the level of the adenylyl cyclase, ACIII. The Na+, K(+)-2Cl(-)-cotransporter NKCC1 regulates Cl(-)-channel activity. In an attempt to localize the subcellular sites olfactory signal-termination and -regulation we used four antibodies to GRK3, two to beta-arrestin-2, five to CaMKII (one to both the alpha and beta form, and two each specific to CaMKII alpha and beta), two to PDE1C2, and three to Cl(-)-cotransporters. Only antibodies to Cl(-)-cotransporters labeled cytoplasmic compartments of, especially, supporting cells but also those of receptor cells. For all other antibodies, immunoreactivity was mostly restricted to the olfactory epithelial luminal border, confirming light microscopic studies that had shown that antibodies to GRK3, beta- arrestin-2, CaMKII, and PDE1C2 labeled this region. Labeling did indeed include receptor cell cilia but occurred in microvilli of neighboring supporting cells as well. Apical parts of microvillous cells that are distinct from supporting cells, and also of ciliated respiratory cells, immunoreacted slightly with most antibodies. When peptides were available, antibody preabsorption with an excess of peptide reduced labeling intensities. Though some of the antibodies did label apices and microvilli of vomeronasal (VNO) supporting cells, none immunoreacted with VNO sensory structures.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/analysis , Olfactory Mucosa/chemistry , Olfactory Mucosa/enzymology , Phosphoric Diester Hydrolases/analysis , Protein Serine-Threonine Kinases/analysis , Sodium-Potassium-Chloride Symporters/analysis , Animals , Antibodies/analysis , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/immunology , Cilia/chemistry , Cilia/enzymology , Cilia/ultrastructure , G-Protein-Coupled Receptor Kinase 3 , Immunohistochemistry , Mice , Microscopy, Electron, Transmission , Microvilli/chemistry , Microvilli/enzymology , Microvilli/ultrastructure , Olfactory Mucosa/ultrastructure , Phosphoric Diester Hydrolases/immunology , Protein Serine-Threonine Kinases/immunology , Rats , Rats, Sprague-Dawley , Receptors, Odorant/analysis , Sodium-Potassium-Chloride Symporters/immunology
6.
Nephron Exp Nephrol ; 96(3): e67-76, 2004.
Article in English | MEDLINE | ID: mdl-15056983

ABSTRACT

BACKGROUND/AIMS: Whether the postobstructive natriuresis and diuresis is related with an altered regulation of sodium transporters in the kidney was examined. METHODS: Male Sprague-Dawley rats underwent either bilateral (BUO) or unilateral obstruction (UUO) of the proximal ureters for 24 h. The expression of Na,K-ATPase, type-3 sodium-hydrogen exchanger (NHE3), type-1 bumetanide-sensitive sodium cotransporter (BSC1), and thiazide-sensitive sodium cotransporter (TSC) proteins was determined in the obstructed kidney by Western blot analysis and immunohistochemistry. Catalytic activity of Na,K-ATPase was also determined. RESULTS: The expression of alpha1 and beta1 subunit proteins and the catalytic activity of Na,K-ATPase were significantly decreased in the obstructed kidney in BUO. The expressions of NHE3, BSC1 and TSC proteins were also significantly decreased. Immunohistochemistry confirmed the downregulation of these sodium transporters in the obstructed kidney. In UUO, the expression of sodium transporters was similarly decreased in the obstructed kidney. CONCLUSION: The postobstructive natriuresis and diuresis may in part be accounted for by a reduced abundance of sodium transporters in the kidney.


Subject(s)
Ion Pumps/metabolism , Kidney/metabolism , Natriuresis , Symporters , Animals , Carrier Proteins/immunology , Carrier Proteins/metabolism , Diuresis , Immunohistochemistry , Ligation , Male , Rats , Rats, Sprague-Dawley , Receptors, Drug/immunology , Receptors, Drug/metabolism , Sodium Chloride Symporters , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/immunology , Sodium-Hydrogen Exchangers/metabolism , Sodium-Potassium-Chloride Symporters/immunology , Sodium-Potassium-Chloride Symporters/metabolism , Sodium-Potassium-Exchanging ATPase/immunology , Sodium-Potassium-Exchanging ATPase/metabolism , Solute Carrier Family 12, Member 1 , Solute Carrier Family 12, Member 3 , Ureter/surgery
7.
J Biol Chem ; 277(40): 37551-8, 2002 Oct 04.
Article in English | MEDLINE | ID: mdl-12145305

ABSTRACT

The Na-K-Cl cotransporter NKCC1 is activated by phosphorylation of a regulatory domain in its N terminus. In the accompanying paper (Darman, R. B., and Forbush, B. (2002) J. Biol. Chem. 277, 37542-37550), we identify three phosphothreonines important in this process. Using a phospho-specific antibody (anti-phospho-NKCC1 antibody R5) raised against a diphosphopeptide containing Thr(212) and Thr(217) of human NKCC, we were readily able to monitor the cotransporter activation state. In (32)P phosphorylation experiments with rectal gland tubules, we show that the R5 antibody signal is proportional to the amount of (32)P incorporated into NKCC1; and in experiments with NKCC1-transfected HEK-293 cells, we demonstrate that R5-detected phosphorylation directly mirrors functional activation. Immunofluorescence analysis of shark rectal gland shows activation-dependent R5 antibody staining along the basolateral membrane. In perfused rat parotid glands, isoproterenol induced staining of both acinar and ductal cells along the basolateral membrane. Isoproterenol also induced basolateral staining of the epithelial cells in rat trachea, whereas basal cells in the subepithelial tissue displayed heavy, non-polarized staining of the cell membrane. In rat colon, agonist stimulation induced staining along the basolateral membrane of crypt cells. These data provide direct evidence of NKCC1 regulation in these tissues, and they further link phosphorylation of NKCC1 with its activation in transfected cells and native tissue. The high conservation of the regulatory threonine residues among NKCC1, NKCC2, and NCC family members, together with the fact that tissues from divergent vertebrate species exhibit similar R5-binding profiles, lends further support to the role of this regulatory locus in vivo.


Subject(s)
Antibody Specificity , Phosphates/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Amino Acid Sequence , Animals , Binding Sites , Colon/drug effects , Colon/physiology , Dipeptides/chemistry , Epinephrine/pharmacology , Humans , Ion Transport , Isoproterenol/pharmacology , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphopeptides/chemistry , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sodium-Potassium-Chloride Symporters/chemistry , Sodium-Potassium-Chloride Symporters/immunology , Solute Carrier Family 12, Member 2 , Threonine , Trachea/physiology
8.
Eur J Neurosci ; 15(2): 233-45, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11849291

ABSTRACT

GABA, a major inhibitory neurotransmitter, depolarizes hippocampal pyramidal neurons during the first postnatal week. These depolarizations result from an efflux of Cl- through GABAA-gated anion channels. The outward Cl- gradient that provides the driving force for Cl- efflux might be generated and maintained by the Na+, K+, 2Cl- cotransporter (NKCC) that keeps intracellular Cl- concentration above electrochemical equilibrium. The developmental pattern of expression of the cotransporter in the hippocampus is not known. We studied the postnatal distribution pattern of NKCC in the hippocampus using a monoclonal antibody (T4) against a conserved epitope in the C-terminus of the cotransporter molecule. We also examined the temporal relationships between the developmental pattern of NKCC expression and the formation of perisomatic GABAergic synapses. This study was aimed at determining, with antivesicular inhibitory amino acid transporter (VIAAT) antibodies, whether perisomatic GABAergic synapses are formed preferentially at the time when GABA is depolarizing. During the first postnatal week, NKCC immunolabelling was restricted to cell bodies in the pyramidal cell layer and in the strata oriens and radiatum. In contrast, at postnatal day 21 (P21) and in adult animals little or no labelling occurred in cell bodies; instead, a prominent dendritic labelling appeared in both pyramidal and nonpyramidal neurons. The ultrastructural immunogold study in P21 rat hippocampi corroborated the light-microscopy results. In addition, this study revealed that a portion of the silver-intensified colloidal gold particles were located on neuronal plasmalemma, as expected for a functional cotransporter. The formation of inhibitory synapses on perikarya of the pyramidal cell layer was a late process. The density of VIAAT-immunoreactive puncta in the stratum pyramidale at P21 reached four times the P7 value in CA3, and six times the P7 value in CA1. Electron microscopy revealed that the number of synapses per neuronal perikaryal profile in the stratum pyramidale of the CA3 area at P21 was three times higher than at P7, even if a concomitant 20% increase in the area of these neuronal perikaryal profiles occurred. It is concluded that, in hippocampal pyramidal cells, there is a developmental shift in the NKCC localization from a predominantly somatic to a predominantly dendritic location. The presence of NKCC during the first postnatal week is consistent with the hypothesis that this transporter might be involved in the depolarizing effects of GABA. The depolarizing effects of GABA may not be required for the establishment of the majority of GABAergic synapses in the stratum pyramidale, because their number increases after the first postnatal week, when GABA action becomes hyperpolarizing.


Subject(s)
Amino Acid Transport Systems , Hippocampus/growth & development , Hippocampus/metabolism , Neural Inhibition/physiology , Sodium-Potassium-Chloride Symporters/biosynthesis , Synapses/metabolism , Vesicular Transport Proteins , Animals , Antibodies , Carrier Proteins/analysis , Carrier Proteins/immunology , Female , Immunohistochemistry , Microscopy, Electron , Pyramidal Cells/chemistry , Pyramidal Cells/metabolism , Pyramidal Cells/ultrastructure , Rats , Rats, Wistar , Sodium-Potassium-Chloride Symporters/analysis , Sodium-Potassium-Chloride Symporters/immunology , Synapses/chemistry , Synapses/ultrastructure , Vesicular Inhibitory Amino Acid Transport Proteins
SELECTION OF CITATIONS
SEARCH DETAIL