Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Physiol Renal Physiol ; 315(2): F386-F394, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29412698

ABSTRACT

The sodium-glucose cotransporter SGLT2 inhibitor empagliflozin (plasma protein binding ~88%) may reach its target in the brush border of the early proximal tubule by glomerular filtration and tubular secretion. Here we determined whether empagliflozin is secreted by renal tubules in mice and whether genetic knockout of the basolateral organic anion transporter 3 ( Oat3-/-) affects its tubular secretion or glucosuric effect. Renal clearance studies in wild-type (WT) mice showed that tubular secretion accounted for 50-70% of empagliflozin urinary excretion. Immunostaining indicated that SGLT2 and OAT3 localization partially overlapped in proximal tubule S1 and S2 segments. Glucosuria in metabolic cage studies was reduced in Oat3-/- vs. WT mice for acute empagliflozin doses of 1, 3, and 10 mg/kg, whereas 30 mg/kg induced similar maximal glucosuria in both genotypes. Chronic application of empagliflozin (~25 mg·kg-1 ·day-1) in Oat3-/- mice was associated with lower urinary glucose-to-creatinine ratios despite maintaining slightly higher blood glucose levels than WT. On a whole kidney level, renal secretion of empagliflozin was largely unchanged in Oat3-/- mice. However, the absence of OAT3 attenuated the influence of empagliflozin on fractional glucose excretion; higher levels of plasma or filtered empagliflozin were needed to induce similar increases in fractional renal glucose excretion. We conclude that empagliflozin is excreted into the urine to similar extent by glomerular filtration and tubular secretion. The latter can occur largely independent of OAT3. However, OAT3 increases the glucosuric effect of empagliflozin, which may relate to the partial overlap of its localization with SGLT2 and thus OAT3-mediated tubular secretion of empagliflozin in the early proximal tubule.


Subject(s)
Benzhydryl Compounds/pharmacology , Blood Glucose/drug effects , Glucosides/pharmacology , Glycosuria/metabolism , Kidney Tubules, Proximal/drug effects , Organic Anion Transporters, Sodium-Independent/metabolism , Renal Elimination , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2/drug effects , Animals , Benzhydryl Compounds/pharmacokinetics , Benzhydryl Compounds/urine , Blood Glucose/metabolism , Glomerular Filtration Rate , Glucosides/pharmacokinetics , Glucosides/urine , Glycosuria/genetics , Glycosuria/prevention & control , Kidney Tubules, Proximal/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Organic Anion Transport Protein 1/genetics , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Independent/deficiency , Organic Anion Transporters, Sodium-Independent/genetics , Sodium-Glucose Transporter 2/metabolism , Sodium-Glucose Transporter 2 Inhibitors/pharmacokinetics , Sodium-Glucose Transporter 2 Inhibitors/urine
2.
Pflugers Arch ; 469(12): 1545-1565, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28842746

ABSTRACT

The expression and localization of sodium-D-glucose cotransporter SGLT1 (SLC5A1), which is involved in small intestinal glucose absorption and renal glucose reabsorption, is of high biomedical relevance because SGLT1 inhibitors are currently tested for antidiabetic therapy. In human and rat organs, detailed expression profiling of SGLT1/Sglt1 mRNA and immunolocalization of the transporter protein has been performed. Using polyspecific antibodies and preabsorption with antigenic peptide as specificity control, in several organs, different immunolocalizations of SGLT1/Sglt1 between human and rat were obtained. Because the preabsorption control does not exclude cross-reactivity with similar epitopes, some localizations remained ambiguous. In the present study, we performed an immunocytochemical localization of Sglt1 in various organs of mice. Specificities of the immunoreactions were evaluated using antibody preabsorption with the Sglt1 peptide and the respective organs of Sglt1 knockout mice. Because staining in some locations was abolished after antibody preabsorption but remained in the knockout mice, missing staining in knockout mice was used as specificity criterion. The immunolocalization in mouse was identical or similar to rat in many organs, including small intestine, liver, and kidney. However, the male-dominant renal Sglt1 protein expression in mice differed from the female-dominant expression in rats, and localization in lung, heart, and brain observed in rats was not detected in mice. In mice, several novel locations of Sglt1, e.g., in eyes, tongue epithelial cells, pancreatic ducts, prostate, and periurethral glands were detected. Using end-point and quantitative RT-PCR in various organs, different Sglt1 expression in mice and rats was confirmed.


Subject(s)
Sodium-Glucose Transporter 1/analysis , Sodium-Glucose Transporter 1/biosynthesis , Animals , Female , Gene Expression Profiling , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats
3.
Cell Physiol Biochem ; 42(4): 1358-1365, 2017.
Article in English | MEDLINE | ID: mdl-28704812

ABSTRACT

BACKGROUND: Cellular glucose uptake may involve either non-concentrative glucose carriers of the GLUT family or Na+-coupled glucose-carrier SGLT1, which accumulates glucose against glucose gradients and may thus accomplish cellular glucose uptake even at dramatically decreased extracellular glucose concentrations. SGLT1 is not only expressed in epithelia but as well in tumour cells and immune cells. Immune cell functions strongly depend on their metabolism, therefore we hypothesized that deficiency of SGLT1 modulates the defence against bacterial infection. To test this hypothesis, we infected wild type mice and gene targeted mice lacking functional SGLT1 with Listeria monocytogenes. METHODS: SGLT1 deficient mice and wild type littermates were infected with 1x104 CFU Listeria monocytogenes intravenously. Bacterial titers were determined by colony forming assay, SGLT1, TNF-α, IL-6 and IL-12a transcript levels were determined by qRT-PCR, as well as SGLT1 protein abundance and localization by immunohistochemistry. RESULTS: Genetic knockout of SGLT1 (Slc5a1-/- mice) significantly compromised bacterial clearance following Listeria monocytogenes infection with significantly enhanced bacterial load in liver, spleen, kidney and lung, and significantly augmented hepatic expression of TNF-α and IL-12a. While all wild type mice survived, all SGLT1 deficient mice died from the infection. CONCLUSIONS: SGLT1 is required for bacterial clearance and host survival following murine Listeria infection.


Subject(s)
Gene Expression/immunology , Glucose/immunology , Listeriosis/genetics , Listeriosis/mortality , Liver/immunology , Sodium-Glucose Transporter 1/immunology , Animals , Bacterial Load , Biological Transport , Glucose/metabolism , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Kidney/immunology , Kidney/microbiology , Kidney/pathology , Listeria monocytogenes , Listeriosis/immunology , Listeriosis/microbiology , Liver/microbiology , Liver/pathology , Lung/immunology , Lung/microbiology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/immunology , Sodium-Glucose Transporter 1/deficiency , Sodium-Glucose Transporter 1/genetics , Spleen/immunology , Spleen/microbiology , Spleen/pathology , Survival Analysis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
4.
Am J Physiol Renal Physiol ; 311(1): F227-38, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27053689

ABSTRACT

The initial step in renal secretion of organic anions (OAs) is mediated by transporters in the basolateral membrane (BLM). Contributors to this process are primary active Na(+)-K(+)-ATPase (EC 3.6.3.9), secondary active Na(+)-dicarboxylate cotransporter 3 (NaDC3/SLC13A3), and tertiary active OA transporters (OATs) OAT1/SLC22A6, OAT2/SLC22A7, and OAT3/SLC22A8. In human kidneys, we analyzed the localization of these transporters by immunochemical methods in tissue cryosections and isolated membranes. The specificity of antibodies was validated with human embryonic kidney-293 cells stably transfected with functional OATs. Na(+)-K(+)-ATPase was immunolocalized to the BLM along the entire human nephron. NaDC3-related immunostaining was detected in the BLM of proximal tubules and in the BLM and/or luminal membrane of principal cells in connecting segments and collecting ducts. The thin and thick ascending limbs, macula densa, and distal tubules exhibited no reactivity with the anti-NaDC3 antibody. OAT1-OAT3-related immunostaining in human kidneys was detected only in the BLM of cortical proximal tubules; all three OATs were stained more intensely in S1/S2 segments compared with S3 segment in medullary rays, whereas the S3 segment in the outer stripe remained unstained. Expression of NaDC3, OAT1, OAT2, and OAT3 proteins exhibited considerable interindividual variability in both male and female kidneys, and sex differences in their expression could not be detected. Our experiments provide a side-by-side comparison of basolateral transporters cooperating in renal OA secretion in the human kidney.


Subject(s)
Dicarboxylic Acid Transporters/metabolism , Nephrons/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Organic Anion Transporters/metabolism , Symporters/metabolism , Adult , Female , HEK293 Cells , Humans , Kidney Medulla/metabolism , Kidney Tubules, Collecting/metabolism , Kidney Tubules, Distal/metabolism , Male , Membranes/metabolism , Middle Aged , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Sex Characteristics , Sodium-Potassium-Exchanging ATPase/metabolism
5.
Cell Physiol Biochem ; 39(3): 1209-28, 2016.
Article in English | MEDLINE | ID: mdl-27595398

ABSTRACT

BACKGROUND: Similar to tumor cells, activated T-lymphocytes generate ATP mainly by glycolytic degradation of glucose. Lymphocyte glucose uptake involves non-concentrative glucose carriers of the GLUT family. In contrast to GLUT isoforms, Na+-coupled glucose-carrier SGLT1 accumulates glucose against glucose gradients and is effective at low extracellular glucose concentrations. The present study explored expression and regulation of SGLT1 in activated murine splenic cytotoxic T cells (CTLs) and human Jurkat T cells. METHODS: FACS analysis, immunofluorescence, confocal microscopy, chemiluminescence and Western blotting were employed to estimate SGLT1 expression, function and regulation in lymphocytes, as well as dual electrode voltage clamp in SGLT1 ± JAK3 expressing Xenopus oocytes to quantify the effect of janus kinase3 (JAK3) on SGLT1 function. RESULTS: SGLT1 is expressed in murine CTLs and also in human Jurkat T cells. 2-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-2-deoxyglucose uptake was significantly decreased by SGLT1-blocker phloridzin (0.2 mM) and by pharmacological inhibition of JAK3 with WHI-P131 (156 µM), WHI-P154 (11.2 µM) and JAK3 inhibitor VI (0.5 µM). Electrogenic glucose transport (Iglucose) in Xenopus oocytes expressing human SGLT1 was increased by additional expression of human wild type JAK3, active A568VJAK3 but not inactive K851AJAK3. Coexpression of JAK3 enhanced the maximal transport rate without significantly modifying affinity of the carrier. Iglucose in SGLT1+JAK3 expressing oocytes was significantly decreased by WHI-P154 (11.2 µM). JAK3 increased the SGLT1 protein abundance in the cell membrane. Inhibition of carrier insertion by brefeldin A (5 µM) in SGLT1+JAK3 expressing oocytes resulted in a decline of Iglucose, which was similar in presence and absence of JAK3. CONCLUSIONS: SGLT1 is expressed in murine cytotoxic T cells and human Jurkat T cells and significantly contributes to glucose uptake in those cells post activation. JAK3 up-regulates SGLT1 activity by increasing the carrier protein abundance in the cell membrane, an effect enforcing cellular glucose uptake into activated lymphocytes and thus contributing to the immune response.


Subject(s)
Glucose/immunology , Janus Kinase 3/genetics , Oocytes/metabolism , Sodium-Glucose Transporter 1/genetics , T-Lymphocytes, Cytotoxic/immunology , 4-Chloro-7-nitrobenzofurazan/analogs & derivatives , 4-Chloro-7-nitrobenzofurazan/pharmacology , Animals , Biological Transport , Brefeldin A/pharmacology , Caco-2 Cells , Deoxyglucose/analogs & derivatives , Deoxyglucose/pharmacology , Gene Expression Regulation , Glucose/pharmacology , Humans , Janus Kinase 3/immunology , Jurkat Cells , Lymphocyte Activation , Mice , Oocytes/cytology , Oocytes/drug effects , Patch-Clamp Techniques , Phlorhizin/pharmacology , Primary Cell Culture , Quinazolines/pharmacology , Signal Transduction , Sodium-Glucose Transporter 1/immunology , Spleen/cytology , Spleen/drug effects , Spleen/immunology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/drug effects , Transgenes , Xenopus laevis
6.
Arch Toxicol ; 90(3): 525-41, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25588984

ABSTRACT

Cadmium (Cd)-intoxicated experimental animals exhibit impaired renal secretion of organic anions (OA) and cations (OC), indicating their transporters (Oats and Octs) in the proximal tubule (PT) basolateral membrane as possible targets of Cd. To correlate transport data from the literature with the expression of relevant transporters, we performed immunochemical and RT-PCR studies of renal Oats and Octs in the subchronic (treatment with CdCl2; 2 mg Cd/kg b.m./day, for 2 weeks) and acute (treatment with Cd-metallothionein (CdMT); 0.4 mg Cd/kg b.m., 6 or 12 h before killing) models of Cd nephrotoxicity. In the subchronic model, PT exhibited a minor loss of basolateral invaginations and overall unchanged expression of Na(+)/K(+)-ATPase and GAPDH proteins and mRNAs, while the expression of Oat and Oct proteins and their mRNAs was strongly downregulated. In the acute model, a time-related redistribution of basolateral transporters to the intracellular vesicular compartment was a major finding. However, 6 h following CdMT treatment, the total abundance of Oat and Oct proteins in the renal tissue remained unchanged, the expression of mRNAs decreased only for Oats, while a limited Oat1 and Na(+)/K(+)-ATPase immunoreactivity in the PT apical membrane indicated loss of cell polarity. As tested in rats treated with colchicine, the observed loss/redistribution of basolateral transporters in both models may be independent on microtubules. Therefore, the diminished renal secretion of OA and OC via PT in Cd nephrotoxicity may result from (a) limited loss of secretory surface (basolateral invaginations), (b) selective loss of Oats and Octs, and


Subject(s)
Cadmium/toxicity , Kidney Diseases/chemically induced , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/metabolism , Animals , Catecholamine Plasma Membrane Transport Proteins/genetics , Catecholamine Plasma Membrane Transport Proteins/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Kidney Tubules, Proximal/metabolism , Male , Metallothionein/toxicity , Organic Anion Transport Protein 1/genetics , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters/genetics , Organic Cation Transport Proteins/genetics , Rats, Wistar , Sodium-Potassium-Exchanging ATPase/metabolism , Toxicity Tests, Acute , Toxicity Tests, Subchronic
7.
Pflugers Arch ; 467(9): 1881-98, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25304002

ABSTRACT

Novel affinity-purified antibodies against human SGLT1 (hSGLT1) and SGLT2 (hSGLT2) were used to localize hSGLT2 in human kidney and hSGLT1 in human kidney, small intestine, liver, lung, and heart. The renal locations of both transporters largely resembled those in rats and mice; hSGLT2 and SGLT1 were localized to the brush border membrane (BBM) of proximal tubule S1/S2 and S3 segments, respectively. Different to rodents, the renal expression of hSGLT1 was absent in thick ascending limb of Henle (TALH) and macula densa, and the expression of both hSGLTs was sex-independent. In small intestinal enterocytes, hSGLT1 was localized to the BBM and subapical vesicles. Performing double labeling with glucagon-like peptide 1 (GLP-1) or glucose-dependent insulinotropic peptide (GIP), hSGLT1 was localized to GLP-1-secreting L cells and GIP-secreting K cells as has been shown in mice. In liver, hSGLT1 was localized to biliary duct cells as has been shown in rats. In lung, hSGLT1 was localized to alveolar epithelial type 2 cells and to bronchiolar Clara cells. Expression of hSGLT1 in Clara cells was verified by double labeling with the Clara cell secretory protein CC10. Double labeling of human heart with aquaporin 1 immunolocalized the hSGLT1 protein in heart capillaries rather than in previously assumed myocyte sarcolemma. The newly identified locations of hSGLT1 implicate several extra renal functions of this transporter, such as fluid absorption in the lung, energy supply to Clara cells, regulation of enteroendocrine cells secretion, and release of glucose from heart capillaries. These functions may be blocked by reversible SGLT1 inhibitors which are under development.


Subject(s)
Sodium-Glucose Transporter 1/biosynthesis , Sodium-Glucose Transporter 2/biosynthesis , Adult , Blotting, Western , Female , Heart , Humans , Immunohistochemistry , Intestine, Small/metabolism , Kidney/metabolism , Liver/metabolism , Lung/metabolism , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Sodium-Glucose Transporter 1/analysis , Sodium-Glucose Transporter 2/analysis
8.
Am J Physiol Renal Physiol ; 308(8): F809-21, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25656365

ABSTRACT

In the mammalian kidney, nonglycosylated and glycosylated forms of aquaporin protein 1 (AQP1) coexist in the luminal and basolateral plasma membranes of proximal tubule and descending thin limb. Factors that influence AQP1 expression in (patho)physiological conditions are poorly known. Thus far, only angiotensin II and hypertonicity were found to upregulate AQP1 expression in rat proximal tubule in vivo and in vitro (Bouley R, Palomino Z, Tang SS, Nunes P, Kobori H, Lu HA, Shum WW, Sabolic I, Brown D, Ingelfinger JR, Jung FF. Am J Physiol Renal Physiol 297: F1575-F1586, 2009), a phenomenon that may be relevant for higher blood pressure observed in men and male experimental animals. Here we investigated the sex-dependent AQP1 protein and mRNA expression in the rat kidney by immunochemical methods and qRT-PCR in tissue samples from prepubertal and intact gonadectomized animals and sex hormone-treated gonadectomized adult male and female animals. In adult rats, the overall renal AQP1 protein and mRNA expression was ∼80% and ∼40% higher, respectively, in males than in females, downregulated by gonadectomy in both sexes and upregulated strongly by testosterone and moderately by progesterone treatment; estradiol treatment had no effect. In prepubertal rats, the AQP1 protein expression was low compared with adults and slightly higher in females, whereas the AQP1 mRNA expression was low and similar in both sexes. The observed differences in AQP1 protein expression in various experiments mainly reflect changes in the glycosylated form. The male-dominant expression of renal AQP1 in rats, which develops after puberty largely in the glycosylated form of the protein, may contribute to enhanced fluid reabsorption following the androgen- or progesterone-stimulated activities of sodium-reabsorptive mechanisms in proximal tubules.


Subject(s)
Aquaporin 1/metabolism , Nephrons/metabolism , Age Factors , Animals , Aquaporin 1/drug effects , Aquaporin 1/genetics , Estradiol/administration & dosage , Estrogen Replacement Therapy , Female , Gene Expression Regulation , Glomerular Filtration Rate , Glycosylation , Male , Nephrons/drug effects , Orchiectomy , Osmolar Concentration , Ovariectomy , Progesterone/administration & dosage , RNA, Messenger/metabolism , Rats, Wistar , Renal Elimination , Sex Factors , Sexual Maturation , Testosterone/administration & dosage , Urodynamics
9.
Biometals ; 28(4): 701-12, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25947387

ABSTRACT

Detoxification of mercury (Hg) with selenium (Se) in the early postnatal period with regard to the expression of metallothionein protein (MT), essential element status, and lipid peroxidation level in tissues has not been studied. Seven-day-old Wistar pups were orally pretreated with Se [6 µmol Na2SeO3/kg body weight (b.w.)] for 3 days and then cotreated with Hg (6 µmol HgCl2/kg b.w.) for the following 4 days. This group (Se + Hg) was compared to the groups treated with Hg, Se, or vehicle (control). Compared to the Hg-group, Se + Hg-group exhibited lower renal MT expression, reduced accumulation of Hg, Cu and Zn, and reduced excretion of Se, Hg and Zn in urine. In the liver, MT was stimulated by Se treatment in both, Se and Se + Hg-group. Hepatic and brain levels of the endogenous essential elements Cu, Fe, Mg, and Zn remained unchanged in all of the studied groups. Brain Hg levels and oxidation of lipids measured as thiobarbituric acid reactive substances were diminished in Se + Hg-group of pups compared to the Hg-group. This study suggests that Se pretreatment can help reduce Hg in the tissues of suckling rats, simultaneously preventing impairment of essential element levels in the kidneys and their excessive excretion via urine. Also, Se was shown to prevent oxidative damage of lipids in the brain, which is particularly susceptible to Hg during the early postnatal period.


Subject(s)
Lipid Peroxidation/drug effects , Mercury/metabolism , Metallothionein/metabolism , Selenium/pharmacology , Administration, Oral , Animals , Animals, Suckling , Female , Kidney/metabolism , Male , Mercury/administration & dosage , Mercury/toxicity , Rats , Rats, Wistar , Selenium/administration & dosage , Selenium/metabolism , Tissue Distribution
10.
Croat Med J ; 56(5): 447-59, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26526882

ABSTRACT

AIM: To investigate whether the sex-dependent expression of hepatic and renal oxalate transporter sat-1 (Slc26a1) changes in a rat model of ethylene glycol (EG)-induced hyperoxaluria. METHODS: Rats were given tap water (12 males and 12 females; controls) or EG (12 males and 12 females; 0.75% v/v in tap water) for one month. Oxaluric state was confirmed by biochemical parameters in blood plasma, urine, and tissues. Expression of sat-1 and rate-limiting enzymes of oxalate synthesis, alcohol dehydrogenase 1 (Adh1) and hydroxy-acid oxidase 1 (Hao1), was determined by immunocytochemistry (protein) and/or real time reverse transcription polymerase chain reaction (mRNA). RESULTS: EG-treated males had significantly higher (in µmol/L; mean±standard deviation) plasma (59.7±27.2 vs 12.9±4.1, P<0.001) and urine (3716±1726 vs 241±204, P<0.001) oxalate levels, and more abundant oxalate crystaluria than controls, while the liver and kidney sat-1 protein and mRNA expression did not differ significantly between these groups. EG-treated females, in comparison with controls had significantly higher (in µmol/L) serum oxalate levels (18.8±2.9 vs 11.6±4.9, P<0.001), unchanged urine oxalate levels, low oxalate crystaluria, and significantly higher expression (in relative fluorescence units) of the liver (1.59±0.61 vs 0.56±0.39, P=0.006) and kidney (1.77±0.42 vs 0.69±0.27, P<0.001) sat-1 protein, but not mRNA. The mRNA expression of Adh1 was female-dominant and that of Hao1 male-dominant, but both were unaffected by EG treatment. CONCLUSIONS: An increased expression of hepatic and renal oxalate transporting protein sat-1 in EG-treated female rats could protect from hyperoxaluria and oxalate urolithiasis.


Subject(s)
Anion Transport Proteins/metabolism , Antiporters/metabolism , Ethylene Glycol/therapeutic use , Hyperoxaluria/prevention & control , Kidney/drug effects , Liver/drug effects , Alcohol Dehydrogenase/genetics , Alcohol Dehydrogenase/metabolism , Animals , Anion Transport Proteins/genetics , Antiporters/genetics , Blotting, Western , Calcium Oxalate/blood , Calcium Oxalate/urine , Chromatography, High Pressure Liquid , Female , Hyperoxaluria/metabolism , Kidney/metabolism , Liver/metabolism , Male , RNA, Messenger/metabolism , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Sex Factors , Sulfate Transporters
11.
Am J Physiol Renal Physiol ; 304(8): F1114-26, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23389457

ABSTRACT

In the mouse kidney, organic anion transporter 3 (mOat3, Slc22a8) was previously localized to the basolateral membrane (BLM) of proximal tubule (PT), thick ascending limb of Henle, macula densa, distal tubule, and cortical collecting duct. However, the specificity of anti-Oat3 antibodies (Oat3-Ab) used in these studies was not properly verified. Moreover, the sex-dependent expression of mOat3, and of the functionally similar transporter mOat1 (Slc22a6), in the mouse kidney has been studied at mRNA level, whereas their protein expression is poorly documented. Here we investigated 1) specificity of Oat3-Abs by using Oat3 knockout (KO) mice, 2) cell localization of renal mOat3 with a specific mOat3-Ab, 3) sex-dependent expression of renal mOat3 and mOat1 proteins, and 4) hormone(s) responsible for observed sex differences. As previously shown, an Oat3-Ab against the rat protein stained the BLM of various nephron segments in wild-type (WT) mice, but the same staining pattern was noted along the nephron of Oat3 KO mice. However, the mOat3-Ab exclusively stained the BLM of PT in WT mice, where it colocalized with the mOat1 protein, whereas no staining of Oat3 protein was noted in the kidney of Oat3 KO mice. The expression of mOat3 protein was lower in male mice, upregulated by castration, and downregulated by testosterone treatment. The expression of mOat1 protein was stronger in males, downregulated by castration, and upregulated by testosterone treatment. Thus, at the protein level, mOat3 and mOat1 exhibit sex-dependent expression with an opposite pattern; mOat3 is female dominant due to androgen inhibition, while mOat1 is male dominant due to androgen stimulation.


Subject(s)
Nephrons/physiology , Organic Anion Transport Protein 1/genetics , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Sex Characteristics , Androgens/metabolism , Androgens/pharmacology , Animals , Blotting, Western , Down-Regulation/physiology , Female , Immunohistochemistry , Kidney/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orchiectomy
12.
Mol Pharm ; 10(6): 2370-80, 2013 Jun 03.
Article in English | MEDLINE | ID: mdl-23607617

ABSTRACT

Kidney transplanted patients are often treated with immunosuppressive, antihypertensive, and antibiotic drugs such as cyclosporine A (CsA), ß-blockers, and fluoroquinolones, respectively. Organic cation transporters (OCT) expressed in the basolateral membrane of proximal tubules represent an important drug excretion route. In this work, the renal expression of OCT after syngeneic and allogeneic kidney transplantation in rats with or without CsA immunosuppression was studied. Moreover, the interactions of CsA, ß-blockers (pindolol/atenolol), and fluoroquinolones (ofloxacin/norfloxacin) with rOCT1, rOCT2, hOCT1, and hOCT2 in stably transfected HEK293-cells were studied. Kidney transplantation was associated with reduced expression of rOCT1, while rOCT2 showed only reduced expression after allogeneic transplantation. All drugs interacted subtype- and species-dependently with OCT. However, only atenolol, pindolol, and ofloxacin were transported by hOCT2, the main OCT in human kidneys. While CsA is not an OCT substrate, it exerts a short-term effect on OCT activity, changing their affinity for some substrates. In conclusion, appropriate drug dosing in transplanted patients is difficult partly because OCT are down-regulated and because concomitant CsA treatment may influence the affinity of the transporters. Moreover, drug-drug competition at the transporter can also alter drug excretion rate.


Subject(s)
Adrenergic beta-Antagonists/metabolism , Fluoroquinolones/metabolism , Kidney Transplantation/adverse effects , Kidney Tubules, Proximal/metabolism , Organic Cation Transport Proteins/metabolism , Animals , Blotting, Western , Cell Line , Cyclosporine/therapeutic use , Humans , Immunohistochemistry , In Vitro Techniques , Kidney Tubules, Proximal/drug effects , Male , Organic Cation Transport Proteins/genetics , Organic Cation Transporter 1/genetics , Organic Cation Transporter 1/metabolism , Organic Cation Transporter 2 , Rats , Real-Time Polymerase Chain Reaction
13.
Am J Physiol Cell Physiol ; 302(8): C1174-88, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22262063

ABSTRACT

With a novel antibody against the rat Na(+)-D-glucose cotransporter SGLT2 (rSGLT2-Ab), which does not cross-react with rSGLT1 or rSGLT3, the ∼75-kDa rSGLT2 protein was localized to the brush-border membrane (BBM) of the renal proximal tubule S1 and S2 segments (S1 > S2) with female-dominant expression in adult rats, whereas rSglt2 mRNA expression was similar in both sexes. Castration of adult males increased the abundance of rSGLT2 protein; this increase was further enhanced by estradiol and prevented by testosterone treatment. In the renal BBM vesicles, the rSGLT1-independent uptake of [(14)C]-α-methyl-D-glucopyranoside was similar in females and males, suggesting functional contribution of another Na(+)-D-glucose cotransporter to glucose reabsorption. Since immunoreactivity of rSGLT2-Ab could not be detected with certainty in rat extrarenal organs, the SGLT2 protein was immunocharacterized with the same antibody in wild-type (WT) mice, with SGLT2-deficient (Sglt2 knockout) mice as negative control. In WT mice, renal localization of mSGLT2 protein was similar to that in rats, whereas in extrarenal organs neither mSGLT2 protein nor mSglt2 mRNA expression was detected. At variance to the findings in rats, the abundance of mSGLT2 protein in the mouse kidneys was male dominant, whereas the expression of mSglt2 mRNA was female dominant. Our results indicate that in rodents the expression of SGLT2 is kidney-specific and point to distinct sex and species differences in SGLT2 protein expression that cannot be explained by differences in mRNA.


Subject(s)
Sodium-Glucose Transporter 2/biosynthesis , Symporters/biosynthesis , Animals , Castration/methods , Estradiol/pharmacology , Female , Galactose/metabolism , Glucose/metabolism , Kidney/drug effects , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Microvilli/drug effects , Microvilli/metabolism , RNA, Messenger/genetics , Rats , Rats, Wistar , Sex Factors , Sodium-Glucose Transporter 2/genetics , Sodium-Glucose Transporter 2/metabolism , Symporters/genetics , Symporters/metabolism , Testosterone/pharmacology
14.
Front Mol Biosci ; 9: 792829, 2022.
Article in English | MEDLINE | ID: mdl-35252349

ABSTRACT

Pals1 is part of the evolutionary conserved Crumbs polarity complex and plays a key role in two processes, the formation of apicobasal polarity and the establishment of cell-cell contacts. In the human kidney, up to 1.5 million nephrons control blood filtration, as well as resorption and recycling of inorganic and organic ions, sugars, amino acids, peptides, vitamins, water and further metabolites of endogenous and exogenous origin. All nephron segments consist of polarized cells and express high levels of Pals1. Mice that are functionally haploid for Pals1 develop a lethal phenotype, accompanied by heavy proteinuria and the formation of renal cysts. However, on a cellular level, it is still unclear if reduced cell polarization, incomplete cell-cell contact formation, or an altered Pals1-dependent gene expression accounts for the renal phenotype. To address this, we analyzed the transcriptomes of Pals1-haploinsufficient kidneys and the littermate controls by gene set enrichment analysis. Our data elucidated a direct correlation between TGFß pathway activation and the downregulation of more than 100 members of the solute carrier (SLC) gene family. Surprisingly, Pals1-depleted nephrons keep the SLC's segment-specific expression and subcellular distribution, demonstrating that the phenotype is not mainly due to dysfunctional apicobasal cell polarization of renal epithelia. Our data may provide first hints that SLCs may act as modulating factors for renal cyst formation.

15.
Article in English | MEDLINE | ID: mdl-35483776

ABSTRACT

Aging-related impaired body structure and functions may be, at least partially, caused by elevated oxidative stress. Melatonin (MEL) and resveratrol (RSV) may act as antioxidant and anti-aging compounds, but these actions in experimental animals and humans are controversial. Herein, a rat model of aging was used to study the long-term sex-related effects of MEL and RSV treatment on body mass and blood/plasma parameters of DNA damage, oxidative status (glutathione and malondialdehyde levels), and concentrations of sex hormones. Starting from the age of 3mo, for the next 9mo or 21mo male and female Wistar rats (n = 4-7 per group) were given water to drink (controls) or 0.1 % ethanol in water (vehicle), or MEL or RSV (each 10 mg/L vehicle). DNA damage in whole blood cells was tested by comet assay, whereas in plasma, glutathione, malondialdehyde, and sex hormones were determined by established methods. Using statistical analysis of data by ANOVA/Scheffe post hoc, we observed a similar sex- and aging-dependent rise of body mass in both sexes and drop of plasma testosterone in control and vehicle-treated male rats, whose pattern remained unaffected by MEL and RSV treatment. Compared with controls, all other parameters remained largely unchanged in aging and differently treated male and female rats. We concluded that the sex- and aging-related pattern of growth and various blood parameters in rats were not affected by the long-term treatment with MEL and RSV at the estimated daily doses (300-400 µg/kg b.m.) that exceed usual moderate consumption in humans.


Subject(s)
Melatonin , Aging , Animals , Biomarkers , Female , Glutathione , Male , Malondialdehyde , Melatonin/pharmacology , Rats , Rats, Wistar , Resveratrol/pharmacology , Water
16.
Am J Physiol Renal Physiol ; 301(5): F1026-34, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21865262

ABSTRACT

Organic anions are taken up from the blood into proximal tubule cells by organic anion transporters 1 and 3 (OAT1 and OAT3) in exchange for dicarboxylates. The released dicarboxylates are recycled by the sodium dicarboxylate cotransporter 3 (NaDC3). In this study, we tested the substrate specificities of human NaDC3, OAT1, and OAT3 to identify those dicarboxylates for which the three cooperating transporters have common high affinities. All transporters were stably expressed in HEK293 cells, and extracellularly added dicarboxylates were used as inhibitors of [(14)C]succinate (NaDC3), p-[(3)H]aminohippurate (OAT1), or [(3)H]estrone-3-sulfate (OAT3) uptake. Human NaDC3 was stably expressed as proven by immunochemical methods and by sodium-dependent uptake of succinate (K(0.5) for sodium activation, 44.6 mM; Hill coefficient, 2.1; K(m) for succinate, 18 µM). NaDC3 was best inhibited by succinate (IC(50) 25.5 µM) and less by α-ketoglutarate (IC(50) 69.2 µM) and fumarate (IC(50) 95.2 µM). Dicarboxylates with longer carbon backbones (adipate, pimelate, suberate) had low or no affinity for NaDC3. OAT1 exhibited the highest affinity for glutarate, α-ketoglutarate, and adipate (IC(50) between 3.3 and 6.2 µM), followed by pimelate (18.6 µM) and suberate (19.3 µM). The affinity of OAT1 to succinate and fumarate was low. OAT3 showed the same dicarboxylate selectivity with ∼13-fold higher IC(50) values compared with OAT1. The data 1) reveal α-ketoglutarate as a common high-affinity substrate of NaDC3, OAT1, and OAT3 and 2) suggest potentially similar molecular structures of the binding sites in OAT1 and OAT3 for dicarboxylates.


Subject(s)
Dicarboxylic Acid Transporters/metabolism , Dicarboxylic Acids/metabolism , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Symporters/metabolism , Amino Acid Sequence , Binding Sites , Blotting, Western , Dicarboxylic Acid Transporters/genetics , Dicarboxylic Acids/chemistry , Electrophoresis, Polyacrylamide Gel , Estrone/pharmacology , HEK293 Cells , Humans , Immunohistochemistry , Ketoglutaric Acids/metabolism , Molecular Sequence Data , Organic Anion Transport Protein 1/genetics , Organic Anion Transporters, Sodium-Dependent/genetics , Organic Anion Transporters, Sodium-Independent/genetics , Structure-Activity Relationship , Succinates/metabolism , Symporters/genetics , Transfection , p-Aminohippuric Acid/metabolism
17.
Biometals ; 23(5): 897-926, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20549307

ABSTRACT

Metallothioneins are cysteine-rich, small metal-binding proteins present in various mammalian tissues. Of the four common metallothioneins, MT-1 and MT-2 (MTs) are expressed in most tissues, MT-3 is predominantly present in brain, whereas MT-4 is restricted to the squamous epithelia. The expression of MT-1 and MT-2 in some organs exhibits sex, age, and strain differences, and inducibility with a variety of stimuli. In adult mammals, MTs have been localized largely in the cell cytoplasm, but also in lysosomes, mitochondria and nuclei. The major physiological functions of MTs include homeostasis of essential metals Zn and Cu, protection against cytotoxicity of Cd and other toxic metals, and scavenging free radicals generated in oxidative stress. The role of MTs in Cd-induced acute and chronic toxicity, particularly in liver and kidneys, is reviewed in more details. In acute toxicity, liver is the primary target, whereas in chronic toxicity, kidneys are major targets of Cd. The intracellular MTs bind Cd ions and form CdMT. In chronic intoxication, Cd stimulates de novo synthesis of MTs; it is assumed that toxicity in the cells starts when loading with Cd ions exceeds the buffering capacity of intracellular MTs. CdMT, released from the Cd-injured organs, or when applied parenterally for experimental purposes, reaches the kidneys via circulation, where it is filtered, endocytosed in the proximal tubule cells, and degraded in lysosomes. Liberated Cd can immediately affect the cell structures and functions. The resulting proteinuria and CdMT in the urine can be used as biomarkers of tubular injury.


Subject(s)
Cadmium/pharmacokinetics , Cadmium/toxicity , Kidney/drug effects , Kidney/metabolism , Metallothionein/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Compartmentation , Female , Humans , Intestinal Mucosa/metabolism , Intestines/drug effects , Liver/drug effects , Liver/metabolism , Lung/drug effects , Lung/metabolism , Male , Metallothionein/chemistry , Metallothionein/genetics , Models, Biological , Molecular Sequence Data , Sequence Homology, Amino Acid , Transcriptional Activation/drug effects , Zinc/metabolism
18.
ACS Omega ; 5(45): 29127-29139, 2020 Nov 17.
Article in English | MEDLINE | ID: mdl-33225144

ABSTRACT

In enterocytes, protein RS1 (RSC1A1) mediates an increase of glucose absorption after ingestion of glucose-rich food via upregulation of Na+-d-glucose cotransporter SGLT1 in the brush-border membrane (BBM). Whereas RS1 decelerates the exocytotic pathway of vesicles containing SGLT1 at low glucose levels between meals, RS1-mediated deceleration is relieved after ingestion of glucose-rich food. Regulation of SGLT1 is mediated by RS1 domain RS1-Reg, in which Gln-Ser-Pro (QSP) is effective. In contrast to QSP and RS1-Reg, Gln-Glu-Pro (QEP) and RS1-Reg with a serine to glutamate exchange in the QSP motif downregulate the abundance of SGLT1 in the BBM at high intracellular glucose concentrations by about 50%. We investigated whether oral application of QEP improves diabetes in db/db mice and affects the induction of diabetes in New Zealand obese (NZO) mice under glucolipotoxic conditions. After 6-day administration of drinking water containing 5 mM QEP to db/db mice, fasting glucose was decreased, increase of blood glucose in the oral glucose tolerance test was blunted, and insulin sensitivity was increased. When QEP was added for several days to a high fat/high carbohydrate diet that induced diabetes in NZO mice, the increase of random plasma glucose was prevented, accompanied by lower plasma insulin levels. QEP is considered a lead compound for development of new antidiabetic drugs with more rapid cellular uptake. In contrast to SGLT1 inhibitors, QEP-based drugs may be applied in combination with insulin for the treatment of type 1 and type 2 diabetes, decreasing the required insulin amount, and thereby may reduce the risk of hypoglycemia.

19.
Pflugers Arch ; 457(6): 1381-92, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19002488

ABSTRACT

The sulfate anion transporter (sat-1, Slc26a1) has been cloned from rat liver, functionally characterized, and localized to the sinusoidal membrane in hepatocytes and basolateral membrane (BLM) in proximal tubules (PT). Here, we confirm previously described localization of sat-1 protein in rat liver and kidneys and report on gender differences (GD) in its expression by immunochemical, transport, and excretion studies in rats. The approximately 85-kDa sat-1 protein was localized to the sinusoidal membrane in hepatocytes and BLM in renal cortical PT, with the male-dominant expression. However, the real-time reverse-transcription polymerase chain reaction data indicated no GD at the level of sat-1 mRNA. In agreement with the protein data, isolated membranes from both organs exhibited the male-dominant exchange of radiolabeled sulfate for oxalate, whereas higher oxalate in plasma and 24-h urine indicated higher oxalate production and excretion in male rats. Furthermore, the expression of liver, but not renal, sat-1 protein was: unaffected by castration, upregulated by ovariectomy, and downregulated by estrogen or progesterone treatment in males. Therefore, GD (males > females) in the expression of sat-1 protein in rat liver (and, possibly, kidneys) are caused by the female sex-hormone-driven inhibition at the posttranscriptional level. The male-dominant abundance of sat-1 protein in liver may conform to elevated uptake of sulfate and extrusion of oxalate, causing higher plasma oxalate in males. Oxalate is then excreted by the kidneys via the basolateral sat-1 (males > females) and the apical CFEX (Slc26a6; GD unknown) in PT and eliminated in the urine (males > females), where it may contribute to the male-prevailing development of oxalate urolithiasis.


Subject(s)
Anion Transport Proteins/biosynthesis , Antiporters/biosynthesis , Kidney/metabolism , Liver/metabolism , Animals , Castration , Cell Membrane/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Female , Gene Expression , Immunohistochemistry , Liver/drug effects , Male , Oxalates/urine , Progesterone/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors , Sulfate Transporters , Sulfates/metabolism
20.
Am J Physiol Renal Physiol ; 297(6): F1575-86, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19776169

ABSTRACT

Aquaporin 1 (AQP1) is the major water channel in the renal proximal tubule (PT) and thin descending limb of Henle, but its regulation remains elusive. Here, we investigated the effect of ANG II, a key mediator of body water homeostasis, on AQP1 expression in immortalized rat proximal tubule cells (IRPTC) and rat kidney. Real-time PCR on IRPTC exposed to ANG II for 12 h revealed a biphasic effect AQP1 mRNA increased dose dependently in response to 10(-12) to 10(-8) M ANG II but decreased by 50% with 10(-7) M ANG II. The twofold increase of AQP1 mRNA in the presence of 10(-8) M ANG II was abolished by the AT(1) receptor blocker losartan. Hypertonicity due to either NaCl or mannitol also upregulated AQP1 mRNA by three- and twofold, respectively. Immunocytochemistry and Western blotting revealed a two- to threefold increase in AQP1 protein expression in IRPTC exposed concomitantly to ANG II (10(-8)M) and hypertonic medium (either NaCl or mannitol), indicating that these stimuli were not additive. Three-dimensional reconstruction of confocal images suggested that AQP1 expression was increased by ANG II in both the apical and basolateral poles of IRPTC. In vivo studies showed that short-term ANG II infusion had a diuretic effect, while this effect was attenuated after several days of ANG II infusion. After 10 days, we observed a twofold increase in AQP1 expression in the PT and thin descending limb of Henle of ANG II-infused rats that was abolished when rats were treated with the selective AT(1)-receptor antagonist olmesartan. Thus ANG II increases AQP1 expression in vitro and in vivo via direct interaction with the AT(1) receptor, providing an important regulatory mechanism to link PT water reabsorption to body fluid homeostasis via the renin-angiotensin system.


Subject(s)
Angiotensin II/administration & dosage , Aquaporin 1/metabolism , Hypertonic Solutions/pharmacology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Aquaporin 1/genetics , Cell Line, Transformed , Diuresis , Dose-Response Relationship, Drug , Drug Synergism , Imidazoles/pharmacology , Kidney/drug effects , Kidney/metabolism , Kidney/physiology , Kidney Tubules, Proximal/cytology , Loop of Henle/drug effects , Loop of Henle/metabolism , Losartan/pharmacology , Male , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sodium Chloride/pharmacology , Tetrazoles/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL