Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Endocr J ; 70(3): 295-304, 2023 Mar 28.
Article in English | MEDLINE | ID: mdl-36450452

ABSTRACT

The symptoms of diabetes insipidus may be masked by the concurrence of adrenal insufficiency and emerge after the administration of hydrocortisone, occasionally at high doses. To elucidate the mechanism underlying polyuria induced by the administration of high-dose corticosteroids in the deficiency of arginine vasopressin (AVP), we first examined the secretion of AVP in three patients in whom polyuria was observed only after the administration of high-dose corticosteroids. Next, we examined the effects of dexamethasone or aldosterone on water balance in wild-type and familial neurohypophyseal diabetes insipidus (FNDI) model mice. A hypertonic saline test showed that AVP secretion was partially impaired in all patients. In one patient, there were no apparent changes in AVP secretion before and after the administration of high-dose corticosteroids. In FNDI mice, unlike dexamethasone, the administration of aldosterone increased urine volumes and decreased urine osmolality. Immunohistochemical analyses showed that, after the administration of aldosterone in FNDI mice, aquaporin-2 expression was decreased in the apical membrane and increased in the basolateral membrane in the collecting duct. These changes were not observed in wild-type mice. The present data suggest that treatment with mineralocorticoids induces polyuria by reducing aquaporin-2 expression in the apical membrane of the kidney in partial AVP deficiency.


Subject(s)
Diabetes Insipidus, Neurogenic , Diabetes Insipidus , Mice , Animals , Polyuria/genetics , Aquaporin 2/genetics , Mineralocorticoids , Aldosterone , Kidney/metabolism , Arginine Vasopressin/genetics , Arginine Vasopressin/metabolism , Dexamethasone/pharmacology
2.
J Am Soc Nephrol ; 32(6): 1339-1354, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33727367

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs), formed by cleavage of pre-microRNA by the endoribonuclease Dicer, are critical modulators of cell function by post-transcriptionally regulating gene expression. METHODS: Selective ablation of Dicer in AQP2-expressing cells (DicerAQP2Cre+ mice) was used to investigate the role of miRNAs in the kidney collecting duct of mice. RESULTS: The mice had severe polyuria and nephrogenic diabetes insipidus, potentially due to greatly reduced AQP2 and AQP4 levels. Although epithelial sodium channel levels were decreased in cortex and increased in inner medulla, amiloride-sensitive sodium reabsorption was equivalent in DicerAQP2Cre+ mice and controls. Small-RNA sequencing and proteomic analysis revealed 31 and 178 significantly regulated miRNAs and proteins, respectively. Integrated bioinformatic analysis of the miRNAome and proteome suggested alterations in the epigenetic machinery and various transcription factors regulating AQP2 expression in DicerAQP2Cre+ mice. The expression profile and function of three miRNAs (miR-7688-5p, miR-8114, and miR-409-3p) whose predicted targets were involved in epigenetic control (Phf2, Kdm5c, and Kdm4a) or transcriptional regulation (GATA3, GATA2, and ELF3) of AQP2 were validated. Luciferase assays could not demonstrate direct interaction of AQP2 or the three potential transcription factors with miR-7688-5p, miR-8114, and miR-409-3p. However, transfection of respective miRNA mimics reduced AQP2 expression. Chromatin immunoprecipitation assays demonstrated decreased Phf2 and significantly increased Kdm5c interactions at the Aqp2 gene promoter in DicerAQP2Cre+ mice, resulting in decreased RNA Pol II association. CONCLUSIONS: Novel evidence indicates miRNA-mediated epigenetic regulation of AQP2 expression.


Subject(s)
Aquaporin 2/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation , MicroRNAs/genetics , Ribonuclease III/genetics , Animals , Aquaporin 2/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diabetes Insipidus, Nephrogenic/genetics , Diabetes Insipidus, Nephrogenic/metabolism , Down-Regulation , Epithelial Sodium Channels/metabolism , Female , GATA2 Transcription Factor/genetics , GATA3 Transcription Factor/genetics , Histone Demethylases/genetics , Histone Demethylases/metabolism , Homeodomain Proteins/genetics , Kidney Tubules, Collecting/physiology , Male , Mice , Polyuria/genetics , Polyuria/metabolism , Proteome , RNA Processing, Post-Transcriptional , Renal Reabsorption , Sequence Analysis, RNA , Transcription Factors/genetics , Transcription Factors/metabolism
3.
FASEB J ; 33(2): 2156-2170, 2019 02.
Article in English | MEDLINE | ID: mdl-30252533

ABSTRACT

cAMP is a universal second messenger regulating a plethora of processes in the kidney. Two downstream effectors of cAMP are PKA and exchange protein directly activated by cAMP (Epac), which, unlike PKA, is often linked to elevation of [Ca2+]i. While both Epac isoforms (Epac1 and Epac2) are expressed along the nephron, their relevance in the kidney remains obscure. We combined ratiometric calcium imaging with quantitative immunoblotting, immunofluorescent confocal microscopy, and balance studies in mice lacking Epac1 or Epac2 to determine the role of Epac in renal water-solute handling. Epac1-/- and Epac2-/- mice developed polyuria despite elevated arginine vasopressin levels. We did not detect major deficiencies in arginine vasopressin [Ca2+]i signaling in split-opened collecting ducts or decreases in aquaporin water channel type 2 levels. Instead, sodium-hydrogen exchanger type 3 levels in the proximal tubule were dramatically reduced in Epac1-/- and Epac2-/- mice. Water deprivation revealed persisting polyuria, impaired urinary concentration ability, and augmented urinary excretion of Na+ and urea in both mutant mice. In summary, we report a nonredundant contribution of Epac isoforms to renal function. Deletion of Epac1 and Epac2 decreases sodium-hydrogen exchanger type 3 expression in the proximal tubule, leading to polyuria and osmotic diuresis.-Cherezova, A., Tomilin, V., Buncha, V., Zaika, O., Ortiz, P. A., Mei, F., Cheng, X., Mamenko, M., Pochynyuk, O. Urinary concentrating defect in mice lacking Epac1 or Epac2.


Subject(s)
Guanine Nucleotide Exchange Factors/genetics , Kidney Concentrating Ability/genetics , Animals , Aquaporin 2/metabolism , Arginine Vasopressin/metabolism , Calcium Signaling , Diuresis , Gene Deletion , Kidney/metabolism , Kidney/physiology , Mice , Mice, Knockout , Osmosis , Polyuria/genetics , Sodium-Hydrogen Exchanger 3/metabolism
4.
J Am Soc Nephrol ; 28(10): 2887-2900, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28507058

ABSTRACT

The transcription factor hepatocyte nuclear factor-1ß (HNF-1ß) is essential for normal kidney development and function. Inactivation of HNF-1ß in mouse kidney tubules leads to early-onset cyst formation and postnatal lethality. Here, we used Pkhd1/Cre mice to delete HNF-1ß specifically in renal collecting ducts (CDs). CD-specific HNF-1ß mutant mice survived long term and developed slowly progressive cystic kidney disease, renal fibrosis, and hydronephrosis. Compared with wild-type littermates, HNF-1ß mutant mice exhibited polyuria and polydipsia. Before the development of significant renal structural abnormalities, mutant mice exhibited low urine osmolality at baseline and after water restriction and administration of desmopressin. However, mutant and wild-type mice had similar plasma vasopressin and solute excretion levels. HNF-1ß mutant kidneys showed increased expression of aquaporin-2 mRNA but mislocalized expression of aquaporin-2 protein in the cytoplasm of CD cells. Mutant kidneys also had decreased expression of the UT-A urea transporter and collectrin, which is involved in apical membrane vesicle trafficking. Treatment of HNF-1ß mutant mIMCD3 cells with hypertonic NaCl inhibited the induction of osmoregulated genes, including Nr1h4, which encodes the transcription factor FXR that is required for maximal urinary concentration. Chromatin immunoprecipitation and sequencing experiments revealed HNF-1ß binding to the Nr1h4 promoter in wild-type kidneys, and immunoblot analysis revealed downregulated expression of FXR in HNF-1ß mutant kidneys. These findings reveal a novel role of HNF-1ß in osmoregulation and identify multiple mechanisms, whereby mutations of HNF-1ß produce defects in urinary concentration.


Subject(s)
Hepatocyte Nuclear Factor 1-beta/physiology , Kidney Tubules, Collecting/physiology , Animals , Cell Line , Female , Gene Expression Regulation , Male , Mice, Transgenic , Polyuria/genetics , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Urine
5.
Am J Physiol Renal Physiol ; 313(4): F1026-F1037, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28701310

ABSTRACT

The renal collecting duct (CD) contains two major cell types, intercalated (ICs) and principal cells (PCs). A previous report showed that deletion of ß1-integrin in the entire renal CD causes defective CD morphogenesis resulting in kidney dysfunction. However, subsequent deletion of ß1-integrin specifically in ICs and PCs, respectively, did not cause any morphological defects in the CDs. The discrepancy between these studies prompts us to reinvestigate the role of ß1-integrin in CD cells, specifically in the PCs. We conditionally deleted ß1-integrin in mouse CD PCs using a specific aquaporin-2 (AQP2) promoter Cre-LoxP system. The resulting mutant mice, ß-1f/fAQP2-Cre+, had lower body weight, failed to thrive, and died around 8-12 wk. Their CD tubules were dilated, and some of them contained cellular debris. Increased apoptosis and proliferation of PCs were observed in the dilated CDs. Trichrome staining and electron microscopy revealed the presence of peritubular and interstitial fibrosis that is associated with increased production of extracellular matrix proteins including collagen type IV and fibronectin, as detected by immunoblotting. Further analysis revealed a significantly increased expression of transforming growth factor-ß (TGF-ß)-induced protein, fibronectin, and TGF-ß receptor-1 mRNAs and concomitantly increased phosphorylation of SMAD-2 that indicates the activation of the TGF-ß signaling pathway. Therefore, our data reveal that normal expression of ß1-integrin in PCs is a critical determinant of CD structural and functional integrity and further support the previously reported critical role of ß1-integrin in the development and/or maintenance of the CD structure and function.


Subject(s)
Extracellular Matrix/metabolism , Gene Deletion , Integrin beta1/metabolism , Kidney Medulla/metabolism , Kidney Tubules, Collecting/metabolism , Polyuria/metabolism , Renal Insufficiency/metabolism , Age Factors , Animals , Apoptosis , Aquaporin 2/genetics , Cell Proliferation , Extracellular Matrix/ultrastructure , Failure to Thrive/genetics , Failure to Thrive/metabolism , Failure to Thrive/pathology , Fibrosis , Genetic Predisposition to Disease , Integrases/genetics , Integrin beta1/genetics , Kidney Medulla/ultrastructure , Kidney Tubules, Collecting/ultrastructure , Mice, Knockout , Phenotype , Phosphorylation , Polyuria/genetics , Polyuria/pathology , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Renal Insufficiency/genetics , Renal Insufficiency/pathology , Signal Transduction , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism
6.
Kidney Int ; 91(4): 842-855, 2017 04.
Article in English | MEDLINE | ID: mdl-28143656

ABSTRACT

Dent disease is a rare X-linked tubulopathy caused by mutations in the endosomal chloride-proton exchanger (ClC-5) resulting in defective receptor-mediated endocytosis and severe proximal tubule dysfunction. Bone marrow transplantation has recently been shown to preserve kidney function in cystinosis, a lysosomal storage disease causing proximal tubule dysfunction. Here we test the effects of bone marrow transplantation in Clcn5Y/- mice, a faithful model for Dent disease. Transplantation of wild-type bone marrow in Clcn5Y/- mice significantly improved proximal tubule dysfunction, with decreased low-molecular-weight proteinuria, glycosuria, calciuria, and polyuria four months after transplantation, compared to Clcn5Y/- mice transplanted with ClC-5 knockout bone marrow. Bone marrow-derived cells engrafted in the interstitium, surrounding proximal tubule cells, which showed a rescue of the apical expression of ClC-5 and megalin receptors. The improvement of proximal tubule dysfunction correlated with Clcn5 gene expression in kidneys of mice transplanted with wild-type bone marrow cells. Coculture of Clcn5Y/- proximal tubule cells with bone marrow-derived cells confirmed rescue of ClC-5 and megalin, resulting in improved endocytosis. Nanotubular extensions between the engrafted bone marrow-derived cells and proximal tubule cells were observed in vivo and in vitro. No rescue was found when the formation of the tunneling nanotubes was prevented by actin depolymerization or when cells were physically separated by transwell inserts. Thus, bone marrow transplantation may rescue the epithelial phenotype due to an inherited endosomal defect. Direct contacts between bone marrow-derived cells and diseased tubular cells play a key role in the rescue mechanism.


Subject(s)
Bone Marrow Transplantation , Chloride Channels/deficiency , Dent Disease/surgery , Kidney Tubules, Proximal/physiopathology , Animals , Cell Communication , Cells, Cultured , Chloride Channels/genetics , Coculture Techniques , Dent Disease/genetics , Dent Disease/metabolism , Dent Disease/physiopathology , Disease Models, Animal , Endocytosis , Genetic Predisposition to Disease , Glycosuria/genetics , Glycosuria/metabolism , Glycosuria/physiopathology , Glycosuria/prevention & control , Hypercalciuria/genetics , Hypercalciuria/metabolism , Hypercalciuria/physiopathology , Hypercalciuria/prevention & control , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Polyuria/genetics , Polyuria/metabolism , Polyuria/physiopathology , Polyuria/prevention & control , Proteinuria/genetics , Proteinuria/metabolism , Proteinuria/physiopathology , Proteinuria/prevention & control , Recovery of Function , Transplantation Chimera
7.
Neurourol Urodyn ; 36(4): 1034-1038, 2017 04.
Article in English | MEDLINE | ID: mdl-27348079

ABSTRACT

AIMS: The pathophysiologies of nocturia (NOC) and nocturnal polyuria (NP) are multifactorial and their etiologies remain unclear in a large number of patients. Clock genes exist in most cells and organs, and the products of Clock regulate circadian rhythms as representative clock genes. Clock genes regulate lower urinary tract function, and a newly suggested concept is that abnormalities in clock genes cause lower urinary tract symptoms. In the present study, we investigated the voiding behavior of Clock mutant (ClockΔ19/Δ19 ) mice in order to determine the effects of clock genes on NOC/NP. METHODS: Male C57BL/6 mice aged 8-12 weeks (WT) and male C57BL/6 ClockΔ19/Δ19 mice aged 8 weeks were used. They were bred under 12 hr light/dark conditions for 2 weeks and voiding behavior was investigated by measuring water intake volume, urine volume, urine volume/void, and voiding frequency in metabolic cages in the dark and light periods. RESULTS: No significant differences were observed in behavior patterns between ClockΔ19/Δ19 and WT mice. ClockΔ19/Δ19 mice showed greater voiding frequencies and urine volumes during the sleep phase than WT mice. The diurnal change in urine volume/void between the dark and light periods in WT mice was absent in ClockΔ19/Δ19 mice. Additionally, functional bladder capacity was significantly lower in ClockΔ19/Δ19 mice than in WT mice. CONCLUSIONS: We demonstrated that ClockΔ19/Δ19 mice showed the phenotype of NOC/NP. The ClockΔ19/Δ19 mouse may be used as an animal model of NOC and NP. Neurourol. Urodynam. 36:1034-1038, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
CLOCK Proteins/genetics , Circadian Rhythm/genetics , Nocturia/genetics , Polyuria/genetics , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL
8.
Adv Exp Med Biol ; 969: 131-148, 2017.
Article in English | MEDLINE | ID: mdl-28258571

ABSTRACT

Several aquaporin (AQP )-type water channels are expressed in kidney: AQP1 in the proximal tubule, thin descending limb of Henle, and vasa recta; AQP2 -6 in the collecting duct; AQP7 in the proximal tubule; AQP8 in the proximal tubule and collecting duct; and AQP11 in the endoplasmic reticulum of proximal tubule cells. AQP2 is the vasopressin-regulated water channel that is important in hereditary and acquired diseases affecting urine-concentrating ability. The roles of AQPs in renal physiology and transepithelial water transport have been determined using AQP knockout mouse models. This chapter describes renal physiologic insights revealed by phenotypic analysis of AQP knockout mice and the prospects for further basic and clinical studies.


Subject(s)
Aquaporin 1/metabolism , Kidney/metabolism , Polyuria/metabolism , Urea/metabolism , Water/metabolism , Animals , Aquaporin 1/genetics , Biological Transport , Gene Expression Regulation , Humans , Kidney/cytology , Kidney Concentrating Ability/physiology , Mice , Mice, Knockout , Osmolar Concentration , Polyuria/genetics , Polyuria/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Water-Electrolyte Balance/physiology
9.
Proc Natl Acad Sci U S A ; 111(27): E2817-26, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-24958889

ABSTRACT

Mammalian target of rapamycin complex 1 (mTORC1) is a key regulator of cell metabolism and autophagy. Despite widespread clinical use of mTORC1 inhibitors, the role of mTORC1 in renal tubular function and kidney homeostasis remains elusive. By using constitutive and inducible deletion of conditional Raptor alleles in renal tubular epithelial cells, we discovered that mTORC1 deficiency caused a marked concentrating defect, loss of tubular cells, and slowly progressive renal fibrosis. Transcriptional profiling revealed that mTORC1 maintains renal tubular homeostasis by controlling mitochondrial metabolism and biogenesis as well as transcellular transport processes involved in countercurrent multiplication and urine concentration. Although mTORC2 partially compensated for the loss of mTORC1, exposure to ischemia and reperfusion injury exaggerated the tubular damage in mTORC1-deficient mice and caused pronounced apoptosis, diminished proliferation rates, and delayed recovery. These findings identify mTORC1 as an important regulator of tubular energy metabolism and as a crucial component of ischemic stress responses.


Subject(s)
Homeostasis/physiology , Ischemia/physiopathology , Kidney Tubules/physiology , Multiprotein Complexes/physiology , TOR Serine-Threonine Kinases/physiology , Animals , Blotting, Western , Kidney Tubules/blood supply , Magnetic Resonance Imaging , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/genetics , Polyuria/genetics , TOR Serine-Threonine Kinases/genetics , Transcription, Genetic
10.
J Biol Chem ; 290(51): 30406-16, 2015 Dec 18.
Article in English | MEDLINE | ID: mdl-26453302

ABSTRACT

ClC-K chloride channels are crucial for auditory transduction and urine concentration. Mutations in CLCNKB, the gene encoding the renal chloride channel hClC-Kb, cause Bartter syndrome type III, a human genetic condition characterized by polyuria, hypokalemia, and alkalosis. In recent years, several Bartter syndrome-associated mutations have been described that result in truncations of the intracellular carboxyl terminus of hClC-Kb. We here used a combination of whole-cell patch clamp, confocal imaging, co-immunoprecipitation, and surface biotinylation to study the functional consequences of a frequent CLCNKB mutation that creates a premature stop codon at Trp-610. We found that W610X leaves the association of hClC-Kb and the accessory subunit barttin unaffected, but impairs its regulation by barttin. W610X attenuates hClC-Kb surface membrane insertion. Moreover, W610X results in hClC-Kb channel opening in the absence of barttin and prevents further barttin-mediated activation. To describe how the carboxyl terminus modifies the regulation by barttin we used V166E rClC-K1. V166E rClC-K1 is active without barttin and exhibits prominent, barttin-regulated voltage-dependent gating. Electrophysiological characterization of truncated V166E rClC-K1 demonstrated that the distal carboxyl terminus is necessary for slow cooperative gating. Since barttin modifies this particular gating process, channels lacking the distal carboxyl-terminal domain are no longer regulated by the accessory subunit. Our results demonstrate that the carboxyl terminus of hClC-Kb is not part of the binding site for barttin, but functionally modifies the interplay with barttin. The loss-of-activation of truncated hClC-Kb channels in heterologous expression systems fully explains the reduced basolateral chloride conductance in affected kidneys and the clinical symptoms of Bartter syndrome patients.


Subject(s)
Chloride Channels/metabolism , Ion Channel Gating , Animals , Binding Sites , Chloride Channels/genetics , Codon, Nonsense/genetics , Dogs , HEK293 Cells , Humans , Hypokalemia/genetics , Hypokalemia/metabolism , Kidney/metabolism , Kidney/pathology , Madin Darby Canine Kidney Cells , Polyuria/genetics , Polyuria/metabolism , Polyuria/pathology , Protein Transport/genetics
11.
J Am Soc Nephrol ; 26(1): 149-59, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24904084

ABSTRACT

A disintegrin and metalloproteinase domain 10 (Adam10), a member of the ADAM family of cell membrane-anchored proteins, has been linked to the regulation of the Notch, EGF, E-cadherin, and other signaling pathways. However, it is unclear what role Adam10 has in the kidney in vivo. In this study, we showed that Adam10 deficiency in ureteric bud (UB) derivatives leads to a decrease in urinary concentrating ability, polyuria, and hydronephrosis in mice. Furthermore, Adam10 deficiency led to a reduction in the percentage of aquaporin 2 (Aqp2)(+) principal cells (PCs) in the collecting ducts that was accompanied by a proportional increase in the percentage of intercalated cells (ICs). This increase was more prominent in type A ICs than in type B ICs. Foxi1, a transcription factor important for the differentiation of ICs, was upregulated in the Adam10 mutants. The observed reduction of Notch activity in Adam10 mutant collecting duct epithelium and the similar reduction of PC/IC ratios in the collecting ducts in mice deficient for mindbomb E3 ubiquitin protein ligase 1, a key regulator of the Notch and Wnt/receptor-like tyrosine kinase signaling pathways, suggest that Adam10 regulates cell fate determination through the activation of Notch signaling, probably through the regulation of Foxi1 expression. However, phenotypic differences between the Adam10 mutants, the Mib1 mutants, and the Foxi1 mutants suggest that the functions of Adam10 in determining the fate of collecting duct cells are more complex than those of a simple upstream factor in a linear pathway involving Notch and Foxi1.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Kidney/metabolism , Membrane Proteins/metabolism , ADAM10 Protein , Animals , Apoptosis , Aquaporin 2/metabolism , Cadherins/metabolism , Cell Proliferation , Epithelial Cells/cytology , Forkhead Transcription Factors/metabolism , Hydronephrosis/genetics , Kidney Tubules/cytology , Kidney Tubules, Collecting/metabolism , Ligands , Mice , Mice, Transgenic , Mutation , Polyuria/genetics , Receptors, Notch/metabolism , Signal Transduction , Stem Cells/cytology , Up-Regulation , Wnt Signaling Pathway
12.
Am J Physiol Renal Physiol ; 308(6): F650-60, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25608967

ABSTRACT

In mammals, glycogen synthase kinase (GSK)3 comprises GSK3α and GSK3ß isoforms. GSK3ß has been shown to play a role in the ability of kidneys to concentrate urine by regulating vasopressin-mediated water permeability of collecting ducts, whereas the role of GSK3α has yet to be discerned. To investigate the role of GSK3α in urine concentration, we compared GSK3α knockout (GSK3αKO) mice with wild-type (WT) littermates. Under normal conditions, GSK3αKO mice had higher water intake and urine output. GSK3αKO mice also showed reduced urine osmolality and aquaporin-2 levels but higher urinary vasopressin. When water deprived, they failed to concentrate their urine to the same level as WT littermates. The addition of 1-desamino-8-d-arginine vasopressin to isolated inner medullary collecting ducts increased the cAMP response in WT mice, but this response was reduced in GSK3αKO mice, suggesting reduced responsiveness to vasopressin. Gene silencing of GSK3α in mpkCCD cells also reduced forskolin-induced aquaporin-2 expression. When treated with LiCl, an isoform nonselective inhibitor of GSK3 and known inducer of polyuria, WT mice developed significant polyuria within 6 days. However, in GSK3αKO mice, the polyuric response was markedly reduced. This study demonstrates, for the first time, that GSK3α could play a crucial role in renal urine concentration and suggest that GSK3α might be one of the initial targets of Li(+) in LiCl-induced nephrogenic diabetes insipidus.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Kidney Tubules, Collecting/enzymology , Urine/physiology , Animals , Aquaporin 2/metabolism , Gene Silencing , Glycogen Synthase Kinase 3/genetics , Lithium Chloride , Mice, Knockout , Polyuria/genetics
13.
Am J Physiol Renal Physiol ; 309(6): F501-13, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26136556

ABSTRACT

Mammalian class IX myosin Myo9a is a single-headed, actin-dependent motor protein with Rho GTPase-activating protein activity that negatively regulates Rho GTPase signaling. Myo9a is abundantly expressed in ciliated epithelial cells of several organs. In mice, genetic deletion of Myo9a leads to the formation of hydrocephalus. Whether Myo9a also has essential functions in the epithelia of other organs of the body has not been explored. In the present study, we report that Myo9a-deficient mice develop bilateral renal disease, characterized by dilation of proximal tubules, calyceal dilation, and thinning of the parenchyma and fibrosis. These structural changes are accompanied by polyuria (with normal vasopressin levels) and low-molecular-weight proteinuria. Immunohistochemistry revealed that Myo9a is localized to the circumferential F-actin belt of proximal tubule cells. In kidneys lacking Myo9a, the multiligand binding receptor megalin and its ligand albumin accumulated at the luminal surface of Myo9a-deficient proximal tubular cells, suggesting that endocytosis is dysregulated. In addition, we found, surprisingly, that levels of murine diaphanous-related formin-1, a Rho effector, were decreased in Myo9a-deficient kidneys as well as in Myo9a knockdown LLC-PK1 cells. In summary, deletion of the Rho GTPase-activating protein Myo9a in mice causes proximal tubular dilation and fibrosis, and we speculate that downregulation of murine diaphanous-related formin-1 and impaired protein reabsorption contribute to the pathophysiology.


Subject(s)
GTPase-Activating Proteins/physiology , Kidney Tubules/physiology , Myosins/physiology , Albumins/metabolism , Animals , Carrier Proteins/metabolism , Cells, Cultured , Endocytosis/physiology , Formins , GTPase-Activating Proteins/genetics , Hydronephrosis/genetics , Hydronephrosis/metabolism , Kidney Tubules/anatomy & histology , Kidney Tubules/cytology , LLC-PK1 Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Myosins/genetics , Nephrons/physiology , Polyuria/genetics , Polyuria/metabolism , Swine , Vasopressins/metabolism , rho-Associated Kinases/metabolism
14.
Calcif Tissue Int ; 96(4): 265-73, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25555744

ABSTRACT

Familial hypomagnesemia with hypercalciuria and nephrocalcinosis (FHHNC) is an autosomal recessive disorder caused by mutations in the CLDN16 or CLDN19 genes, encoding claudin-16 and claudin-19 in the thick ascending limb of Henle's loop. In patients with claudin-19 mutations, severe ocular involvement (macular coloboma, pigmentary retinitis, nystagmus, or visual loss) has been described. In this report, we presented a 12-year-old girl with rickets, polyuria, and polydipsia. She was the daughter of consanguineous parents, and she had a history of recurred hypocalcemic and hypomagnesemic tetany. On physical examination, bilateral horizontal nystagmus and severe myopia were detected. Laboratory examination revealed hypomagnesemia, hypocalcemia, hypercalciuria, nephrocalcinosis, and renal stone. A clinical diagnosis of FHHNC caused possibly by claudin-19 mutation was decided with the ocular findings. DNA analysis revealed a novel homozygous missense mutation c.241C>T in the CLDN19 gene. In conclusion, in a patient with hypomagnesemia, hypercalciuria, nephrocalcinosis, and ocular findings, a diagnosis of FHHNC caused by claudin-19 mutation should be considered. This is the first study of FHHNC in Chinese population. Our findings of the novel mutation c.241C>T in exon 2 add to the list of more than 16 mutations of CLDN19 gene reported.


Subject(s)
Claudins/genetics , Hypercalciuria/genetics , Hypocalcemia/genetics , Magnesium Deficiency/congenital , Mutation, Missense , Nephrocalcinosis/genetics , Amino Acid Sequence , Child , China , Consanguinity , DNA Mutational Analysis , Exons , Female , Humans , Magnesium Deficiency/genetics , Molecular Sequence Data , Polydipsia/genetics , Polyuria/genetics , Rickets/genetics , Sequence Homology, Amino Acid
15.
FASEB J ; 28(8): 3645-59, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24784577

ABSTRACT

One of the clinical alterations observed in chronic renal disease (CRD) is the impaired urine concentration, known as diabetes insipidus (DI). Tubulointerstitial fibrosis of the kidney is also a pathological finding observed in CRD and involves composition of extracellular matrix (ECM). However, an association between these two events has not been elucidated. In this study, we showed that the extracellular-to-intracellular scaffold protein integrin-linked kinase (ILK) regulates expression of tubular water channel aquaporin-2 (AQP2) and its apical membrane presence in the renal tubule. Basally, polyuria and decreased urine osmolality were present in ILK conditional-knockdown (cKD-ILK) adult mice compared with nondepleted ILK littermates. No changes were observed in arginine-vasopressin (AVP) blood levels, renal receptor (V2R), or AQP3 expression. However, tubular AQP2 was decreased in expression and apical membrane presence in cKD-ILK mice, where the canonical V2R/cAMP axis activation is still functional, but independent of the absence of ILK. Thus, cKD-ILK constitutes a nephrogenic diabetes insipidus (NDI) model. AQP2 and ILK colocalize in cultured inner medullary collecting duct (mIMCD3) cells. Specific ILK siRNAs and collagen I (Col) decrease ILK and AQP2 levels and AQP2 presence on the membrane of tubular mIMCD3 cells, which impairs the capacity of the cells to transport water under hypotonic stress. The present work points to ILK as a therapeutic target in NDI.


Subject(s)
Aquaporin 2/physiology , Body Water/metabolism , Extracellular Matrix Proteins/physiology , Kidney Concentrating Ability/physiology , Kidney Tubules, Collecting/metabolism , Polyuria/metabolism , Protein Serine-Threonine Kinases/physiology , Animals , Aquaporin 2/biosynthesis , Aquaporin 2/genetics , Aquaporin 3/biosynthesis , Aquaporin 3/genetics , Arginine Vasopressin/blood , Biological Transport, Active , Cell Membrane/chemistry , Cell Polarity , Cells, Cultured , Collagen Type I/pharmacology , Deamino Arginine Vasopressin/pharmacology , Diabetes Insipidus, Nephrogenic/metabolism , Disease Models, Animal , Kidney Tubules, Collecting/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osmolar Concentration , Osmotic Pressure/physiology , Phosphorylation , Polyuria/genetics , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , RNA Interference , RNA, Small Interfering/pharmacology , Receptors, Vasopressin/biosynthesis , Receptors, Vasopressin/genetics
16.
J Am Soc Nephrol ; 24(2): 243-52, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23308014

ABSTRACT

The mammalian collecting duct comprises principal and intercalated cells, which maintain sodium/water and acid/base balance, respectively, but the epigenetic contributors to the differentiation of these cell types remain unknown. Here, we investigated whether the histone H3 K79 methyltransferase Dot1l, which is highly expressed in principal cells, participates in this process. Taking advantage of the distribution of aquaporin 2 (Aqp2), which localizes to principal cells of the collecting duct, we developed mice lacking Dot1l in Aqp2-expressing cells (Dot1l(AC)) and found that these mice had approximately 20% fewer principal cells and 13%-16% more intercalated cells than control mice. This deletion of Dot1l in principal cells abolished histone H3 K79 methylation in these cells, but unexpectedly, most intercalated cells also had undetectable di-methyl K79, suggesting that Aqp2(+) cells give rise to intercalated cells. These Aqp2(+) cell-derived intercalated cells were present in both developing and mature kidneys. Furthermore, compared with control mice, Dot1l(AC) mice had 40% higher urine volume and 18% lower urine osmolarity with relatively normal electrolyte and acid-base homeostasis. In conclusion, these data suggest that Dot1l deletion facilitates the differentiation of some α- and ß-intercalated cells from Aqp2-expressing progenitor cells or mature principal cells.


Subject(s)
Aquaporin 2/genetics , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/physiology , Methyltransferases/genetics , Water-Electrolyte Balance/genetics , Acid-Base Imbalance/genetics , Acid-Base Imbalance/pathology , Acid-Base Imbalance/physiopathology , Animals , Aquaporin 2/metabolism , Cell Differentiation/physiology , Cell Lineage/physiology , Epigenesis, Genetic/physiology , Female , Histone-Lysine N-Methyltransferase , Histones/metabolism , Integrases/genetics , Male , Methylation , Methyltransferases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Polyuria/genetics , Polyuria/pathology , Polyuria/physiopathology , Water-Electrolyte Imbalance/genetics , Water-Electrolyte Imbalance/pathology , Water-Electrolyte Imbalance/physiopathology
17.
Cell Physiol Biochem ; 32(7): 173-83, 2013.
Article in English | MEDLINE | ID: mdl-24429824

ABSTRACT

BACKGROUND AND AIMS: The thiazide-sensitive Na(+)-Cl(-) cotransporter NCC and the Cl(-)/HCO3(-)exchanger pendrin are expressed on apical membranes of distal cortical nephron segments and mediate salt absorption, with pendrin working in tandem with the epithelial Na(+) channel (ENaC) and the Na(+)-dependent chloride/bicarbonate exchanger (NDCBE), whereas NCC is working by itself. A recent study showed that NCC and pendrin compensate for loss of each other under basal conditions, therefore masking the role that each plays in salt reabsorption. Carbonic anhydrase II (CAII, CA2 or CAR2) plays an important role in acid-base transport and salt reabsorption in the proximal convoluted tubule and acid-base transport in the collecting duct. Animals with CAII deletion show remodeling of intercalated cells along with the downregulation of pendrin. NCC KO mice on the other hand show significant upregulation of pendrin and ENaC. Neither model shows any significant salt wasting under baseline conditions. We hypothesized that the up-regulation of pendrin is essential for the prevention of salt wasting in NCC KO mice. METHODS AND RESULTS: To test this hypothesis, we generated NCC/CAII double KO (dKO) mice by crossing mice with single deletion of NCC and CAII. The NCC/CAII dKO mice displayed significant downregulation of pendrin, along with polyuria and salt wasting. As a result, the dKO mice developed volume depletion, which was associated with the inability to concentrate urine. CONCLUSIONS: We conclude that the upregulation of pendrin is essential for the prevention of salt and water wasting in NCC deficient animals and its downregulation or inactivation will result in salt wasting, impaired water conservation and volume depletion in the setting of NCC inactivation or inhibition.


Subject(s)
Anion Transport Proteins/genetics , Carbonic Anhydrase II/metabolism , Kidney Tubules, Collecting/metabolism , Animals , Anion Transport Proteins/biosynthesis , Carbonic Anhydrase II/genetics , Chloride-Bicarbonate Antiporters/metabolism , Gene Expression Regulation , Mice , Mice, Knockout , Polyuria/genetics , Polyuria/metabolism , Salts/urine , Sodium Chloride/metabolism , Solute Carrier Family 12, Member 3/biosynthesis , Solute Carrier Family 12, Member 3/genetics , Solute Carrier Family 12, Member 3/metabolism , Sulfate Transporters
18.
Am J Kidney Dis ; 62(6): 1160-4, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23871407

ABSTRACT

A pregnant woman presented at gestational week 28 with loss of consciousness and profound polyuria. Further characterization revealed osmotic diuresis due to massive glycosuria without hyperglycemia. Glycosuria reduced substantially postpartum, from approximately 100 to approximately 30 g/1.73 m2 per day. DNA sequencing analysis of the SLC5A2 gene encoding the renal glucose transporter SGLT2 showed a homozygous frame-shift mutation (occurring after the glutamine at amino acid 168 and leading to premature termination of the protein at amino acid 186) diagnostic of familial renal glycosuria. Pregnant women with familial renal glycosuria can be at risk of profound polyuria during pregnancy due to the associated increase in glycosuria. These findings also have implications for the use of SGLT2 inhibitors in clinical practice.


Subject(s)
Glycosuria, Renal/genetics , Polyuria/genetics , Pregnancy Complications/genetics , Adult , Diagnosis, Differential , Female , Frameshift Mutation/genetics , Glucose Intolerance/diagnosis , Glucose Intolerance/genetics , Glycosuria, Renal/diagnosis , Homozygote , Humans , Kidney Function Tests , Pregnancy , Pregnancy Complications/diagnosis , Pregnancy Trimester, Second , Sequence Analysis, DNA , Sodium-Glucose Transporter 2/genetics
19.
Pediatr Nephrol ; 27(1): 115-21, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21786142

ABSTRACT

We report the molecular findings for the CTNS gene in 12 Turkish cystinosis patients aged 7-29 years. All presented initially with severe failure to thrive, polyuria, and polydipsia. Cystinosis was diagnosed at age 1 month to 9 years. Seven patients reached end-stage renal failure at ages ranging from 6.5 to 15 years. Whereas three of the remaining five have renal Fanconi syndrome with proteinuria, two have had kidney failure of varying degrees. Molecular analyses involved an initial multiplex polymerase chain reaction (PCR) to determine the presence or absence of the 57-kb northern European founder deletion in CTNS, followed by sequencing of the ten coding exons of CTNS. Comprehensive mutation analysis verified that none of the 12 patients carried the common 57-kb deletion. We identified four previously reported nucleotide variations associated with cystinosis and five new variants: a 10-kb deletion, three missense variants, and a nucleotide substitution in a potential branch point site of intron 4. This study is the first molecular analysis of Turkish cystinosis patients and provides guidance for the molecular diagnosis of cystinosis in this population.


Subject(s)
Amino Acid Transport Systems, Neutral/genetics , Cystinosis/genetics , Mutation , Adolescent , Adult , Child , Cystinosis/complications , Cystinosis/epidemiology , DNA Mutational Analysis , Disease Progression , Exons , Failure to Thrive/genetics , Fanconi Syndrome/genetics , Female , Genetic Predisposition to Disease , Humans , Introns , Kidney Failure, Chronic/genetics , Male , Mutation, Missense , Phenotype , Point Mutation , Polydipsia/genetics , Polymerase Chain Reaction , Polyuria/genetics , Proteinuria/genetics , Renal Insufficiency/genetics , Sequence Deletion , Turkey/epidemiology , Young Adult
20.
Pituitary ; 15 Suppl 1: S1-5, 2012 Dec.
Article in English | MEDLINE | ID: mdl-20401697

ABSTRACT

A 3-year 5-month-old boy was seen for second opinion regarding polydipsia and polyuria. Previously, a diagnosis of primary polydipsia was made after normal urine concentration after overnight water deprivation testing. The boy's father, paternal grandfather, and paternal aunt had diabetes insipidus treated with desmopressin acetate. Based on this young boy's symptoms, ability to concentrate urine after informal overnight water deprivation, and family history of diabetes insipidus, we performed AVP gene mutation testing. Analysis of the AVP gene revealed a novel mutation G54E that changes a normal glycine to glutamic acid, caused by a guanine to adenine change at nucleotide g.1537 (exon 2) of the AVP gene. Commonly, patients with familial neurohypophyseal diabetes insipidus (FNHDI) present within the first 6 years of life with progressively worsening polyuria and compensatory polydipsia. Since these patients have progressive loss of arginine vasopressin (AVP), they may initially respond normally to water deprivation testing and have normal pituitary findings on brain MRI. Genetic testing may be helpful in these patients, as well as preemptively diagnosing those with a mutation, thereby avoiding unnecessary surveillance of those unaffected.


Subject(s)
Diabetes Insipidus, Neurogenic/diagnosis , Diabetes Insipidus, Neurogenic/genetics , Polydipsia/diagnosis , Polydipsia/genetics , Polyuria/diagnosis , Polyuria/genetics , Child, Preschool , Humans , Male , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL