Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Am J Physiol Cell Physiol ; 325(1): C344-C361, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37125773

ABSTRACT

Kidney stones (KSs) are very common, excruciating, and associated with tremendous healthcare cost, chronic kidney disease (CKD), and kidney failure (KF). Most KSs are composed of calcium oxalate and small increases in urinary oxalate concentration significantly enhance the stone risk. Oxalate also potentially contributes to CKD progression, kidney disease-associated cardiovascular diseases, and poor renal allograft survival. This emphasizes the urgent need for plasma and urinary oxalate lowering therapies, which can be achieved by enhancing enteric oxalate secretion. We previously identified Oxalobacter formigenes (O. formigenes)-derived factors secreted in its culture-conditioned medium (CM), which stimulate oxalate transport by human intestinal Caco2-BBE (C2) cells and reduce urinary oxalate excretion in hyperoxaluric mice by enhancing colonic oxalate secretion. Given their remarkable therapeutic potential, we now identified Sel1-like proteins as the major O. formigenes-derived secreted factors using mass spectrometry and functional assays. Crystal structures for six proteins were determined to confirm structures and better understand functions. OxBSel1-14-derived small peptides P8 and P9 were identified as the major factors, with P8 + 9 closely recapitulating the CM's effects, acting through the oxalate transporters SLC26A2 and SLC26A6 and PKA activation. Besides C2 cells, P8 + 9 also stimulate oxalate transport by human ileal and colonic organoids, confirming that they work in human tissues. In conclusion, P8 and P9 peptides are identified as the major O. formigenes-derived secreted factors and they have significant therapeutic potential for hyperoxalemia, hyperoxaluria, and related disorders, impacting the outcomes of patients suffering from KSs, enteric hyperoxaluria, primary hyperoxaluria, CKD, KF, and renal transplant recipients.NEW & NOTEWORTHY We previously identified Oxalobacter formigenes-derived secreted factors stimulating oxalate transport by human intestinal epithelial cells in vitro and reducing urinary oxalate excretion in hyperoxaluric mice by enhancing colonic oxalate secretion. We now identified Sel1-like proteins and small peptides as the major secreted factors and they have significant therapeutic potential for hyperoxalemia and hyperoxaluria, impacting the outcomes of patients suffering from kidney stones, primary and secondary hyperoxaluria, chronic kidney disease, kidney failure, and renal transplant recipients.


Subject(s)
Hyperoxaluria , Kidney Calculi , Kidney Transplantation , Renal Insufficiency, Chronic , Renal Insufficiency , Humans , Mice , Animals , Oxalobacter formigenes/metabolism , Caco-2 Cells , Oxalates/metabolism , Hyperoxaluria/metabolism , Kidney Calculi/metabolism , Epithelial Cells/metabolism , Peptides/metabolism , Renal Insufficiency/metabolism , Renal Insufficiency, Chronic/metabolism
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 ; 318(2): C372-C379, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31825656

ABSTRACT

Most kidney stones are composed of calcium oxalate, and small increases in urine oxalate enhance the stone risk. The mammalian intestine plays a crucial role in oxalate homeostasis, and we had recently reported that Oxalobacter-derived factors stimulate oxalate transport by human intestinal Caco2-BBE (C2) cells through PKA activation. We therefore evaluated whether intestinal oxalate transport is directly regulated by activation of the PKA signaling pathway. To this end, PKA was activated with forskolin and IBMX (F/I). F/I significantly stimulated (3.7-fold) [14C]oxalate transport by C2 cells [≥49% of which is mediated by the oxalate transporter SLC26A6 (A6)], an effect completely blocked by the PKA inhibitor H89, indicating that it is PKA dependent. PKA stimulation of intestinal oxalate transport is not cell line specific, since F/I similarly stimulated oxalate transport by the human intestinal T84 cells. F/I significantly increased (2.5-fold) A6 surface protein expression by use of immunocytochemistry. Assessing [14C]oxalate transport as a function of increasing [14C]oxalate concentration in the flux medium showed that the observed stimulation is due to a F/I-induced increase (1.8-fold) in Vmax and reduction (2-fold) in Km. siRNA knockdown studies showed that significant components of the observed stimulation are mediated by A6 and SLC26A2 (A2). Besides enhancing A6 surface protein expression, it is also possible that the observed stimulation is due to PKA-induced enhanced A6 and/or A2 transport activity in view of the reduced Km. We conclude that PKA activation positively regulates oxalate transport by intestinal epithelial cells and that PKA agonists might therapeutically impact hyperoxalemia, hyperoxaluria, and related kidney stones.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Intestinal Mucosa/metabolism , Oxalates/metabolism , Signal Transduction/physiology , Animals , Caco-2 Cells , Cell Line, Tumor , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Hyperoxaluria/metabolism , Intestinal Mucosa/drug effects , Intracellular Signaling Peptides and Proteins/pharmacology , Ion Transport/physiology , Kidney Calculi/metabolism , Signal Transduction/drug effects
4.
Am J Physiol Gastrointest Liver Physiol ; 316(1): G1-G14, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30307745

ABSTRACT

Most kidney stones (KS) are composed of calcium oxalate and small increases in urine oxalate enhance the stone risk. Obesity is a risk factor for KS, and urinary oxalate excretion increases with increased body size. We previously established the obese ob/ob ( ob) mice as a model (3.3-fold higher urine oxalate) to define the pathogenesis of obesity-associated hyperoxaluria (OAH). The purpose of this study was to test the hypothesis that the obesity-associated enhanced small intestinal paracellular permeability contributes to OAH by increasing passive paracellular intestinal oxalate absorption. ob Mice have significantly higher jejunal (1.6-fold) and ileal (1.4-fold) paracellular oxalate absorption ex vivo and significantly higher (5-fold) urine [13C]oxalate following oral gavage with [13C]oxalate, indicating increased intestinal oxalate absorption in vivo. The observation of higher oxalate absorption in vivo compared with ex vivo suggests the possibility of increased paracellular permeability along the entire gut. Indeed, ob mice have significantly higher fractions of the administered sucrose (1.7-fold), lactulose (4.4-fold), and sucralose (3.1-fold) excreted in the urine, reflecting increased gastric, small intestinal, and colonic paracellular permeability, respectively. The ob mice have significantly reduced gastrointestinal occludin, zonula occludens-1, and claudins-1 and -3 mRNA and total protein expression. Proinflammatory cytokines and oxidative stress, which are elevated in obesity, significantly enhanced paracellular intestinal oxalate absorption in vitro and ex vivo. We conclude that obese mice have significantly higher intestinal oxalate absorption and enhanced gastrointestinal paracellular permeability in vivo, which would likely contribute to the pathogenesis of OAH, since there is a transepithelial oxalate concentration gradient to drive paracellular intestinal oxalate absorption. NEW & NOTEWORTHY This study shows that the obese ob/ob mice have significantly increased gastrointestinal paracellular oxalate absorption and remarkably enhanced paracellular permeability along the entire gut in vivo, which are likely mediated by the obesity-associated increased systemic and intestinal inflammation and oxidative stress. A transepithelial oxalate concentration gradient driving gastrointestinal paracellular oxalate absorption exists, and therefore, our novel findings likely contribute to the hyperoxaluria observed in the ob/ob mice and hence to the pathogenesis of obesity-associated hyperoxaluria.


Subject(s)
Gastrointestinal Tract/metabolism , Hyperoxaluria/physiopathology , Intestinal Mucosa/metabolism , Obesity/metabolism , Animals , Inflammation/metabolism , Intestinal Absorption/physiology , Intestine, Small/metabolism , Jejunum/metabolism , Mice, Inbred C57BL , Permeability
6.
Am J Physiol Cell Physiol ; 315(5): C687-C698, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30020825

ABSTRACT

Most kidney stones (KS) are composed of calcium oxalate, and small increases in urine oxalate affect the stone risk. Intestinal oxalate secretion mediated by anion exchanger SLC26A6 (PAT1) plays a crucial role in limiting net absorption of ingested oxalate, thereby preventing hyperoxaluria and related KS, reflecting the importance of understanding regulation of intestinal oxalate transport. We previously showed that ATP and UTP inhibit oxalate transport by human intestinal Caco2-BBE cells (C2). Since ATP is rapidly degraded to adenosine (ADO), we examined whether intestinal oxalate transport is regulated by ADO. We measured [14C]oxalate uptake in the presence of an outward Cl gradient as an assay of Cl-oxalate exchange activity, ≥49% of which is PAT1-mediated in C2 cells. We found that ADO significantly inhibited oxalate transport by C2 cells, an effect completely blocked by the nonselective ADO receptor antagonist 8- p-sulfophenyltheophylline. ADO also significantly inhibited oxalate efflux by C2 cells, which is important since PAT1 mediates oxalate efflux in vivo. Using pharmacological antagonists and A2B adenosine receptor (A2B AR) siRNA knockdown studies, we observed that ADO inhibits oxalate transport through the A2B AR, phospholipase C, and PKC. ADO inhibits oxalate transport by reducing PAT1 surface expression as shown by biotinylation studies. We conclude that ADO inhibits oxalate transport by lowering PAT1 surface expression in C2 cells through signaling pathways including the A2B AR, PKC, and phospholipase C. Given higher ADO levels and overexpression of the A2B AR in inflammatory bowel disease (IBD), our findings have potential relevance to pathophysiology of IBD-associated hyperoxaluria and related KS.


Subject(s)
Adenosine/metabolism , Amino Acid Transport Systems/genetics , Inflammatory Bowel Diseases/genetics , Receptor, Adenosine A2B/genetics , Symporters/genetics , Adenosine/administration & dosage , Adenosine A2 Receptor Antagonists/administration & dosage , Adenosine Triphosphate/metabolism , Biological Transport/genetics , Caco-2 Cells , Humans , Hyperoxaluria/genetics , Hyperoxaluria/metabolism , Hyperoxaluria/pathology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/metabolism , Intestines/drug effects , Kidney Calculi/genetics , Kidney Calculi/metabolism , Kidney Calculi/pathology , Oxalates/metabolism , Receptor, Adenosine A2B/metabolism , Risk Factors , Signal Transduction/drug effects , Theophylline/administration & dosage , Type C Phospholipases/genetics
7.
Kidney Int ; 93(5): 1098-1107, 2018 05.
Article in English | MEDLINE | ID: mdl-29395336

ABSTRACT

Most kidney stones are composed of calcium oxalate, and minor changes in urine oxalate affect the stone risk. Obesity is a risk factor for kidney stones and a positive correlation of unknown etiology between increased body size, and elevated urinary oxalate excretion has been reported. Here, we used obese ob/ob (ob) mice to elucidate the pathogenesis of obesity-associated hyperoxaluria. These ob mice have significant hyperoxaluria (3.3-fold) compared with control mice, which is not due to overeating as shown by pair-feeding studies. Dietary oxalate removal greatly ameliorated this hyperoxaluria, confirming that it is largely enteric in origin. Transporter SLC26A6 (A6) plays an essential role in active transcellular intestinal oxalate secretion, and ob mice have significantly reduced jejunal A6 mRNA (- 80%) and total protein (- 62%) expression. While net oxalate secretion was observed in control jejunal tissues mounted in Ussing chambers, net absorption was seen in ob tissues, due to significantly reduced secretion. We hypothesized that the obesity-associated increase in intestinal and systemic inflammation, as reflected by elevated proinflammatory cytokines, suppresses A6-mediated intestinal oxalate secretion and contributes to obesity-associated hyperoxaluria. Indeed, proinflammatory cytokines (elevated in ob mice) significantly decreased intestinal oxalate transport in vitro by reducing A6 mRNA and total protein expression. Proinflammatory cytokines also significantly reduced active mouse jejunal oxalate secretion, converting oxalate transport from net secretion in vehicle-treated tissues to net absorption in proinflammatory cytokines-treated tissues. Thus, reduced active intestinal oxalate secretion, likely secondary to local and systemic inflammation, contributes to the pathogenesis of obesity-associated hyperoxaluria. Hence, proinflammatory cytokines represent potential therapeutic targets.


Subject(s)
Hyperoxaluria/etiology , Intestinal Secretions/metabolism , Jejunum/metabolism , Obesity/complications , Oxalates/metabolism , Animals , Antiporters/metabolism , Caco-2 Cells , Cytokines/metabolism , Disease Models, Animal , Down-Regulation , Humans , Hyperoxaluria/metabolism , Hyperoxaluria/physiopathology , Inflammation Mediators/metabolism , Intestinal Absorption , Jejunum/physiopathology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Obesity/metabolism , Obesity/physiopathology , Secretory Pathway , Sulfate Transporters/metabolism
8.
J Am Soc Nephrol ; 28(3): 876-887, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27738124

ABSTRACT

Hyperoxaluria is a major risk factor for kidney stones and has no specific therapy, although Oxalobacter formigenes colonization is associated with reduced stone risk. O. formigenes interacts with colonic epithelium and induces colonic oxalate secretion, thereby reducing urinary oxalate excretion, via an unknown secretagogue. The difficulties in sustaining O. formigenes colonization underscore the need to identify the derived factors inducing colonic oxalate secretion. We therefore evaluated the effects of O. formigenes culture conditioned medium (CM) on apical 14C-oxalate uptake by human intestinal Caco-2-BBE cells. Compared with control medium, O. formigenes CM significantly stimulated oxalate uptake (>2.4-fold), whereas CM from Lactobacillus acidophilus did not. Treating the O. formigenes CM with heat or pepsin completely abolished this bioactivity, and selective ultrafiltration of the CM revealed that the O. formigenes-derived factors have molecular masses of 10-30 kDa. Treatment with the protein kinase A inhibitor H89 or the anion exchange inhibitor 4,4'-diisothiocyano-2,2'-stilbenedisulfonic acid completely blocked the CM-induced oxalate transport. Knockdown of the oxalate transporter SLC26A6 also significantly restricted the induction of oxalate transport by CM. In a mouse model of primary hyperoxaluria type 1, rectal administration of O. formigenes CM significantly reduced (>32.5%) urinary oxalate excretion and stimulated (>42%) distal colonic oxalate secretion. We conclude that O. formigenes-derived bioactive factors stimulate oxalate transport in intestinal cells through mechanisms including PKA activation. The reduction in urinary oxalate excretion in hyperoxaluric mice treated with O. formigenes CM reflects the in vivo retention of biologic activity and the therapeutic potential of these factors.


Subject(s)
Biological Factors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Oxalates/metabolism , Oxalobacter formigenes , Animals , Humans , Hyperoxaluria/metabolism , Ion Transport , Male , Mice
9.
J Am Soc Nephrol ; 24(7): 1104-13, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23766534

ABSTRACT

Inherited and acquired disorders that enhance the activity of transporters mediating renal tubular Na(+) reabsorption are well established causes of hypertension. It is unclear, however, whether primary activation of an Na(+)-independent chloride transporter in the kidney can also play a pathogenic role in this disease. Here, mice overexpressing the chloride transporter pendrin in intercalated cells of the distal nephron (Tg(B1-hPDS) mice) displayed increased renal absorption of chloride. Compared with normal mice, these transgenic mice exhibited a delayed increase in urinary NaCl and ultimately, developed hypertension when exposed to a high-salt diet. Administering the same sodium intake as NaHCO3 instead of NaCl did not significantly alter BP, indicating that the hypertension in the transgenic mice was chloride-sensitive. Moreover, excessive chloride absorption by pendrin drove parallel absorption of sodium through the epithelial sodium channel ENaC and the sodium-driven chloride/bicarbonate exchanger (Ndcbe), despite an appropriate downregulation of these sodium transporters in response to the expanded vascular volume and hypertension. In summary, chloride transport in the distal nephron can play a primary role in driving NaCl transport in this part of the kidney, and a primary abnormality in renal chloride transport can provoke arterial hypertension. Thus, we conclude that the chloride/bicarbonate exchanger pendrin plays a major role in controlling net NaCl absorption, thereby influencing BP under conditions of high salt intake.


Subject(s)
Blood Pressure/physiology , Chloride-Bicarbonate Antiporters/metabolism , Chlorides/metabolism , Hypertension/metabolism , Kidney/metabolism , Membrane Transport Proteins/metabolism , Nephrons/metabolism , Sodium Chloride/metabolism , Animals , Humans , Immunohistochemistry , Ion Transport , Mice , Mice, Transgenic , Sulfate Transporters
10.
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
11.
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
12.
Comput Intell Neurosci ; 2022: 8491753, 2022.
Article in English | MEDLINE | ID: mdl-35855801

ABSTRACT

Dyslexia is among the most common neurological disorders in children. Detection of dyslexia therefore remains an important pursuit for the research works across various domains which is illustrated by the plethora of work presented in diverse scientific articles. The work presented herein attempted to utilize the potential of a unified gaming test of subjects (dyslexia/controls) in tandem with principal components derived from data to detect dyslexia. The work aims to build a machine learning model for dyslexia detection using comprehensive gaming test data. We have attempted to explore the potential of various kernel functions of the Support Vector Machine (SVM) on different number of principal components to reduce the computational complexity. A detection accuracy of 92% is obtained from the radial basis function with 5 components, and the highest detection accuracy obtained from the radial basis function with 3 components is 93%. On the contrary, the Artificial Neural Network(ANN) shows an added advantage with minimal number of hyperparameters with 3 components for obtaining an accuracy of 95%. The comparison of the proposed method with some of the existing works shows efficacy of this method for dyslexia detection.


Subject(s)
Dyslexia , Machine Learning , Child , Dyslexia/diagnosis , Humans , Neural Networks, Computer , Support Vector Machine
13.
Mt Sinai J Med ; 71(4): 281-4, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15365595

ABSTRACT

BACKGROUND: The low serum anion gap (AG) in patients with hepatic cirrhosis is generally attributed to hypoalbuminemia. Serum immunoglobulin G (IgG) (elevated in chronic viral hepatitis) and IgA (elevated in alcoholic cirrhosis) have different isoelectric points, and thus may affect serum AG in opposite directions. AIM: To define the normal serum AG in patients with liver cirrhosis of diverse etiologies. STUDY DESIGN: We retrospectively compared serum AG of 144 stable cirrhotics and 286 control patients (consecutive hospital admissions with serum creatinine concentration < 2 mg/dL). RESULTS: Serum AG was significantly lower among the cirrhotics, compared to the controls (5.8 +/- 2.2 mEq/L vs. 7.0 +/- 2.2 mEq/L, respectively, p<0.005). However, when patients with serum albumin concentration < 3.5 g/dL were excluded, there was no significant difference between the cirrhotics vs. controls (6.7 +/- 1.8 mEq/L vs. 7.0 +/- 2.2 mEq/L, p=ns). Moreover, patients with liver cirrhosis secondary to chronic viral hepatitis had AG similar to that of the alcoholic cirrhotics (5.6 +/- 2.5 mEq/L vs. 6.0 +/- 1.9 mEq/L, p=ns). There was a positive correlation between serum albumin concentrations > 1.9 g/dL and serum AG, and a tendency toward an inverse correlation between serum globulin concentration and serum AG. CONCLUSION: Our results support the contention that hypoalbuminemia accounts for the decreased serum AG frequently observed in patients with liver cirrhosis. We found no difference in serum AG with different causes of cirrhosis. We also suggest a lower reference range for normal serum AG.


Subject(s)
Acid-Base Equilibrium/physiology , Liver Cirrhosis/blood , Alcoholism/blood , Alcoholism/complications , Female , Hepatitis, Viral, Human/blood , Hepatitis, Viral, Human/complications , Humans , Liver Cirrhosis/etiology , Male , Middle Aged , Reference Values , Retrospective Studies
14.
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
15.
J Clin Invest ; 120(5): 1627-35, 2010 May.
Article in English | MEDLINE | ID: mdl-20389022

ABSTRACT

Regulation of sodium balance is a critical factor in the maintenance of euvolemia, and dysregulation of renal sodium excretion results in disorders of altered intravascular volume, such as hypertension. The amiloride-sensitive epithelial sodium channel (ENaC) is thought to be the only mechanism for sodium transport in the cortical collecting duct (CCD) of the kidney. However, it has been found that much of the sodium absorption in the CCD is actually amiloride insensitive and sensitive to thiazide diuretics, which also block the Na-Cl cotransporter (NCC) located in the distal convoluted tubule. In this study, we have demonstrated the presence of electroneutral, amiloride-resistant, thiazide-sensitive, transepithelial NaCl absorption in mouse CCDs, which persists even with genetic disruption of ENaC. Furthermore, hydrochlorothiazide (HCTZ) increased excretion of Na+ and Cl- in mice devoid of the thiazide target NCC, suggesting that an additional mechanism might account for this effect. Studies on isolated CCDs suggested that the parallel action of the Na+-driven Cl-/HCO3- exchanger (NDCBE/SLC4A8) and the Na+-independent Cl-/HCO3- exchanger (pendrin/SLC26A4) accounted for the electroneutral thiazide-sensitive sodium transport. Furthermore, genetic ablation of SLC4A8 abolished thiazide-sensitive NaCl transport in the CCD. These studies establish what we believe to be a novel role for NDCBE in mediating substantial Na+ reabsorption in the CCD and suggest a role for this transporter in the regulation of fluid homeostasis in mice.


Subject(s)
Chloride-Bicarbonate Antiporters/metabolism , Kidney Tubules, Collecting/metabolism , Sodium-Bicarbonate Symporters/metabolism , Sodium/chemistry , Amiloride/pharmacology , Animals , Electrophysiology/methods , Hydrochlorothiazide/pharmacology , Kidney/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Biological , Oocytes/metabolism , Sodium Chloride Symporter Inhibitors/pharmacology , Xenopus
16.
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
17.
Kidney Int ; 65(3): 897-903, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14871409

ABSTRACT

BACKGROUND: Homologous down-regulation/desensitization of the parathyroid hormone receptor (PTH1R)/adenylate cyclase system has been demonstrated in uremia, and may contribute to parathyroid hormone (PTH) resistance; however, additional studies have shown that parathyroidectomy fails to normalize the down-regulation of the PTH1R. The present studies were designed to test directly, in vitro, the hypothesis that factors circulating in the uremic environment, other than PTH, decrease the response of osteoblastic cells to PTH. METHODS: Studies were conducted in confluent cultures of UMR 106-01 osteoblast-like cells. Uremic ultrafiltrate (UUF) was obtained from patients on hemodialysis. Cells were exposed to media containing 50% uremic ultrafiltrate for periods of up to 72 hours. Control cultures were exposed to a buffered salt solution containing a comparable ionic composition to that of the UUF. PTH-stimulated cyclic adenosine monophosphate (cAMP) generation was determined by radioimmunoassay (RIA), PTH binding and PTH1R mRNA levels were determined by radioligand binding and Northern analysis, respectively. RESULTS: PTH-stimulated cAMP generation from cultures treated with uremic ultrafiltrate for 48 hours was 1385.8 +/- 183.2 pmol/culture/5 minutes, whereas control cultures generated 2389.5 +/- 271 pmol cAMP/culture/5 minutes (P < 0.05). PTH binding was decreased by 30% in cultures incubated with UUF as compared to controls. The decrease in binding induced by UUF was accompanied by a decrease in PTH1R mRNA levels. CONCLUSION: These findings demonstrate that factors present in UUF decrease PTH-stimulated cAMP generation by a mechanism that involves a decrease in the levels of PTH1R mRNA levels. Thus, the skeletal resistance to PTH in the setting of chronic kidney disease, may be explained, at least in part, by circulating factors other than PTH.


Subject(s)
Hemodialysis Solutions/pharmacology , Osteoblasts/drug effects , Osteoblasts/physiology , Receptors, Parathyroid Hormone/genetics , Teriparatide/analogs & derivatives , Uremia/physiopathology , Binding, Competitive , Cells, Cultured , Cyclic AMP/metabolism , Gene Expression Regulation , Humans , Kidney Failure, Chronic/physiopathology , Kidney Failure, Chronic/therapy , Osteoblasts/cytology , Parathyroid Hormone/metabolism , Parathyroid Hormone/pharmacology , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , RNA, Messenger/metabolism , Receptor, Parathyroid Hormone, Type 1 , Receptors, Parathyroid Hormone/metabolism , Renal Dialysis , Teriparatide/metabolism , Teriparatide/pharmacology , Uremia/therapy
18.
Proc Natl Acad Sci U S A ; 101(7): 2064-9, 2004 Feb 17.
Article in English | MEDLINE | ID: mdl-14769928

ABSTRACT

Mutations in the serine-threonine kinase WNK4 [with no lysine (K) 4] cause pseudohypoaldosteronism type II, a Mendelian disease featuring hypertension with hyperkalemia. In the kidney, WNK4 regulates the balance between NaCl reabsorption and K(+) secretion via variable inhibition of the thiazide-sensistive NaCl cotransporter and the K(+) channel ROMK. We now demonstrate expression of WNK4 mRNA and protein outside the kidney. In extrarenal tissues, WNK4 is found almost exclusively in polarized epithelia, variably associating with tight junctions, lateral membranes, and cytoplasm. Epithelia expressing WNK4 include sweat ducts, colonic crypts, pancreatic ducts, bile ducts, and epididymis. WNK4 is also expressed in the specialized endothelium of the blood-brain barrier. These epithelia and endothelium all play important roles in Cl(-) transport. Because WNK4 is known to regulate renal Cl(-) handling, we tested WNK4's effect on the activity of mediators of epithelial Cl(-) flux whose extrarenal expression overlaps with WNK4. WNK4 proved to be a potent inhibitor of the activity of both the Na(+)-K(+)-2Cl(-) cotransporter (NKCC1) and the Cl(-)/base exchanger SLC26A6 (CFEX) (>95% inhibition of NKCC1-mediated (86)Rb influx, P < 0.001; >80% inhibition of CFEX-mediated [(14)C] formate uptake, P < 0.001), mediators of Cl(-) flux across basolateral and apical membranes, respectively. In contrast, WNK4 showed no inhibition of pendrin, a related Cl(-)/base exchanger. These findings indicate a general role for WNK4 in the regulation of electrolyte flux in diverse epithelia. Moreover, they reveal that WNK4 regulates the activities of a diverse group of structurally unrelated ion channels, cotransporters, and exchangers.


Subject(s)
Cell Polarity , Chlorides/metabolism , Epithelium/metabolism , Membrane Transport Proteins , Protein Serine-Threonine Kinases/metabolism , Animals , Blotting, Western , Carrier Proteins/genetics , Carrier Proteins/metabolism , Humans , Immunohistochemistry , Ion Transport , Kidney/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Oocytes/metabolism , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sodium-Potassium-Chloride Symporters/genetics , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 2 , Sulfate Transporters , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL