Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Am J Physiol Renal Physiol ; 319(3): F366-F379, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32657159

ABSTRACT

Carbonic anhydrase (CAII) binds to the electrogenic basolateral Na+-[Formula: see text] cotransporter (NBCe1) and facilitates [Formula: see text] reabsorption across the proximal tubule. However, whether the inhibition of CAII with acetazolamide (ACTZ) alters NBCe1 activity and interferes with the ammoniagenesis pathway remains elusive. To address this issue, we compared the renal adaptation of rats treated with ACTZ to NH4Cl loading for up to 2 wk. The results indicated that ACTZ-treated rats exhibited a sustained metabolic acidosis for up to 2 wk, whereas in NH4Cl-loaded rats, metabolic acidosis was corrected within 2 wk of treatment. [Formula: see text] excretion increased by 10-fold in NH4Cl-loaded rats but only slightly (1.7-fold) in ACTZ-treated rats during the first week despite a similar degree of acidosis. Immunoblot experiments showed that the protein abundance of glutaminase (4-fold), glutamate dehydrogenase (6-fold), and SN1 (8-fold) increased significantly in NH4Cl-loaded rats but remained unchanged in ACTZ-treated rats. Na+/H+ exchanger 3 and NBCe1 proteins were upregulated in response to NH4Cl loading but not ACTZ treatment and were rather sharply downregulated after 2 wk of ACTZ treatment. ACTZ causes renal [Formula: see text] wasting and induces metabolic acidosis but inhibits the upregulation of glutamine transporter and ammoniagenic enzymes and thus suppresses ammonia synthesis and secretion in the proximal tubule, which prevented the correction of acidosis. This effect is likely mediated through the inhibition of the CA-NBCe1 metabolon complex, which results in cell alkalinization. During chronic ACTZ treatment, the downregulation of both NBCe1 and Na+/H+ exchanger 3, along with the inhibition of ammoniagenesis and [Formula: see text] generation, contributes to the maintenance of metabolic acidosis.


Subject(s)
Acetazolamide/pharmacology , Acidosis/metabolism , Ammonia/metabolism , Bicarbonates/metabolism , Diuretics/pharmacology , Kidney/drug effects , Acetazolamide/pharmacokinetics , Acid-Base Equilibrium/drug effects , Adaptation, Physiological , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Animals , Chlorides/blood , Diuretics/pharmacokinetics , Electrolytes/blood , Gene Expression Regulation/drug effects , Half-Life , Kidney/metabolism , Kidney/pathology , Male , Rats , Rats, Sprague-Dawley , Urinalysis
2.
Am J Physiol Renal Physiol ; 316(4): F743-F757, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30623725

ABSTRACT

Chronic adenine feeding is extensively used to develop animal models of chronic renal failure with metabolic features resembling those observed in humans. However, the mechanism by which adenine induces renal failure is poorly understood. In this study, we examined the early effects of adenine on water metabolism and salt balance in rats placed in metabolic cages and fed control or adenine-containing diets for 7 days. Molecular and functional studies demonstrated that adenine-fed rats exhibited a significant reduction in food intake, polyuria, polydipsia, decreased urine osmolality, and increased salt wasting. These effects are independent of changes in food intake and result from a coordinated downregulation of water channel aquaporin-2 (AQP2) and salt transporter (Na+-K+-Cl- cotransporter 2; NKCC2) in the collecting duct and medullary thick ascending limb, respectively. As a result, adenine-fed rats exhibited massive volume depletion, as indicated by a significant body weight loss, increased blood urea nitrogen, and increased hematocrit and hemoglobin levels, all of which were significantly corrected with NaCl replacement. Adenine-induced urinary concentrating defect was not corrected by exogenous arginine vasopressin (AVP), and it correlated with reduced cAMP production in vivo and in vitro. In conclusion, adenine acts on renal tubules as a signaling molecule and causes nephrogenic diabetes insipidus with salt wasting, at least, by directly interfering with AVP V2 receptor signaling with subsequent downregulation of NKCC2 and AQP2 in the kidney. The combination of renal fluid loss and decreased food intake with subsequent massive volume depletion likely plays an important role in the development of early prerenal failure that progresses to chronic kidney disease in long-term adenine feeding.


Subject(s)
Adenine/toxicity , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Kidney/drug effects , Signal Transduction/drug effects , Animals , Aquaporin 2/antagonists & inhibitors , Arginine Vasopressin/pharmacology , Cyclic AMP/metabolism , Diet , Dose-Response Relationship, Drug , Eating , Kidney/pathology , Kidney Diseases/pathology , Male , Osmolar Concentration , Rats , Rats, Sprague-Dawley , Sodium Chloride/pharmacology , Solute Carrier Family 12, Member 1/antagonists & inhibitors , Water/metabolism , Water-Electrolyte Balance/drug effects
3.
Drug Metab Dispos ; 47(3): 184-188, 2019 03.
Article in English | MEDLINE | ID: mdl-30578276

ABSTRACT

A recombinant humanized anticocaine monoclonal antibody, h2E2, has shown potential in the preclinical phases for the treatment of cocaine abuse. The standard tests for cocaine usage are the detection of benzoylecgonine (BE) and cocaine in the urine. This includes workplace drug screens as well as in clinical trials for potential treatments of cocaine abuse. By sequestering cocaine into the plasma compartment, h2E2 prevents cocaine from entering the brain. Due to the altered disposition of cocaine in the presence of h2E2, we investigated the effects of h2E2 on cocaine and metabolite levels in the urine of rats to clarify the use of BE as an endpoint measurement for effectiveness in future clinical trials. The urine concentrations of cocaine and metabolites were considerably altered in the presence of h2E2. After a single injection of h2E2 (120 mg/kg) and cocaine hydrochloride (0.56 mg/kg), the concentration of cocaine and BE excreted into the urine of rats decreased by 92% and 91%, respectively, from vehicle controls. Due to the significant decrease in urinary excretion, BE is not an appropriate indicator of cocaine usage in the presence of h2E2. Another endpoint measurement must be selected for the measurement of cocaine usage in the upcoming clinical trials of h2E2. In contrast to the effects on cocaine and BE urinary excretion, there was a 3-fold increase in ecgonine methyl ester (EME) in the presence of h2E2. Therefore, we conclude that EME is a more appropriate measurement of cocaine intake in the presence of h2E2.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Cocaine-Related Disorders/diagnosis , Cocaine/urine , Renal Elimination/drug effects , Substance Abuse Detection/methods , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Cocaine/analogs & derivatives , Cocaine/antagonists & inhibitors , Cocaine/metabolism , Cocaine-Related Disorders/drug therapy , Cocaine-Related Disorders/urine , Disease Models, Animal , Humans , Male , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use
4.
Am J Physiol Renal Physiol ; 311(2): F249-59, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27194721

ABSTRACT

Estrogen treatment causes renal phosphate (Pi) wasting and hypophosphatemia in rats and humans; however, the signaling mechanisms mediating this effect are still poorly understood. To determine the specific roles of estrogen receptor isoforms (ERα and ERß) and the Klotho pathway in mediating these effects, we studied the effects of estrogen on renal Pi handling in female mice with null mutations of ERα or ERß or Klotho and their wild type (WT) using balance studies in metabolic cages. Estrogen treatment of WT and ERß knockout (KO) mice caused a significant reduction in food intake along with increased renal phosphate wasting. The latter resulted from a significant downregulation of NaPi-IIa and NaPi-IIc protein abundance. The mRNA expression levels of both transporters were unchanged in estrogen-treated mice. These effects on both food intake and renal Pi handling were absent in ERα KO mice. Estrogen treatment of Klotho KO mice or parathyroid hormone (PTH)-depleted thyroparathyroidectomized mice exhibited a significant downregulation of NaPi-IIa with no change in the abundance of NaPi-IIc. Estrogen treatment of a cell line (U20S) stably coexpressing both ERα and ERß caused a significant downregulation of NaPi-IIa protein when transiently transfected with a plasmid containing full-length or open-reading frame (ORF) 3'-untranslated region (UTR) but not 5'-UTR ORF of mouse NaPi-IIa transcript. In conclusion, estrogen causes phosphaturia and hypophosphatemia in mice. These effects result from downregulation of NaPi-IIa and NaPi-IIc proteins in the proximal tubule through the activation of ERα. The downregulation of NaPi-IIa by estrogen involves 3'-UTR of its mRNA and is independent of Klotho/fibroblast growth factor 23 and PTH signaling pathways.


Subject(s)
Estradiol/pharmacology , Estrogen Receptor alpha/physiology , Fibroblast Growth Factors/physiology , Glucuronidase/physiology , Kidney/metabolism , Parathyroid Hormone/physiology , Sodium-Phosphate Cotransporter Proteins, Type IIa/biosynthesis , 3' Untranslated Regions/genetics , 5' Untranslated Regions/genetics , Animals , Cell Line , Down-Regulation/genetics , Down-Regulation/physiology , Estrogen Receptor alpha/genetics , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Glucuronidase/genetics , Kidney/drug effects , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Klotho Proteins , Mice , Mice, Knockout , Parathyroid Hormone/genetics , Phosphates/blood , Phosphates/urine , Sodium-Phosphate Cotransporter Proteins, Type IIa/drug effects , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics
5.
Am J Physiol Renal Physiol ; 308(6): F522-34, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25608964

ABSTRACT

We have previously demonstrated that estrogen (E2) downregulates phosphate transporter NaPi-IIa and causes phosphaturia and hypophosphatemia in ovariectomized rats. In the present study, we examined whether E2 directly targets NaPi-IIa in the proximal tubule (PT) and studied the respective roles of estrogen receptor isoforms (ERα and ERß) in the downregulation of NaPi-IIa using both in vivo and an in vitro expression systems. We found that estrogen specifically downregulates NaPi-IIa but not NaPi-IIc or Pit2 in the kidney cortex. Proximal tubules incubated in a "shake" suspension with E2 for 24 h exhibited a dose-dependent decrease in NaPi-IIa protein abundance. Results from OVX rats treated with specific agonists for either ERα [4,4',4″;-(4-propyl-[1H]-pyrazole-1,3,5-triyl) trisphenol, PPT] or ERß [4,4',4″-(4-propyl-[1H]-pyrazole-1,3,5-triyl) trisphenol, DPN] or both (PPT + DPN), indicated that only the latter caused a sharp downregulation of NaPi-IIa, along with significant phosphaturia and hypophosphatemia. Lastly, heterologous expression studies demonstrated that estrogen downregulated NaPi-IIa only in U20S cells expressing both ERα and ERß, but not in cells expressing either receptor alone. In conclusion, these studies demonstrate that rat PT cells express both ERα and ERß and that E2 induces phosphaturia by directly and specifically targeting NaPi-IIa in the PT cells. This effect is mediated via a mechanism involving coactivation of both ERα and ERß, which likely form a functional heterodimer complex in the rat kidney proximal tubule.


Subject(s)
Estrogens/physiology , Kidney Tubules, Proximal/metabolism , Phosphates/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism , Animals , Cell Line , Chlorides/urine , Eating , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Female , Humans , Kidney Cortex/metabolism , Phosphate Transport Proteins/metabolism , Random Allocation , Rats, Sprague-Dawley
6.
Proc Natl Acad Sci U S A ; 109(33): 13368-73, 2012 Aug 14.
Article in English | MEDLINE | ID: mdl-22847418

ABSTRACT

The Na-Cl cotransporter (NCC), which is the target of inhibition by thiazides, is located in close proximity to the chloride-absorbing transporter pendrin in the kidney distal nephron. Single deletion of pendrin or NCC does not cause salt wasting or excessive diuresis under basal conditions, raising the possibility that these transporters are predominantly active during salt depletion or in response to excess aldosterone. We hypothesized that pendrin and NCC compensate for loss of function of the other under basal conditions, thereby masking the role that each plays in salt absorption. To test our hypothesis, we generated pendrin/NCC double knockout (KO) mice by crossing pendrin KO mice with NCC KO mice. Pendrin/NCC double KO mice displayed severe salt wasting and sharp increase in urine output under basal conditions. As a result, animals developed profound volume depletion, renal failure, and metabolic alkalosis without hypokalemia, which were all corrected with salt replacement. We propose that the combined inhibition of pendrin and NCC can provide a strong diuretic regimen without causing hypokalemia for patients with fluid overload, including patients with congestive heart failure, nephrotic syndrome, diuretic resistance, or generalized edema.


Subject(s)
Anion Transport Proteins/deficiency , Diuresis/physiology , Receptors, Drug/deficiency , Renal Insufficiency/physiopathology , Sodium Chloride/metabolism , Symporters/deficiency , Alkalosis/blood , Alkalosis/complications , Alkalosis/physiopathology , Animals , Anion Transport Proteins/metabolism , Body Weight , Kidney/pathology , Kidney/physiopathology , Kidney Function Tests , Mice , Mice, Knockout , Models, Biological , Potassium/blood , Receptors, Drug/metabolism , Renal Insufficiency/blood , Renal Insufficiency/complications , Sodium/blood , Sodium Chloride, Dietary , Solute Carrier Family 12, Member 3 , Sulfate Transporters , Symporters/metabolism , Vasopressins/administration & dosage , Vasopressins/pharmacology , Water-Electrolyte Balance/physiology
7.
Ocul Surf ; 29: 432-443, 2023 07.
Article in English | MEDLINE | ID: mdl-37355021

ABSTRACT

PURPOSE: Cystinosis is an autosomal recessive lysosomal storage disease (LSDs) caused by mutations in the gene encoding cystinosin (CTNS) that leads to cystine crystal accumulation in the lysosome that compromises cellular functions resulting in tissue damage and organ failure, especially in kidneys and eyes. However, the underlying molecular mechanism of its pathogenesis remains elusive. Two novel mice lines created via CRISPR are used to examine the pathogenesis of cystinosis in the kidney and cornea and the treatment efficacy of corneal pathology using self-complimentary Adeno-associated viral (scAAV-CTNS) vector. METHODS: The CRISPR technique generated two novel cystinotic mouse lines, Ctnsis1 (an insertional mutation) and Ctnsis2 (a nonsense mutation). Immune histochemistry, renal functions test and HRT2 in vivo confocal microscopy were used to evaluate the age-related renal pathogenesis and treatment efficacy of the scAAV-CTNS virus in corneal pathology. RESULTS: Both mutations lead to the production of truncated Ctns proteins. Ctnsis1 and Ctnsis 2 mice exhibit the characteristic of cystinotic corneal crystal phenotype at four-week-old. Treatment with the scAAV-CTNS viral vector decreased the corneal crystals in the treated mice cornea. Ctnsis 1 show renal abnormalities manifested by increased urine volume, reduced urine osmolality, and the loss of response to Desmopressin (dDAVP) at 22-month-old but Ctnsis2 don't manifest renal pathology up to 2 years of age. CONCLUSIONS: Both Ctnsis1 and Ctnsis2 mice exhibit phenotypes resembling human intermediate nephropathic and ocular cystinosis, respectively. scAAV-CTNS viral vectors reduce the corneal cystine crystals and have a great potential as a therapeutic strategy for treating patients suffering from cystinosis.


Subject(s)
Cystinosis , Humans , Animals , Mice , Infant , Cystinosis/therapy , Cystinosis/drug therapy , Cystine/genetics , Cystine/metabolism , Cystine/therapeutic use , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing , Cornea/pathology , Genetic Therapy
8.
Am J Physiol Gastrointest Liver Physiol ; 303(5): G546-60, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22723264

ABSTRACT

Activation of spermine/spermidine-N(1)-acetyltransferase (SSAT) leads to DNA damage and growth arrest in mammalian cells, and its ablation reduces the severity of ischemic and endotoxic injuries. Here we have examined the role of SSAT in the pathogenesis of toxic liver injury caused by carbon tetrachloride (CCl(4)). The expression and activity of SSAT increase in the liver subsequent to CCl(4) administration. Furthermore, the early liver injury after CCl(4) treatment was significantly attenuated in hepatocyte-specific SSAT knockout mice (Hep-SSAT-Cko) compared with wild-type (WT) mice as determined by the reduced serum alanine aminotransferase levels, decreased hepatic lipid peroxidation, and less severe liver damage. Cytochrome P450 2e1 levels remained comparable in both genotypes, suggesting that SSAT deficiency does not affect the metabolism of CCl(4). Hepatocyte-specific deficiency of SSAT also modulated the induction of cytokines involved in inflammation and repair as well as leukocyte infiltration. In addition, Noxa and activated caspase 3 levels were elevated in the livers of WT compared with Hep-SSAT-Cko mice. Interestingly, the onset of cell proliferation was significantly more robust in the WT compared with Hep-SSAT Cko mice. The inhibition of polyamine oxidases protected the animals against CCl(4)-induced liver injury. Our studies suggest that while the abrogation of polyamine back conversion or inhibition of polyamine oxidation attenuate the early injury, they may delay the onset of hepatic regeneration.


Subject(s)
Acetyltransferases/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Hepatocytes/metabolism , Liver/metabolism , Acetyltransferases/genetics , Animals , Carbon Tetrachloride , Liver/pathology , Mice , Mice, Knockout , Polyamines
9.
Nephrol Dial Transplant ; 27(4): 1368-79, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21873623

ABSTRACT

BACKGROUND: The epithelial calcium channel (ECaC) (TRPV5) and the Cl-/HCO3- exchanger pendrin (SLC26A4) are expressed on the apical membrane of tubular cells in the distal nephron and play essential roles in calcium re-absorption and bicarbonate secretion, respectively, in the kidney. METHODS: A combination of functional and molecular biology techniques were employed to examine the role of pendrin deletion in calcium excretion. RESULTS: Here, we demonstrate that deletion of pendrin causes acidic urine [urine pH 4.9 in knockout (KO) versus 5.9 in wild-type (WT) mice, P<0.03)] and downregulates the calcium-absorbing molecules ECaC and Na/Ca exchanger in the kidney, as shown by northern hybridization, immunoblot analysis and/or immunofluorescent labeling. These changes were associated with a ∼100% increase in 24-h urine calcium excretion in pendrin null mice. Subjecting the pendrin WT and KO mice to oral bicarbonate loading for 12 days increased the urine pH to ∼8 in both genotypes, normalized the expression of ECaC and Na/Ca exchanger and reduced the urine calcium excretion in pendrin-null mice to levels comparable to WT mice. CONCLUSIONS: We suggest that pendrin dysfunction should be suspected and investigated in humans with an otherwise unexplained acidic urine and hypercalciuria.


Subject(s)
Anion Transport Proteins/physiology , Calcium Channels/metabolism , Calcium/urine , Chloride-Bicarbonate Antiporters/metabolism , Kidney/metabolism , Sodium-Calcium Exchanger/metabolism , TRPV Cation Channels/metabolism , Absorption , Animals , Bicarbonates/metabolism , Blotting, Northern , Blotting, Western , Down-Regulation , Fluorescent Antibody Technique , Hydrogen-Ion Concentration , Kidney/cytology , Mice , Mice, Knockout , Sulfate Transporters
10.
Am J Physiol Renal Physiol ; 301(5): F969-78, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21795646

ABSTRACT

Hypokalemia is associated with increased ammoniagenesis and stimulation of net acid excretion by the kidney in both humans and experimental animals. The molecular mechanisms underlying these effects remain unknown. Toward this end, rats were placed in metabolic cages and fed a control or K(+)-deficient diet (KD) for up to 6 days. Rats subjected to KD showed normal acid-base status and serum electrolytes composition. Interestingly, urinary NH(4)(+) excretion increased significantly and correlated with a parallel decrease in urine K(+) excretion in KD vs. control animals. Molecular studies showed a specific upregulation of the glutamine transporter SN1, which correlated with the upregulation of glutaminase (GA), glutamate dehydrogenase (GDH), and phosphoenolpyruvate carboxykinase. These effects occurred as early as day 2 of KD. Rats subjected to a combined KD and 280 mM NH(4)Cl loading (to induce metabolic acidosis) for 2 days showed an additive increase in NH(4)(+) excretion along with an additive increment in the expression levels of ammoniagenic enzymes GA and GDH compared with KD or NH(4)Cl loading alone. The incubation of cultured proximal tubule cells NRK 52E or LLC-PK(1) in low-K(+) medium did not affect NH(4)(+) production and did not alter the expression of SN1, GA, or GDH in NRK cells. These results demonstrate that K(+) deprivation stimulates ammoniagenesis through a coordinated upregulation of glutamine transporter SN1 and ammoniagenesis enzymes. This effect is developed before the onset of hypokalemia. The signaling pathway mediating these events is likely independent of KD-induced intracellular acidosis. Finally, the correlation between increased NH(4)(+) production and decreased K(+) excretion indicate that NH(4)(+) synthesis and transport likely play an important role in renal K(+) conservation during hypokalemia.


Subject(s)
Ammonia/urine , Kidney/metabolism , Potassium Deficiency/metabolism , Acids , Amino Acid Transport Systems, Basic/metabolism , Ammonium Chloride/metabolism , Animals , Blotting, Northern , Chlorides/metabolism , Eating/physiology , Glutamate Dehydrogenase/metabolism , Glutaminase/metabolism , Glutathione Peroxidase/metabolism , Kidney Tubules/metabolism , LLC-PK1 Cells , Male , Membranes/metabolism , Potassium/metabolism , RNA/biosynthesis , RNA/isolation & purification , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Swine
11.
Cell Physiol Biochem ; 28(3): 521-6, 2011.
Article in English | MEDLINE | ID: mdl-22116366

ABSTRACT

The Cl(-)/HCO(3)(-) exchanger pendrin (SLC26A4, PDS) and the thiazide-sensitive NaCl cotransporter NCC (SLC12A3) are expressed on the apical membranes of distal nephron segments and mediate salt absorption, with pendrin working in tandem with the epithelial Na channel (ENaC) and NCC working by itself. Pendrin is expressed on the apical membrane of intercalated cells in late distal convoluted tubule (DCT), connecting tubule (CNT) and the cortical collecting duct (CCD) whereas the thiazide-sensitive NaCl cotransporter NCC is primarily detected on the apical membrane of DCT cells. Recent studies indicate that pendrin expression is increased in kidneys of NCC knockout mice, raising the possibility that pendrin and NCC can compensate for loss of the other by increasing their expression and activity. Current investigations in our laboratories demonstrate that pendrin plays an important role in compensatory salt absorption in response to the loop diuretics and the thiazide derivatives. These studies further demonstrate that whereas single deletion of pendrin or NCC does not cause salt wasting in mutant mice under baseline conditions, double knockout of pendrin and NCC causes profound polyuria and polydipsia, along with salt wasting under basal conditions. As a result, animals develop significant dehydration. We propose that pharmacologic inhibition of pendrin and NCC can provide a novel and strong diuretic regimen for patients with fluid overload, including those with congestive heart failure, nephrotic syndrome or renal failure.


Subject(s)
Anion Transport Proteins/physiology , Animals , Anion Transport Proteins/antagonists & inhibitors , Anion Transport Proteins/metabolism , Diuresis/physiology , Diuretics/pharmacology , Kidney Tubules, Distal/drug effects , Kidney Tubules, Distal/metabolism , Receptors, Drug/metabolism , Receptors, Drug/physiology , Solute Carrier Family 12, Member 3 , Sulfate Transporters , Symporters/metabolism , Symporters/physiology , Thiazides/pharmacology
12.
Am J Physiol Cell Physiol ; 299(1): C33-41, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20375274

ABSTRACT

The anion exchanger Pendrin, which is encoded by SLC26A4 (human)/Slc26a4 (mouse) gene, is localized on the apical membrane of non-acid-secreting intercalated (IC) cells in the kidney cortical collecting duct (CCD). To examine its role in the mediation of bicarbonate secretion in vivo and the apical Cl(-)/HCO(3)(-) exchanger in the kidney CCD, mice with genetic deletion of pendrin were generated. The mutant mice show the complete absence of pendrin expression in their kidneys as assessed by Northern blot hybridization, Western blot, and immunofluorescence labeling. Pendrin knockout (KO) mice display significantly acidic urine at baseline [pH 5.20 in KO vs. 6.01 in wild type (WT); P < 0.0001] along with elevated serum HCO(3)(-) concentration (27.4 vs. 24 meq/l in KO vs. WT, respectively; P < 0.02), consistent with decreased bicarbonate secretion in vivo. The urine chloride excretion was comparable in WT and KO mice. For functional studies, CCDs were microperfused and IC cells were identified by their ability to trap the pH fluorescent dye BCECF. The apical Cl(-)/HCO(3)(-) exchanger activity in B-IC and non-A, non-B-IC cells, as assessed by intracellular pH monitoring, was significantly reduced in pendrin-null mice. The basolateral Cl(-)/HCO(3)(-) exchanger activity in A-IC cells and in non-A, non-B-IC cells, was not different in pendrin KO mice relative to WT animals. Urine NH(4)(+) (ammonium) excretion increased significantly, consistent with increased trapping of NH(3) in the collecting duct in pendrin KO mice. We conclude that Slc26a4 (pendrin) deletion impairs the secretion of bicarbonate in vivo and reduces apical Cl(-)/HCO(3)(-) exchanger activity in B-IC and non-A, non-B-IC cells in CCD. Additional apical Cl(-)/HCO(3)(-) exchanger(s) is (are) present in the CCD.


Subject(s)
Anion Transport Proteins/deficiency , Bicarbonates/metabolism , Chloride-Bicarbonate Antiporters/metabolism , Kidney Tubules, Collecting/metabolism , Alkalosis/metabolism , Alkalosis/prevention & control , Animals , Anion Transport Proteins/genetics , Bicarbonates/blood , Chlorides/blood , Chlorides/urine , Down-Regulation , Hydrogen-Ion Concentration , Kidney Tubules, Collecting/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Quaternary Ammonium Compounds/urine , Sodium-Bicarbonate Symporters/metabolism , Sulfate Transporters , Time Factors
13.
Am J Physiol Cell Physiol ; 299(1): C164-74, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20392931

ABSTRACT

The expression of catabolic enzymes spermidine/spermine N(1)-acetyltransferase (SSAT) and spermine oxidase (SMO) increases after ischemic reperfusion injury. We hypothesized that polyamine catabolism is upregulated and that this increase in catabolic response contributes to tissue damage in endotoxin-induced acute kidney injury (AKI). SSAT mRNA expression peaked at threefold 24 h following LPS injection and returned to background levels by 48 h. The activity of SSAT correlated with its mRNA levels. The expression of SMO also increased in the kidney after LPS administration. Serum creatinine levels increased significantly at approximately 15 h, peaking by 24 h, and returned to background levels by 72 h. To test the role of SSAT in endotoxin-induced AKI, we injected wild-type (SSAT-wt) and SSAT-deficient (SSAT-ko) mice with LPS. Compared with SSAT-wt mice, the SSAT-ko mice subjected to endotoxic-AKI had less severe kidney damage as indicated by better preservation of kidney function. The role of polyamine oxidation in the mediation of kidney injury was examined by comparing the severity of renal damage in SSAT-wt mice treated with MDL72527, an inhibitor of both polyamine oxidase and SMO. Animals treated with MDL72527 showed significant protection against endotoxin-induced AKI. We conclude that increased polyamine catabolism through generation of by-products of polyamine oxidation contributes to kidney damage and that modulation of polyamine catabolism may be a viable approach for the treatment of endotoxin-induced AKI.


Subject(s)
Acetyltransferases/metabolism , Kidney Diseases/enzymology , Kidney/enzymology , Polyamines/metabolism , Acetyltransferases/deficiency , Acetyltransferases/genetics , Acute Disease , Animals , Creatinine/blood , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic , Kidney/drug effects , Kidney/pathology , Kidney Diseases/chemically induced , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Diseases/prevention & control , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidoreductases Acting on CH-NH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Putrescine/analogs & derivatives , Putrescine/pharmacology , RNA, Messenger/metabolism , Severity of Illness Index , Time Factors , Polyamine Oxidase
14.
J Biol Chem ; 284(43): 29470-9, 2009 Oct 23.
Article in English | MEDLINE | ID: mdl-19723628

ABSTRACT

SLC26A7 (human)/Slc26a7 (mouse) is a recently identified chloride-base exchanger and/or chloride transporter that is expressed on the basolateral membrane of acid-secreting cells in the renal outer medullary collecting duct (OMCD) and in gastric parietal cells. Here, we show that mice with genetic deletion of Slc26a7 expression develop distal renal tubular acidosis, as manifested by metabolic acidosis and alkaline urine pH. In the kidney, basolateral Cl(-)/HCO3(-) exchange activity in acid-secreting intercalated cells in the OMCD was significantly decreased in hypertonic medium (a normal milieu for the medulla) but was reduced only mildly in isotonic medium. Changing from a hypertonic to isotonic medium (relative hypotonicity) decreased the membrane abundance of Slc26a7 in kidney cells in vivo and in vitro. In the stomach, stimulated acid secretion was significantly impaired in isolated gastric mucosa and in the intact organ. We propose that SLC26A7 dysfunction should be investigated as a potential cause of unexplained distal renal tubular acidosis or decreased gastric acid secretion in humans.


Subject(s)
Acidosis, Renal Tubular/metabolism , Chloride-Bicarbonate Antiporters , Gastric Acid/metabolism , Gastric Mucosa/metabolism , Kidney Medulla/metabolism , Acidosis, Renal Tubular/genetics , Animals , Humans , Hydrogen-Ion Concentration , Mice , Mice, Knockout , Mice, Mutant Strains , Sulfate Transporters
15.
Am J Physiol Renal Physiol ; 297(3): F604-11, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19587148

ABSTRACT

Chronic metabolic acidosis (CMA) is associated with an inhibition of fluid reabsorption in the renal proximal tubule. The effects of CMA on paracellular transport across the renal epithelial tight junction (TJ) is unknown. Claudin-2 is a transmembrane TJ-associated protein which confers TJ paracellular permeability to Na(+). We examined the effects of CMA on the expression of TJ transport proteins using both in vivo and in vitro models of CMA. The results showed downregulation of claudin-2 mRNA and protein expression in the cortex of rats subjected to the NH(4)Cl loading model of CMA. Madin-Darby canine kidney (MDCK) and HK-2 cells are models of renal epithelial cells and express claudin-2 protein in their TJ. We examined the effects of acidic pH exposure on the expression of claudin-2 in MDCK and HK-2 renal epithelial cells. Exposure of MDCK cells to pH 6.96 medium caused a significant and reversible decrease in claudin-2 protein abundance. A dose-response analysis of acidic medium exposure of MDCK and HK-2 cells demonstrated a downregulation of claudin-2 protein. The downregulation effect of acidic pH is specific to claudin-2 expression as the expression of other TJ-associated proteins (i.e., claudin-1, -3, -4, and -7, occludin, and zonula occludens-1) remained unchanged compared with control pH (7.40). Collectively, these data demonstrate that CMA downregulates the expression of claudin-2 likely through a direct effect of acidic pH. Potential physiological significance of these changes is discussed.


Subject(s)
Acidosis/metabolism , Epithelial Cells/metabolism , Kidney/metabolism , Membrane Proteins/metabolism , Tight Junctions/metabolism , Animals , Cell Line , Chronic Disease , Claudins , Disease Models, Animal , Dogs , Down-Regulation , Electric Impedance , Hydrogen-Ion Concentration , Membrane Proteins/genetics , Permeability , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sodium/metabolism , Time Factors
16.
Kidney Int ; 74(4): 438-47, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18496516

ABSTRACT

Increased dietary fructose in rodents recapitulates many aspects of the Metabolic Syndrome with hypertension, insulin resistance and dyslipidemia. Here we show that fructose increased jejunal NaCl and water absorption which was significantly decreased in mice whose apical chloride/base exchanger Slc26a6 (PAT1, CFEX) was knocked out. Increased dietary fructose intake enhanced expression of this transporter as well as the fructose-absorbing transporter Slc2a5 (Glut5) in the small intestine of wild type mice. Fructose feeding decreased salt excretion by the kidney and resulted in hypertension, a response almost abolished in the knockout mice. In parallel studies, a chloride-free diet blocked fructose-induced hypertension in Sprague Dawley rats. Serum uric acid remained unchanged in animals on increased fructose intake with hypertension. We suggest that fructose-induced hypertension is likely caused by increased salt absorption by the intestine and kidney and the transporters Slc26a6 and Slc2a5 are essential in this process.


Subject(s)
Amino Acid Transport Systems, Neutral/metabolism , Amino Acid Transport Systems/metabolism , Chlorides/metabolism , Fructose/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Glucose Transporter Type 5/metabolism , Hypertension/chemically induced , Symporters/metabolism , Amino Acid Transport Systems/genetics , Amino Acid Transport Systems, Neutral/genetics , Animals , Diet , Female , Fructose/genetics , Fructose/pharmacology , Glucose Transport Proteins, Facilitative/genetics , Glucose Transporter Type 5/genetics , Male , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Symporters/genetics
17.
Am J Nephrol ; 28(2): 330-8, 2008.
Article in English | MEDLINE | ID: mdl-18046080

ABSTRACT

BACKGROUND/AIM: Slc26a6 (PAT1, CFEX) is a major chloride/base exchanger located on the apical membrane of the kidney proximal tubule. The purpose of the present study was to examine the effect of Slc26a6 deletion on the apical Na+/H+ exchanger 3 (NHE3) in the straight segment (S3) of the proximal tubule, which is the major site for the reabsorption of filtered chloride in the kidney. METHODS: The proximal tubule S3 segment was perfused and the intracellular pH and apical Na+/H+ exchanger activity and expression were measured. RESULTS: In the proximal tubule straight segments that were microperfused in vitro, baseline intracellular pH, measured by BCPCF-AM, was 7.10 +/- 0.02 in Slc26a6-/- and 7.33 +/- 0.02 in Slc26a6+/+ animals, a significant reduction in Slc26a6 mutant mice (p < 0.00001). The activity of the apical Na+/H+ exchanger was 0.49 +/- 0.02 pH units/min in Slc26a6+/+ and 0.26 +/- 0.03 pH units/min in Slc26a6-/- animals, a significant reduction in Slc26a6-/- mice (p < 0.0001). Formate-induced intracellular alkalinization, which is mediated via NHE3, was significantly blunted in Slc26a6-/- animals, with an alkalinization magnitude of 0.16 pH unit in Slc26a6-/- versus 0.37 in Slc26a6+/+ animals (p < 0.00001, n = 5 separate animals). Angiotensin II stimulation of NHE3 activity was intact in Slc26a6-/- animals. Buffering capacity was comparable in Slc26a6+/+ and Slc26a6-/- mice. Immunoblotting and immunofluorescent labeling demonstrated comparable NHE3 abundance and distribution in kidney proximal tubules of Slc26a6+/+ and Slc26a6-/- mice. CONCLUSION: In conclusion, Slc26a6 deletion downregulates the apical Na+/H+ exchanger activity in the straight segment of the proximal tubule. The absence of a significant renal sodium loss in Slc26a6-null mice, despite NHE3 downregulation in the in vitro perfused tubules, points to possible activation of signaling pathways that can stimulate the apical Na+/H+ exchanger in vivo.


Subject(s)
Antiporters/genetics , Antiporters/physiology , Down-Regulation , Gene Deletion , Kidney Tubules, Proximal/metabolism , Animals , Female , Hydrogen-Ion Concentration , Male , Mice , Mice, Transgenic , Microscopy, Fluorescence , Models, Biological , Perfusion , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism , Sulfate Transporters
18.
Cancer Res ; 66(7): 3706-14, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16585197

ABSTRACT

Normal breast tissue mainly expresses the neuropeptide Y (NPY) Y2 receptor whereas primary human breast carcinomas express the Y1 receptor (Y1R) subtype. We hypothesized that activation of estrogen signaling systems plays a role in the induction of Y1R. To investigate this possibility, we used estrogen receptor-positive (ER+) human breast carcinoma cell line, MCF-7, and examined the effect of estrogen on Y1R gene expression and its signaling pathways. Saturation binding studies revealed that MCF-7 cells express high-affinity NPY receptor. NPY inhibited forskolin-stimulated adenosine 3'5'-cyclic monophosphate (cAMP) accumulation and mobilized intracellular Ca(2+) in MCF-7 cells. Chronic estrogen treatment enhanced NPY-mediated inhibition of cAMP accumulation by 4-fold and caused a significant increase in Y1R mRNA expression through ERalpha. Similarly, estrogen increased Y1R mRNA expression in T-47D (ER+) but not in MDA-MB231 or MDA-MB468 (ER-) cell lines. Cycloheximide decreased basal Y1R mRNA expression; however, it did not affect its increase by estrogen. Moreover, estrogen treatment of MCF-7 cells did not increase Y1R mRNA stability. The up-regulation of Y1R expression by estrogen is prevented by hydroxyurea but not by nocodazole or IB-MECA (cell cycle inhibitors). Lastly, NPY inhibited estrogen-induced cell proliferation through Y1R. In conclusion, MCF-7 cells express a functional Y1R coupled to both Ca(2+) and cAMP pathways. Estrogen up-regulates Y1R expression through ERalpha. This effect is independent of increased Y1R mRNA stability or new protein synthesis, and likely occurs during S phase completion of the cell cycle. Estrogen plays an important role in the up-regulation of Y1R, which in turn regulates estrogen-induced cell proliferation in breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Estrogens/pharmacology , Receptors, Neuropeptide Y/biosynthesis , Binding, Competitive , Breast Neoplasms/genetics , Calcium/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Colforsin/pharmacology , Cyclic AMP/metabolism , Cycloheximide/pharmacology , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Iodine Radioisotopes , Neuropeptide Y/metabolism , Neuropeptide Y/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism , Up-Regulation/drug effects
19.
J Am Diet Assoc ; 107(8): 1404-8, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17659909

ABSTRACT

There is conflicting evidence regarding the effects of high protein intake on kidney health, especially as it relates to age. We investigated the short-term effects of a high-protein diet on kidney function and systemic acid-base homeostasis in older compared to younger adults. The subjects were healthy men and women either between the ages of 25 and 40 years (n=12) or 55 and 70 years (n=10). They underwent a two-period crossover trial with each period consisting of 2 weeks of usual diet followed by a 1-week experimental diet. During the experimental diet period subjects consumed metabolic meals that provided either low protein content (0.5 g protein/kg/day) or high protein content (2.0 g protein/kg/day). Outcome measures included blood and urine markers of renal function and acid-base balance. An analysis of variance was used to assess differences between age groups with respect to experimental diet. The older group, mainly women, showed an increase in glomerular filtration rate after the high-protein compared to low-protein diet; the younger group did not. Urinary pH was significantly lower, and ammonium excretion was significantly higher after the high-protein diet in both age groups, but neither group developed a clinically detectable acidosis after the week of receiving a high-protein diet.


Subject(s)
Acid-Base Equilibrium/drug effects , Aging/physiology , Dietary Proteins/administration & dosage , Glomerular Filtration Rate/drug effects , Urine/chemistry , Adult , Age Factors , Aged , Analysis of Variance , Biomarkers/blood , Biomarkers/urine , Blood Chemical Analysis , Cross-Over Studies , Dietary Proteins/adverse effects , Dose-Response Relationship, Drug , Female , Humans , Hydrogen-Ion Concentration , Kidney Function Tests , Male , Middle Aged , Quaternary Ammonium Compounds/urine , Sex Factors
20.
Sci Transl Med ; 7(313): 313ra181, 2015 Nov 11.
Article in English | MEDLINE | ID: mdl-26560359

ABSTRACT

Pulmonary alveolar microlithiasis (PAM) is a rare, autosomal recessive lung disorder associated with progressive accumulation of calcium phosphate microliths. Inactivating mutations in SLC34A2, which encodes the NPT2b sodium-dependent phosphate cotransporter, has been proposed as a cause of PAM. We show that epithelial deletion of Npt2b in mice results in a progressive pulmonary process characterized by diffuse alveolar microlith accumulation, radiographic opacification, restrictive physiology, inflammation, fibrosis, and an unexpected alveolar phospholipidosis. Cytokine and surfactant protein elevations in the alveolar lavage and serum of PAM mice and confirmed in serum from PAM patients identify serum MCP-1 (monocyte chemotactic protein 1) and SP-D (surfactant protein D) as potential biomarkers. Microliths introduced by adoptive transfer into the lungs of wild-type mice produce marked macrophage-rich inflammation and elevation of serum MCP-1 that peaks at 1 week and resolves at 1 month, concomitant with clearance of stones. Microliths isolated by bronchoalveolar lavage readily dissolve in EDTA, and therapeutic whole-lung EDTA lavage reduces the burden of stones in the lungs. A low-phosphate diet prevents microlith formation in young animals and reduces lung injury on the basis of reduction in serum SP-D. The burden of pulmonary calcium deposits in established PAM is also diminished within 4 weeks by a low-phosphate diet challenge. These data support a causative role for Npt2b in the pathogenesis of PAM and the use of the PAM mouse model as a preclinical platform for the development of biomarkers and therapeutic strategies.


Subject(s)
Biomarkers/blood , Calcinosis/etiology , Calcinosis/physiopathology , Calcinosis/therapy , Genetic Diseases, Inborn/etiology , Genetic Diseases, Inborn/physiopathology , Genetic Diseases, Inborn/therapy , Lung Diseases/etiology , Lung Diseases/physiopathology , Lung Diseases/therapy , Sodium-Phosphate Cotransporter Proteins, Type IIb/deficiency , Sodium-Phosphate Cotransporter Proteins, Type IIb/genetics , Animals , Diet , Disease Models, Animal , Epithelium/metabolism , Epithelium/pathology , Lung/metabolism , Lung/pathology , Mice , Mutation , Phosphates/metabolism , Pulmonary Alveoli/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL