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
2.
Curr Opin Nephrol Hypertens ; 30(2): 264-274, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33427760

ABSTRACT

PURPOSE OF REVIEW: The gut-kidney axis plays a critical role in oxalate homeostasis, and better understanding of oxalate transport regulatory mechanisms is essential for developing novel therapies. RECENT FINDINGS: Oxalate potentially contributes to chronic kidney disease (CKD) progression, CKD - and end stage renal disease (ESRD)-associated cardiovascular diseases, polycystic kidney disease (PKD) progression, and/or poor renal allograft survival, emphasizing the need for plasma and urinary oxalate lowering therapies. One promising strategy would be to enhance the bowel's ability to secrete oxalate, which might be facilitated by the following findings. Oxalobacter formigenes (O. formigenes)-derived factors recapitulate O. formigenes colonization effects by reducing urinary oxalate excretion in hyperoxaluric mice by inducing colonic oxalate secretion. Protein kinase A activation stimulates intestinal oxalate transport by enhancing the surface expression of the oxalate transporter SLC26A6 (A6). Glycosylation also stimulates A6-mediated oxalate transport. The colon adapts to chronic acidosis in rats through increased colonic oxalate secretion as previously reported in CKD rats, and A6-mediated enteric oxalate secretion is critical in reducing the body oxalate burden in CKD mice. Intestinal oxalate transport is negatively regulated by proinflammatory cytokines and cholinergic, purinergic, and adenosinergic signaling. SUMMARY: These findings could facilitate the development of novel therapeutics for hyperoxalemia, hyperoxaluria, and related disorders if similar regulatory mechanisms are confirmed in humans.


Subject(s)
Kidney Transplantation , Oxalates , Animals , Antiporters , Homeostasis , Humans , Kidney , Mice , Oxalobacter formigenes , Rats , Sulfate Transporters
3.
Am J Physiol Cell Physiol ; 305(1): C78-89, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23596171

ABSTRACT

Nephrolithiasis remains a major health problem in Western countries. Seventy to 80% of kidney stones are composed of calcium oxalate, and small changes in urinary oxalate affect risk of kidney stone formation. Intestinal oxalate secretion mediated by the anion exchanger SLC26A6 plays an essential role in preventing hyperoxaluria and calcium oxalate nephrolithiasis, indicating that understanding the mechanisms regulating intestinal oxalate transport is critical for management of hyperoxaluria. Purinergic signaling modulates several intestinal processes through pathways including PKC activation, which we previously found to inhibit Slc26a6 activity in mouse duodenal tissue. We therefore examined whether purinergic stimulation with ATP and UTP affects oxalate transport by human intestinal Caco-2-BBe (C2) cells. We measured [¹4C]oxalate uptake in the presence of an outward Cl⁻ gradient as an assay of Cl⁻/oxalate exchange activity, ≥50% of which is mediated by SLC26A6. We found that ATP and UTP significantly inhibited oxalate transport by C2 cells, an effect blocked by the PKC inhibitor Gö-6983. Utilizing pharmacological agonists and antagonists, as well as PKC-δ knockdown studies, we observed that ATP inhibits oxalate transport through the P2Y2 receptor, PLC, and PKC-δ. Biotinylation studies showed that ATP inhibits oxalate transport by lowering SLC26A6 surface expression. These findings are of potential relevance to pathophysiology of inflammatory bowel disease-associated hyperoxaluria, where supraphysiological levels of ATP/UTP are expected and overexpression of the P2Y2 receptor has been reported. We conclude that ATP and UTP inhibit oxalate transport by lowering SLC26A6 surface expression in C2 cells through signaling pathways including the P2Y2 purinergic receptor, PLC, and PKC-δ.


Subject(s)
Oxalates/metabolism , Protein Kinase C-delta/metabolism , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Animals , Biological Transport/drug effects , Biological Transport/physiology , Caco-2 Cells , Enzyme Activation , Gene Knockdown Techniques , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Protein Kinase C-delta/genetics , Receptors, Purinergic P2Y2/genetics , Receptors, Purinergic P2Y2/metabolism , Sulfate Transporters , Uridine Triphosphate/genetics , Uridine Triphosphate/metabolism
4.
Am J Physiol Cell Physiol ; 302(1): C46-58, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21956166

ABSTRACT

Urolithiasis remains a very common disease in Western countries. Seventy to eighty percent of kidney stones are composed of calcium oxalate, and minor changes in urinary oxalate affect stone risk. Intestinal oxalate secretion mediated by anion exchanger SLC26A6 plays a major constitutive role in limiting net absorption of ingested oxalate, thereby preventing hyperoxaluria and calcium oxalate urolithiasis. Using the relatively selective PKC-δ inhibitor rottlerin, we had previously found that PKC-δ activation inhibits Slc26a6 activity in mouse duodenal tissue. To identify a model system to study physiologic agonists upstream of PKC-δ, we characterized the human intestinal cell line T84. Knockdown studies demonstrated that endogenous SLC26A6 mediates most of the oxalate transport by T84 cells. Cholinergic stimulation with carbachol modulates intestinal ion transport through signaling pathways including PKC activation. We therefore examined whether carbachol affects oxalate transport in T84 cells. We found that carbachol significantly inhibited oxalate transport by T84 cells, an effect blocked by rottlerin. Carbachol also led to significant translocation of PKC-δ from the cytosol to the membrane of T84 cells. Using pharmacological inhibitors, we observed that carbachol inhibits oxalate transport through the M(3) muscarinic receptor and phospholipase C. Utilizing the Src inhibitor PP2 and phosphorylation studies, we found that the observed regulation downstream of PKC-δ is partially mediated by c-Src. Biotinylation studies revealed that carbachol inhibits oxalate transport by reducing SLC26A6 surface expression. We conclude that carbachol negatively regulates oxalate transport by reducing SLC26A6 surface expression in T84 cells through signaling pathways including the M(3) muscarinic receptor, phospholipase C, PKC-δ, and c-Src.


Subject(s)
Calcium Oxalate/antagonists & inhibitors , Cholinergic Antagonists/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/physiology , Signal Transduction/physiology , Calcium Oxalate/metabolism , Carbachol/pharmacology , Cell Line , Cholinergic Antagonists/metabolism , Humans , Intestinal Mucosa/cytology , Membrane Transport Proteins/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Transport/drug effects , Protein Transport/physiology , Receptor, Muscarinic M3/metabolism , Signal Transduction/drug effects , Sulfate Transporters
5.
Am J Physiol Cell Physiol ; 292(4): C1485-92, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17151144

ABSTRACT

SLC26A6 (CFEX, PAT1) is an anion exchanger expressed in several tissues including renal proximal tubule, pancreatic duct, small intestine, liver, stomach, and heart. It has recently been reported that PKC activation inhibits A6-mediated Cl/HCO(3) exchange by disrupting binding of carbonic anhydrase to A6. However, A6 can operate in HCO(3)-independent exchange modes of physiological importance, as A6-mediated Cl/oxalate exchange plays important roles in proximal tubule NaCl reabsorption and intestinal oxalate secretion. We therefore examined whether PKC activation affects HCO(3)-independent exchange modes of Slc26a6 functionally expressed in Xenopus oocytes. We found that PKC activation inhibited Cl/formate exchange mediated by Slc26a6 but failed to inhibit the related anion exchanger pendrin (SLC26A4) under identical conditions. PKC activation inhibited Slc26a6-mediated Cl/formate exchange, Cl/oxalate exchange, and Cl/Cl exchange to a similar extent. The inhibitor sensitivity profile and the finding that PMA-induced inhibition was calcium independent suggested a potential role for PKC-delta. Indeed, the PKC-delta-selective inhibitor rottlerin significantly blocked PMA-induced inhibition of Slc26a6 activity. Localization of Slc26a6 by immunofluorescence microscopy demonstrated that exposure to PKC activation led to redistribution of Slc26a6 from the oocyte plasma membrane to the intracellular compartment immediately below it. We also observed that PMA decreased the pool of Slc26a6 available to surface biotinylation but had no effect on total Slc26a6 expression. The physiological significance of these findings was supported by the observation that PKC activation inhibited mouse duodenal oxalate secretion, an effect blocked by rottlerin. We conclude that multiple modes of anion exchange mediated by Slc26a6 are negatively regulated by PKC-delta activation.


Subject(s)
Antiporters/metabolism , Protein Kinase C-delta/physiology , Acetophenones/pharmacology , Animals , Anion Transport Proteins/metabolism , Benzopyrans/pharmacology , Biological Transport, Active , Carbazoles/pharmacology , Cell Membrane/metabolism , Chlorides/metabolism , Cytoplasm/metabolism , Enzyme Activation , Female , Formates/metabolism , In Vitro Techniques , Indoles/pharmacology , Maleimides/pharmacology , Mice , Oocytes/metabolism , Oxalates/metabolism , Protein Kinase C-delta/antagonists & inhibitors , Protein Transport , Sulfate Transporters , Tetradecanoylphorbol Acetate/pharmacology , Xenopus
6.
ASAIO J ; 48(3): 316-9, 2002.
Article in English | MEDLINE | ID: mdl-12059008

ABSTRACT

To study the effect of the reversal of the blood ports on blood flow rate (QB), percentage recirculation, and blood urea nitrogen (BUN) clearance, we compared 12 well functioning chronic hemodialysis catheters (7 PermCaths and 5 Tesio Twin Catheters) in both standard and reversed blood flow setups. The reversal of PermCath ports caused no change in the QB (307+/-20 ml/min vs. 314+/-9 ml/min, standard vs. reversed hook-up, respectively), but a significant increase in percentage recirculation (2.5+/-1.8% vs. 12+/-4.6%, standard vs. reversed hook-up, respectively, p = 0.02). Reversal of the Tesio Twin Catheter ports caused a significant decline in QB (296+/-13 ml/min vs. 250+/-16 ml/min, standard vs. reversed hook-up, respectively, p = 0.02), but no significant change in percentage recirculation (2.8+/-1.4% vs. 3.8+/-2.5%, standard vs. reversed hook-up, respectively, p = not significant). Reversal of the ports caused no significant change in BUN clearance with the PermCath (264+/-18 ml/min vs. 257+/-17, standard vs. reversed hook-up, respectively, p = 0.8), but a significant decline in BUN clearance with the Tesio Twin Catheter (247+/-11 ml/min vs. 216+/-13.5 ml/min, standard vs. reversed hook-up, respectively, p = 0.015). In conclusion, our results suggest that reversed hook-up of a well functioning Tesio Twin Catheter is associated with a significant decline in QB and BUN clearance, but no change in percentage recirculation; however, inadvertent reversed hook-up of a well functioning PermCath can lead to a considerable increase in percentage recirculation but no change in QB or BUN clearance.


Subject(s)
Catheterization, Central Venous/instrumentation , Catheterization , Renal Dialysis/instrumentation , Adult , Aged , Blood Circulation , Blood Urea Nitrogen , Female , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...