51 results on '"Dominique Loffing-Cueni"'
Search Results
2. Antiaging hormone Klotho derived from renal distal-convolution regulates calcium but not phosphate homeostasis
- Author
-
Ganesh Pathare, Klaudia Kopper, Adisa Trnjanin, Dominique Loffing-Cueni, Agnieszka Wengi, and Johannes Loffing
- Subjects
Physiology - Abstract
Soluble-Klotho (sKl) is the shed ectodomain of the transmembrane protein-Klotho (mKl) that exhibits pleiotropic actions, including lifespan extension, mineral metabolism, slowing-down kidney diseases and cardioprotection. The sKl is derived from the kidneys, but what type/s of renal cells secrete it is unknown. Secondly the respective roles of mKl versus sKl in regulating mineral metabolism is unclear due to the lack of appropriate in vivo models. Here, using scRNA-seq of renal distal-convolution (DC) cells, we found an unexpected pattern revealing that Klotho transcripts ( Kl) are moderately expressed in overall distal convoluted tubule (DCT), but highly enriched in the end of DCT and in connecting tubule (CNT). Immunohistochemistry further confirmed this pattern for mKl protein as well. Next, Kl was knocked-out only in renal DC to check if it affects sKl production. Interestingly, deleting Kl in the DCT and late-DCT+CNT in mice showed ~20% and ~80% reduction in sKl levels, respectively. Expectedly, knocking-out Kl along the entire DC in mice (Kl-KODC) abolished sKl levels. Furthermore, we found that compared to control mice, Kl-KODC mice exhibited reduced renal TRPV5-Ca2+ channel expression, profound calciuria, and loss of bone mineral density. The RNA-seq of automated-sorted DC cells from Kl-KODC mice revealed enhanced caveolae-mediated endocytosis of TRPV5. On the other hand, Kl-KODC mice had normal phosphate metabolism as confirmed by unchanged serum FGF23, serum phosphate, urinary phosphate excretion, and renal NaPi-IIa expression. Our findings reveal that a small population of renal DC cells accounts for the sKl levels. The lack of sKl may leads to disturbed Ca2+ homeostasis and bone loss without affecting phosphate balance in mice. This is the full abstract presented at the American Physiology Summit 2023 meeting and is only available in HTML format. There are no additional versions or additional content available for this abstract. Physiology was not involved in the peer review process.
- Published
- 2023
3. Deletion of the transcription factor Prox-1 specifically in the renal distal convoluted tubule causes hypomagnesemia via reduced expression of TRPM6 and NCC
- Author
-
Johannes Loffing, Dominique Loffing-Cueni, Denise V. Kratschmar, Christina Schnoz, Sandra Moser, Alex Odermatt, University of Zurich, and Loffing, Johannes
- Subjects
0301 basic medicine ,Renal distal convoluted tubule (DCT) ,10017 Institute of Anatomy ,Physiology ,Clinical Biochemistry ,TRPM Cation Channels ,610 Medicine & health ,1308 Clinical Biochemistry ,Kidney ,03 medical and health sciences ,Mice ,2737 Physiology (medical) ,0302 clinical medicine ,Physiology (medical) ,TRPM6 ,medicine ,Transcriptional regulation ,Animals ,Magnesium ,Solute Carrier Family 12, Member 3 ,Distal convoluted tubule ,Kidney Tubules, Distal ,Transcription factor ,Solute Carrier Family 12, Member 1 ,Homeodomain Proteins ,Aquaporin 2 ,NaCl cotransporter (NCC) ,Chemistry ,Kinase ,Tumor Suppressor Proteins ,DCT adaptation ,Sodium ,1314 Physiology ,Transient receptor potential cation channel subfamily M member 6 (TRPM6) ,Cell biology ,body regions ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Transcription factor Prox-1 ,Potassium ,570 Life sciences ,biology ,Cotransporter ,030217 neurology & neurosurgery ,Homeostasis ,Ion Channels, Receptors and Transporters ,Gene Deletion - Abstract
The renal distal convoluted tubule (DCT) is critical for the fine-tuning of urinary ion excretion and the control of blood pressure. Ion transport along the DCT is tightly controlled by posttranscriptional mechanisms including a complex interplay of kinases, phosphatases, and ubiquitin ligases. Previous work identified the transcription factor Prox-1 as a gene significantly enriched in the DCT of adult mice. To test if Prox-1 contributes to the transcriptional regulation of DCT function and structure, we developed a novel mouse model (NCCcre:Prox-1flox/flox) for an inducible deletion of Prox-1 specifically in the DCT. The deletion of Prox-1 had no obvious impact on DCT structure and growth independent whether the deletion was achieved in newborn or adult mice. Furthermore, DCT-specific Prox-1 deficiency did not alter DCT-proliferation in response to loop diuretic treatment. Likewise, the DCT-specific deletion of Prox-1 did not cause other gross phenotypic abnormalities. Body weight, urinary volume, Na+ and K+ excretion as well as plasma Na+, K+, and aldosterone levels were similar in Prox-1DCTKO and Prox-1DCTCtrl mice. However, Prox-1DCTKO mice exhibited a significant hypomagnesemia with a profound downregulation of the DCT-specific apical Mg2+ channel TRPM6 and the NaCl cotransporter (NCC) at both mRNA and protein levels. The expression of other proteins involved in distal tubule Mg2+ and Na+ handling was not affected. Thus, Prox-1 is a DCT-enriched transcription factor that does not control DCT growth but contributes to the molecular control of DCT-dependent Mg2+ homeostasis in the adult kidney.
- Published
- 2020
4. A five amino acids deletion in NKCC2 of C57BL/6 mice affects analysis of NKCC2 phosphorylation but does not impact kidney function
- Author
-
Viktoria Fisi, Agnieszka Wengi, Dominique Loffing-Cueni, Robert A. Fenton, Marta Mariniello, James A. McCormick, Sandra Moser, Yuya Sugano, Johannes Loffing, Lena Lindtoft Rosenbaek, University of Zurich, and Loffing, Johannes
- Subjects
0301 basic medicine ,C57BL/6 ,10017 Institute of Anatomy ,Physiology ,PROTEIN ,030204 cardiovascular system & hematology ,10052 Institute of Physiology ,NKCC2 ,Mice ,0302 clinical medicine ,Regular Paper ,Amino Acids ,Solute Carrier Family 12, Member 1 ,chemistry.chemical_classification ,Kidney ,biology ,STRAIN DIFFERENCES ,Chemistry ,phosphorylation ,NACL COTRANSPORTER ,SALT ,SPAK ,strain differences ,K-CL COTRANSPORTER ,Amino acid ,medicine.anatomical_structure ,Phosphorylation ,THICK ASCENDING LIMB ,Antibody ,kidney ,Sodium-Potassium-Chloride Symporters ,DISTAL CONVOLUTED TUBULE ,610 Medicine & health ,Sequence alignment ,03 medical and health sciences ,medicine ,Animals ,ion homeostasis ,Renal Physiology ,DEOXYCORTICOSTERONE ACETATE ,urogenital system ,1314 Physiology ,biology.organism_classification ,Molecular biology ,Mice, Inbred C57BL ,SODIUM-CHLORIDE COTRANSPORTER ,030104 developmental biology ,Ion homeostasis ,biology.protein ,570 Life sciences ,Cotransporter - Abstract
Aim: The phosphorylation level of the furosemide-sensitive Na +-K +-2Cl − cotransporter (NKCC2) in the thick ascending limb (TAL) is used as a surrogate marker for NKCC2 activation and TAL function. However, in mice, analyses of NKCC2 phosphorylation with antibodies against phosphorylated threonines 96 and 101 (anti-pT96/pT101) give inconsistent results. We aimed (a) to elucidate these inconsistencies and (b) to develop a phosphoform-specific antibody that ensures reliable detection of NKCC2 phosphorylation in mice. Methods: Genetic information, molecular biology, biochemical techniques and mouse phenotyping was used to study NKCC2 and kidney function in two commonly used mouse strains (ie 129Sv and in C57BL/6 mice). Moreover, a new phosphoform-specific mouse NKCC2 antibody was developed and characterized. Results: Amino acids sequence alignment revealed that C57BL/6 mice have a strain-specific five amino acids deletion (ΔF97-T101) in NKCC2 that diminishes the detection of NKCC2 phosphorylation with previously developed pT96/pT101 NKCC2 antibodies. Instead, the antibodies cross-react with the phosphorylated thiazide-sensitive NaCl cotransporter (NCC), which can obscure interpretation of results. Interestingly, the deletion in NKCC2 does not impact on kidney function and/or expression of renal ion transport proteins as indicated by the analysis of the F2 generation of crossbred 129Sv and C57BL/6 mice. A newly developed pT96 NKCC2 antibody detects pNKCC2 in both mouse strains and shows no cross-reactivity with phosphorylated NCC. Conclusion: Our work reveals a hitherto unappreciated, but essential, strain difference in the amino acids sequence of mouse NKCC2 that needs to be considered when analysing NKCC2 phosphorylation in mice. The new pNKCC2 antibody circumvents this technical caveat.
- Published
- 2021
5. Plasma Potassium Determines NCC Abundance in Adult Kidney-Specific γENaC Knockout
- Author
-
Romain Perrier, Emilie Boscardin, Marc Maillard, Johannes Loffing, Robert Koesters, Dominique Loffing-Cueni, Chloé Sergi, Edith Hummler, and Bernard C. Rossier
- Subjects
0301 basic medicine ,Epithelial sodium channel ,medicine.medical_specialty ,Kidney ,Hyperkalemia ,urogenital system ,Chemistry ,Sodium ,Potassium ,chemistry.chemical_element ,Pseudohypoaldosteronism ,General Medicine ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Nephrology ,Internal medicine ,Knockout mouse ,medicine ,Distal convoluted tubule ,medicine.symptom - Abstract
The amiloride-sensitive epithelial sodium channel (ENaC) and the thiazide-sensitive sodium chloride cotransporter (NCC) are key regulators of sodium and potassium and colocalize in the late distal convoluted tubule of the kidney. Loss of the αENaC subunit leads to a perinatal lethal phenotype characterized by sodium loss and hyperkalemia resembling the human syndrome pseudohypoaldosteronism type 1 (PHA-I). In adulthood, inducible nephron-specific deletion of αENaC in mice mimics the lethal phenotype observed in neonates, and as in humans, this phenotype is prevented by a high sodium (HNa+)/low potassium (LK+) rescue diet. Rescue reflects activation of NCC, which is suppressed at baseline by elevated plasma potassium concentration. In this study, we investigated the role of the γENaC subunit in the PHA-I phenotype. Nephron-specific γENaC knockout mice also presented with salt-wasting syndrome and severe hyperkalemia. Unlike mice lacking αENaC or βEΝaC, an HNa+/LK+ diet did not normalize plasma potassium (K+) concentration or increase NCC activation. However, when K+ was eliminated from the diet at the time that γENaC was deleted, plasma K+ concentration and NCC activity remained normal, and progressive weight loss was prevented. Loss of the late distal convoluted tubule, as well as overall reduced βENaC subunit expression, may be responsible for the more severe hyperkalemia. We conclude that plasma K+ concentration becomes the determining and limiting factor in regulating NCC activity, regardless of Na+ balance in γENaC-deficient mice.
- Published
- 2018
6. Protein Phosphatase 1 Inhibitor-1 Mediates the cAMP-Dependent Stimulation of the Renal NaCl Cotransporter
- Author
-
Johannes Loffing, Dominique Loffing-Cueni, Nourdine Faresse, Agnieszka Wengi, Sandra Moser, Jan Czogalla, Lena Lindtoft Rosenbaek, Joana Raquel Martins, David Penton, Robert A. Fenton, Fritz Rigendinger, University of Zurich, and Loffing, Johannes
- Subjects
distal tubule ,IBMX ,10017 Institute of Anatomy ,Phosphatase ,Immunoblotting ,610 Medicine & health ,macromolecular substances ,In Vitro Techniques ,Sodium Chloride ,environment and public health ,Cell & Transport Physiology ,chemistry.chemical_compound ,Mice ,Protein Phosphatase 1 ,medicine ,Animals ,Humans ,Solute Carrier Family 12, Member 3 ,cyclic AMP ,Distal convoluted tubule ,Phosphorylation ,Kidney Tubules, Distal ,Mice, Knockout ,Analysis of Variance ,2727 Nephrology ,Forskolin ,Chemistry ,Kinase ,urogenital system ,Colforsin ,Proteins ,Protein phosphatase 1 ,Biological Transport ,General Medicine ,Na transport ,Cell biology ,enzymes and coenzymes (carbohydrates) ,medicine.anatomical_structure ,Basic Research ,Nephrology ,embryonic structures ,570 Life sciences ,biology ,NaCl cotransporter ,Signal transduction ,signaling ,Signal Transduction - Abstract
Background A number of cAMP-elevating hormones stimulate phosphorylation (and hence activity) of the NaCl cotransporter (NCC) in the distal convoluted tubule (DCT). Evidence suggests that protein phosphatase 1 (PP1) and other protein phosphatases modulate NCC phosphorylation, but little is known about PP1’s role and the mechanism regulating its function in the DCT. Methods We used ex vivo mouse kidney preparations to test whether a DCT-enriched inhibitor of PP1, protein phosphatase 1 inhibitor–1 (I1), mediates cAMP’s effects on NCC, and conducted yeast two-hybrid and coimmunoprecipitation experiments in NCC-expressing MDCK cells to explore protein interactions. Results Treating isolated DCTs with forskolin and IBMX increased NCC phosphorylation via a protein kinase A (PKA)–dependent pathway. Ex vivo incubation of mouse kidney slices with isoproterenol, norepinephrine, and parathyroid hormone similarly increased NCC phosphorylation. The cAMP-induced stimulation of NCC phosphorylation strongly correlated with the phosphorylation of I1 at its PKA consensus phosphorylation site (a threonine residue in position 35). We also found an interaction between NCC and the I1-target PP1. Moreover, PP1 dephosphorylated NCC in vitro , and the PP1 inhibitor calyculin A increased NCC phosphorylation. Studies in kidney slices and isolated perfused kidneys of control and I1-KO mice demonstrated that I1 participates in the cAMP-induced stimulation of NCC. Conclusions Our data suggest a complete signal transduction pathway by which cAMP increases NCC phosphorylation via a PKA-dependent phosphorylation of I1 and subsequent inhibition of PP1. This pathway might be relevant for the physiologic regulation of renal sodium handling by cAMP-elevating hormones, and may contribute to salt-sensitive hypertension in patients with endocrine disorders or sympathetic hyperactivity.
- Published
- 2019
7. Gitelman’s Syndrome: characterization of a novel c.1181G>A point mutation and functional classification of the known mutations
- Author
-
Verdiana Ravarotto, Michèle Heidemeyer, Lorenzo A. Calò, Johannes Loffing, Elisa Pagnin, Gian Paolo Rossi, Dominique Loffing-Cueni, University of Zurich, and Rossi, Gian Paolo
- Subjects
Adult ,0301 basic medicine ,10017 Institute of Anatomy ,Protein Conformation ,Physiology ,DNA Mutational Analysis ,Mutant ,Xenopus ,Context (language use) ,610 Medicine & health ,Biology ,2705 Cardiology and Cardiovascular Medicine ,03 medical and health sciences ,Complementary DNA ,parasitic diseases ,Internal Medicine ,Humans ,Point Mutation ,Missense mutation ,Solute Carrier Family 12, Member 3 ,urogenital system ,Siblings ,Point mutation ,Transfection ,1314 Physiology ,biology.organism_classification ,Molecular biology ,Pedigree ,Blot ,HEK293 Cells ,030104 developmental biology ,2724 Internal Medicine ,embryonic structures ,570 Life sciences ,biology ,Female ,Cardiology and Cardiovascular Medicine ,Gitelman Syndrome - Abstract
We have investigated the mechanisms by which a novel missense point mutation (c.1181G>A) found in two sisters causes Gitelman’s syndrome by impairing the sodium chloride co-transporter (NCC, encoded by SLC12A3 gene) function. The cDNA and in vitro transcribed mRNA of either wild-type or mutated SLC12A3 were transfected into HEK293 cells and injected into Xenopus laevis oocytes, respectively. The expression, maturation, trafficking, and function of the mutated and wild-type NCC were assessed by Western blotting, immunohistochemistry and 22Na+ uptake studies. By immunoblotting of lysates from HEK293 cells and oocytes expressing wild-type NCC, two NCC-related bands of approximately 130 kDa and 115 kDa, corresponding to fully and core-glycosylated NCC, respectively, were identified. In contrast, the mutant NCC only showed a single band of approximately 115 kDa, indicating impaired maturation of the protein. Moreover, oocytes injected with wild-type NCC showed thiazide-sensitive 22Na+ uptake, which was absent in those injected with the mutant NCC. The novel mutation was discussed in the context of the functionally characterized NCC mutations causing Gitelman’s syndrome, which fit into five classes. In conclusion, the functional characterization of this novel Gly394Asp NCC and its localization on the NCC structure, alongside that of previously known mutations causing Gitelman’s syndrome, may provide novel information on the function of the different domains of the human NCC.
- Published
- 2018
8. The β‐adrenergic stimulation of the renal NaCl cotransporter is mediated via a cAMP‐dependent activation of protein phosphatase 1 inhibitor 1
- Author
-
Agnieszka Wengi, Nourdine Faresse, Johannes Loffing, Jan Czogalla, Sandra Moser, Robert A. Fenton, Lena Lindtoft Rosenbaek, David Penton Ribas, and Dominique Loffing-Cueni
- Subjects
medicine.medical_specialty ,Endocrinology ,Chemistry ,Internal medicine ,Genetics ,medicine ,β adrenergic stimulation ,Protein phosphatase 1 ,Cotransporter ,Molecular Biology ,Biochemistry ,Biotechnology - Published
- 2018
9. ENaC activity in collecting ducts modulates NCC in cirrhotic mice
- Author
-
David Mordasini, Bruno Vogt, Michel Burnier, Johannes Loffing, Rohrbach Beatrice, Dominique Loffing-Cueni, Geneviève Escher, Marc Maillard, Edith Hummler, University of Zurich, and Mordasini, David
- Subjects
Epithelial sodium channel ,medicine.medical_specialty ,Cirrhosis ,10017 Institute of Anatomy ,Physiology ,Sodium ,Clinical Biochemistry ,chemistry.chemical_element ,610 Medicine & health ,1308 Clinical Biochemistry ,030204 cardiovascular system & hematology ,Liver Cirrhosis, Experimental ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,2737 Physiology (medical) ,0302 clinical medicine ,Downregulation and upregulation ,Physiology (medical) ,Internal medicine ,Ascites ,medicine ,Animals ,Aldosterone/blood ,Bile Ducts/metabolism ,Epithelial Sodium Channels/genetics ,Epithelial Sodium Channels/metabolism ,Liver Cirrhosis, Experimental/metabolism ,Potassium/urine ,Sodium/urine ,Sodium Chloride Symporters/metabolism ,Epithelial Sodium Channels ,Receptor ,Aldosterone ,030304 developmental biology ,0303 health sciences ,Kidney ,urogenital system ,1314 Physiology ,medicine.disease ,Sodium Chloride Symporters ,3. Good health ,medicine.anatomical_structure ,Endocrinology ,chemistry ,Potassium ,570 Life sciences ,biology ,Bile Ducts ,medicine.symptom - Abstract
Cirrhosis is a frequent and severe disease, complicated by renal sodium retention leading to ascites and oedema. A better understanding of the complex mechanisms responsible for renal sodium handling could improve clinical management of sodium retention. Our aim was to determine the importance of the amiloride-sensitive epithelial sodium channel (ENaC) in collecting ducts in compensate and decompensate cirrhosis. Bile duct ligation was performed in control mice (CTL) and collecting duct-specific αENaC knockout (KO) mice, and ascites development, aldosterone plasma concentration, urinary sodium/potassium ratio and sodium transporter expression were compared. Disruption of ENaC in collecting ducts (CDs) did not alter ascites development, urinary sodium/potassium ratio, plasma aldosterone concentrations or Na,K-ATPase abundance in CCDs. Total αENaC abundance in whole kidney increased in cirrhotic mice of both genotypes and cleaved forms of α and γ ENaC increased only in ascitic mice of both genotypes. The sodium chloride cotransporter (NCC) abundance was lower in non-ascitic KO, compared to non-ascitic CTL, and increased when ascites appeared. In ascitic mice, the lack of αENaC in CDs induced an upregulation of total ENaC and NCC and correlated with the cleavage of ENaC subunits. This revealed compensatory mechanisms which could also take place when treating the patients with diuretics. These compensatory mechanisms should be considered for future development of therapeutic strategies.
- Published
- 2015
10. Comparative transcriptomic analysis identifies evolutionarily conserved gene products in the vertebrate renal distal convoluted tubule
- Author
-
Johannes Loffing, Dominique Loffing-Cueni, Chiara Cianciolo Cosentino, Yuya Sugano, Stephan C.F. Neuhauss, University of Zurich, and Loffing, Johannes
- Subjects
0301 basic medicine ,Dopamine and cAMP-Regulated Phosphoprotein 32 ,10017 Institute of Anatomy ,Physiology ,Receptors, Drug ,Clinical Biochemistry ,610 Medicine & health ,1308 Clinical Biochemistry ,Transcriptome ,Evolution, Molecular ,03 medical and health sciences ,Mice ,2737 Physiology (medical) ,Physiology (medical) ,Gene expression ,medicine ,Animals ,Distal convoluted tubule ,Kidney Tubules, Distal ,Zebrafish ,Conserved Sequence ,Genetics ,Gene knockdown ,biology ,1314 Physiology ,Zebrafish Proteins ,biology.organism_classification ,Sodium Chloride Symporters ,10124 Institute of Molecular Life Sciences ,Pronephros ,Cell biology ,Gene expression profiling ,030104 developmental biology ,medicine.anatomical_structure ,embryonic structures ,570 Life sciences ,mCherry - Abstract
Understanding the molecular basis of the complex regulatory networks controlling renal ion transports is of major physiological and clinical importance. In this study, we aimed to identify evolutionarily conserved critical players in the function of the renal distal convoluted tubule (DCT) by a comparative transcriptomic approach. We generated a transgenic zebrafish line with expression of the red fluorescent mCherry protein under the control of the zebrafish DCT-specific promoter of the thiazide-sensitive NaCl cotransporter (NCC). The mCherry expression was then used to isolate from the zebrafish mesonephric kidneys the distal late (DL) segments, the equivalent of the mammalian DCT, for subsequent RNA-seq analysis. We next compared this zebrafish DL transcriptome to the previously established mouse DCT transcriptome and identified a subset of gene products significantly enriched in both the teleost DL and the mammalian DCT, including SLCs and nuclear transcription factors. Surprisingly, several of the previously described regulators of NCC (e.g., SPAK, KLHL3, ppp1r1a) in the mouse were not found enriched in the zebrafish DL. Nevertheless, the zebrafish DL expressed enriched levels of related homologues. Functional knockdown of one of these genes, ppp1r1b, reduced the phosphorylation of NCC in the zebrafish pronephros, similar to what was seen previously in knockout mice for its homologue, Ppp1r1a. The present work is the first report on global gene expression profiling in a specific nephron portion of the zebrafish kidney, an increasingly used model system for kidney research. Our study suggests that comparative analysis of gene expression between phylogenetically distant species may be an effective approach to identify novel regulators of renal function.
- Published
- 2017
11. Severe hyperkalemia is rescued by low-potassium diet in renal βENaC-deficient mice
- Author
-
Emilie Boscardin, Johannes Loffing, Dominique Loffing-Cueni, Edith Hummler, Bernard C. Rossier, Chloé Sergi, Romain Perrier, Marc Maillard, Robert Koesters, University of Zurich, and Hummler, Edith
- Subjects
0301 basic medicine ,Epithelial sodium channel ,medicine.medical_specialty ,Hyperkalemia ,10017 Institute of Anatomy ,Physiology ,Sodium ,Clinical Biochemistry ,030232 urology & nephrology ,chemistry.chemical_element ,Mice, Transgenic ,610 Medicine & health ,1308 Clinical Biochemistry ,Kidney ,03 medical and health sciences ,0302 clinical medicine ,2737 Physiology (medical) ,Downregulation and upregulation ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Potassium Channels, Inwardly Rectifying ,Epithelial Sodium Channels ,Protein kinase A ,urogenital system ,Chemistry ,Pseudohypoaldosteronism ,Kidney metabolism ,Metabolic acidosis ,Nephrons ,1314 Physiology ,Diet, Sodium-Restricted ,medicine.disease ,Phenotype ,030104 developmental biology ,Endocrinology ,Epithelial Sodium Channels/metabolism ,Hyperkalemia/metabolism ,Kidney/metabolism ,Nephrons/metabolism ,Potassium/metabolism ,Potassium Channels, Inwardly Rectifying/metabolism ,Sodium/metabolism ,Pseudohypoaldosteronism type 1 ,STE20/SPS1-related proline-alanine-rich protein kinase ,Thiazide-sensitive Na+/Cl− co-transporter ,Potassium ,570 Life sciences ,biology ,medicine.symptom - Abstract
In adulthood, an induced nephron-specific deficiency of αENaC (Scnn1a) resulted in pseudohypoaldosteronism type 1 (PHA-1) with sodium loss, hyperkalemia, and metabolic acidosis that is rescued through high-sodium/low-potassium (HNa + /LK + ) diet. In the present study, we addressed whether renal βENaC expression is required for sodium and potassium balance or can be compensated by remaining (α and γ) ENaC subunits using adult nephron-specific knockout (Scnn1b Pax8/LC1 ) mice. Upon induction, these mice present a severe PHA-1 phenotype with weight loss, hyperkalemia, and dehydration, but unlike the Scnn1a Pax8/LC1 mice without persistent salt wasting. This is followed by a marked downregulation of STE20/SPS1-related proline-alanine-rich protein kinase (SPAK) and Na + /Cl - co-transporter (NCC) protein expression and activity. Most of the experimental Scnn1b Pax8/LC1 mice survived with a HNa + /LK + diet that partly normalized NCC phosphorylation, but not total NCC expression. Since salt loss was minor, we applied a standard-sodium/LK + diet that efficiently rescued these mice resulting in normokalemia and normalization of NCC phosphorylation, but not total NCC expression. A further switch to LNa + /standard-K + diet induced again a severe PHA-1-like phenotype, but with only transient salt wasting indicating that low-K + intake is critical to decrease hyperkalemia in a NCC-dependent manner. In conclusion, while the βENaC subunit plays only a minor role in sodium balance, severe hyperkalemia results in downregulation of NCC expression and activity. Our data demonstrate the importance to primarily correct the hyperkalemia with a low-potassium diet that normalizes NCC activity.
- Published
- 2017
12. Plasma Potassium Determines NCC Abundance in Adult Kidney-Specific
- Author
-
Emilie, Boscardin, Romain, Perrier, Chloé, Sergi, Marc P, Maillard, Johannes, Loffing, Dominique, Loffing-Cueni, Robert, Koesters, Bernard C, Rossier, and Edith, Hummler
- Subjects
Mice, Knockout ,urogenital system ,Pseudohypoaldosteronism ,Potassium, Dietary ,Sodium, Dietary ,Nephrons ,Mice ,Basic Research ,Dietary Supplements ,Potassium ,Animals ,Hyperkalemia ,Polystyrenes ,Solute Carrier Family 12, Member 3 ,Epithelial Sodium Channels ,Chelating Agents - Abstract
The amiloride-sensitive epithelial sodium channel (ENaC) and the thiazide-sensitive sodium chloride cotransporter (NCC) are key regulators of sodium and potassium and colocalize in the late distal convoluted tubule of the kidney. Loss of the αENaC subunit leads to a perinatal lethal phenotype characterized by sodium loss and hyperkalemia resembling the human syndrome pseudohypoaldosteronism type 1 (PHA-I). In adulthood, inducible nephron-specific deletion of αENaC in mice mimics the lethal phenotype observed in neonates, and as in humans, this phenotype is prevented by a high sodium (HNa(+))/low potassium (LK(+)) rescue diet. Rescue reflects activation of NCC, which is suppressed at baseline by elevated plasma potassium concentration. In this study, we investigated the role of the γENaC subunit in the PHA-I phenotype. Nephron-specific γENaC knockout mice also presented with salt-wasting syndrome and severe hyperkalemia. Unlike mice lacking αENaC or βΕΝaC, an HNa(+)/LK(+) diet did not normalize plasma potassium (K(+)) concentration or increase NCC activation. However, when K(+) was eliminated from the diet at the time that γENaC was deleted, plasma K(+) concentration and NCC activity remained normal, and progressive weight loss was prevented. Loss of the late distal convoluted tubule, as well as overall reduced βENaC subunit expression, may be responsible for the more severe hyperkalemia. We conclude that plasma K(+) concentration becomes the determining and limiting factor in regulating NCC activity, regardless of Na(+) balance in γENaC-deficient mice.
- Published
- 2017
13. Extracellular K(+) rapidly controls NaCl cotransporter phosphorylation in the native distal convoluted tubule by Cl(-) -dependent and independent mechanisms
- Author
-
Johannes Loffing, Dominique Loffing-Cueni, Nina Himmerkus, Olivier Staub, Frank Schweda, David Penton, Jan Czogalla, Monique Carrel, Markus Bleich, Renuga Devi Rajaram, Agnieszka Wengi, University of Zurich, and Loffing, Johannes
- Subjects
0301 basic medicine ,10017 Institute of Anatomy ,Chemistry ,Kinase ,Physiology ,urogenital system ,potassium ,sodium transport ,signal transduction ,610 Medicine & health ,1314 Physiology ,Cell biology ,Dephosphorylation ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Biochemistry ,Extracellular ,medicine ,Phosphorylation ,570 Life sciences ,biology ,Distal convoluted tubule ,Cotransporter ,Protein kinase A ,Ex vivo - Abstract
High dietary potassium (K(+) ) intake dephosphorylates and inactivates the NaCl cotransporter (NCC) in the renal distal convoluted tubule (DCT). Using several ex vivo models, we show that physiological changes in extracellular K(+) , similar to those occurring after a K(+) rich diet, are sufficient to promote a very rapid dephosphorylation of NCC in native DCT cells. Although the increase of NCC phosphorylation upon decreased extracellular K(+) appears to depend on cellular Cl(-) fluxes, the rapid NCC dephosphorylation in response to increased extracellular K(+) is not Cl(-) -dependent. The Cl(-) -dependent pathway involves the SPAK/OSR1 kinases, whereas the Cl(-) independent pathway may include additional signalling cascades. A high dietary potassium (K(+) ) intake causes a rapid dephosphorylation, and hence inactivation, of the thiazide-sensitive NaCl cotransporter (NCC) in the renal distal convoluted tubule (DCT). Based on experiments in heterologous expression systems, it was proposed that changes in extracellular K(+) concentration ([K(+) ]ex ) modulate NCC phosphorylation via a Cl(-) -dependent modulation of the with no lysine (K) kinases (WNK)-STE20/SPS-1-44 related proline-alanine-rich protein kinase (SPAK)/oxidative stress-related kinase (OSR1) kinase pathway. We used the isolated perfused mouse kidney technique and ex vivo preparations of mouse kidney slices to test the physiological relevance of this model on native DCT. We demonstrate that NCC phosphorylation inversely correlates with [K(+) ]ex , with the most prominent effects occurring around physiological plasma [K(+) ]. Cellular Cl(-) conductances and the kinases SPAK/OSR1 are involved in the phosphorylation of NCC under low [K(+) ]ex . However, NCC dephosphorylation triggered by high [K(+) ]ex is neither blocked by removing extracellular Cl(-) , nor by the Cl(-) channel blocker 4,4'-diisothiocyano-2,2'-stilbenedisulphonic acid. The response to [K(+) ]ex on a low extracellular chloride concentration is also independent of significant changes in SPAK/OSR1 phosphorylation. Thus, in the native DCT, [K(+) ]ex directly and rapidly controls NCC phosphorylation by Cl(-) -dependent and independent pathways that involve the kinases SPAK/OSR1 and a yet unidentified additional signalling mechanism.
- Published
- 2016
14. Immunofluorescent localization of the Rab-GAP protein TBC1D4 (AS160) in mouse kidney
- Author
-
Johannes Loffing, Dominique Loffing-Cueni, Nikolay Gresko, Marianna Di Chiara, Natascha Lier, University of Zurich, and Loffing, Johannes
- Subjects
Epithelial sodium channel ,medicine.medical_specialty ,Histology ,10017 Institute of Anatomy ,Fluorescent Antibody Technique ,610 Medicine & health ,Mice, Inbred Strains ,Nephron ,Biology ,Kidney ,2722 Histology ,1307 Cell Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,1312 Molecular Biology ,medicine ,Animals ,Intercalated Cell ,Distal convoluted tubule ,Kidney Tubules, Distal ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,urogenital system ,GTPase-Activating Proteins ,3607 Medical Laboratory Technology ,Cell Biology ,Connecting tubule ,Cell biology ,Medical Laboratory Technology ,Endocrinology ,medicine.anatomical_structure ,Aquaporin 2 ,10076 Center for Integrative Human Physiology ,Loop of Henle ,570 Life sciences ,biology ,030217 neurology & neurosurgery ,Homeostasis - Abstract
TBC1D4 (or AS160) was identified as a Rab-GTPase activating protein (Rab-GAP) that controls insulin-dependent trafficking of the glucose transporter GLUT4 in skeletal muscle cells and in adipocytes. Recent in vitro cell culture studies suggest that TBC1D4 may also regulate the intracellular trafficking of kidney proteins such as the vasopressin-dependent water channel AQP2, the aldosterone-regulated epithelial sodium channel ENaC, and the Na(+)-K(+)-ATPase. To study the possible role of TBC1D4 in the kidney in vivo, we raised a rabbit polyclonal antibody against TBC1D4 to be used for immunoblotting and immunohistochemical studies. In immunoblots on mouse kidney homogenates, the antibody recognizes specific bands at the expected size of 160 kDa and at lower molecular weights, which are absent in kidneys of TBC1D4 deficient mice. Using a variety of nephron-segment-specific marker proteins, immunohistochemistry reveals TBC1D4 in the cytoplasm of the parietal epithelial cells of Bowman's capsule, the thin and thick limbs of Henle's loop, the distal convoluted tubule, the connecting tubule, and the collecting duct. In the latter, both principal as well as intercalated cells are TBC1D4-positive. Thus, with the exception of the proximal tubule, TBC1D4 is highly expressed along the nephron and the collecting duct, where it may interfere with the intracellular trafficking of many renal transport proteins including AQP2, ENaC and Na(+)-K(+)-ATPase. Hence, TBC1D4 may play an important role for the control of renal ion and water handling and hence for the control of extracellular fluid homeostasis.
- Published
- 2012
- Full Text
- View/download PDF
15. Programmed death 1 protects from fatal circulatory failure during systemic virus infection of mice
- Author
-
Annette Oxenius, Johannes Vogel, Thomas Braunschweiler, Burkhard Ludewig, Helge Frebel, Reto A. Schuepbach, Michael O. Kurrer, Veronika Nindl, Kirsten Richter, Dominique Loffing-Cueni, Carsten A. Wagner, University of Zurich, and Oxenius, Annette
- Subjects
10017 Institute of Anatomy ,Programmed Cell Death 1 Receptor ,CD8-Positive T-Lymphocytes ,B7-H1 Antigen ,Pathogenesis ,Mice ,0302 clinical medicine ,Immunopathology ,Immunology and Allergy ,Cytotoxic T cell ,Mice, Knockout ,0303 health sciences ,hemic and immune systems ,Shock ,10081 Institute of Veterinary Physiology ,3. Good health ,medicine.anatomical_structure ,2723 Immunology and Allergy ,10023 Institute of Intensive Care Medicine ,Hypotension ,Signal Transduction ,Pore Forming Cytotoxic Proteins ,T cell ,Immunology ,610 Medicine & health ,chemical and pharmacologic phenomena ,Mice, Transgenic ,Pulmonary Edema ,Lymphocytic Choriomeningitis ,Biology ,Lymphocytic choriomeningitis ,Article ,Virus ,Capillary Permeability ,03 medical and health sciences ,medicine ,Animals ,Histocompatibility Antigen H-2D ,030304 developmental biology ,2403 Immunology ,H-2 Antigens ,medicine.disease ,Mice, Inbred C57BL ,Disease Models, Animal ,Cytolysis ,570 Life sciences ,biology ,Endothelium, Vascular ,CD8 ,030215 immunology - Abstract
The PD-1–PD-L1 pathway inhibits perforin-mediated killing of PD-L1+ vascular endothelial cells by CD8+ T cells, thereby limiting vascular damage during systemic LCMV infection., The inhibitory programmed death 1 (PD-1)–programmed death ligand 1 (PD-L1) pathway contributes to the functional down-regulation of T cell responses during persistent systemic and local virus infections. The blockade of PD-1–PD-L1–mediated inhibition is considered as a therapeutic approach to reinvigorate antiviral T cell responses. Yet previous studies reported that PD-L1–deficient mice develop fatal pathology during early systemic lymphocytic choriomeningitis virus (LCMV) infection, suggesting a host protective role of T cell down-regulation. As the exact mechanisms of pathology development remained unclear, we set out to delineate in detail the underlying pathogenesis. Mice deficient in PD-1–PD-L1 signaling or lacking PD-1 signaling in CD8 T cells succumbed to fatal CD8 T cell–mediated immunopathology early after systemic LCMV infection. In the absence of regulation via PD-1, CD8 T cells killed infected vascular endothelial cells via perforin-mediated cytolysis, thereby severely compromising vascular integrity. This resulted in systemic vascular leakage and a consequential collapse of the circulatory system. Our results indicate that the PD-1–PD-L1 pathway protects the vascular system from severe CD8 T cell–mediated damage during early systemic LCMV infection, highlighting a pivotal physiological role of T cell down-regulation and suggesting the potential development of immunopathological side effects when interfering with the PD-1–PD-L1 pathway during systemic virus infections.
- Published
- 2012
- Full Text
- View/download PDF
16. Adult nephron-specific MR-deficient mice develop a severe renal PHA-1 phenotype
- Author
-
Dominique Loffing-Cueni, Johannes Loffing, Simona Frateschi, Bernard C. Rossier, Robert Koesters, Petra Klusonova, Marc Maillard, Edith Hummler, Chloé Sergi, Jérémie Canonica, Alex Odermatt, University of Zurich, and Hummler, Edith
- Subjects
0301 basic medicine ,Epithelial sodium channel ,medicine.medical_specialty ,10017 Institute of Anatomy ,Hyperkalemia ,Physiology ,Pseudohypoaldosteronism ,Clinical Biochemistry ,610 Medicine & health ,Nephron ,1308 Clinical Biochemistry ,030204 cardiovascular system & hematology ,Biology ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,2737 Physiology (medical) ,0302 clinical medicine ,Mineralocorticoid receptor ,Receptors, Glucocorticoid ,Physiology (medical) ,Internal medicine ,Weight Loss ,medicine ,Animals ,Epithelial Sodium Channels ,Aldosterone ,Renal sodium reabsorption ,Sodium ,Metabolic acidosis ,Epithelial Cells ,1314 Physiology ,Nephrons ,medicine.disease ,Sodium Chloride Symporters ,Aldosterone/blood ,Epithelial Cells/metabolism ,Epithelial Sodium Channels/genetics ,Epithelial Sodium Channels/metabolism ,Gene Deletion ,Nephrons/metabolism ,Phenotype ,Potassium/blood ,Potassium/urine ,Pseudohypoaldosteronism/genetics ,Pseudohypoaldosteronism/metabolism ,Pseudohypoaldosteronism/pathology ,Receptors, Glucocorticoid/genetics ,Receptors, Glucocorticoid/metabolism ,Receptors, Mineralocorticoid/deficiency ,Receptors, Mineralocorticoid/genetics ,Receptors, Mineralocorticoid/metabolism ,Sodium/blood ,Sodium/urine ,Sodium Chloride Symporters/genetics ,Sodium Chloride Symporters/metabolism ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Receptors, Mineralocorticoid ,chemistry ,Potassium ,570 Life sciences ,biology ,medicine.symptom - Abstract
Aldosterone is the main mineralocorticoid hormone controlling sodium balance, fluid homeostasis, and blood pressure by regulating sodium reabsorption in the aldosterone-sensitive distal nephron (ASDN). Germline loss-of-function mutations of the mineralocorticoid receptor (MR) in humans and in mice lead to the "renal" form of type 1 pseudohypoaldosteronism (PHA-1), a case of aldosterone resistance characterized by salt wasting, dehydration, failure to thrive, hyperkalemia, and metabolic acidosis. To investigate the importance of MR in adult epithelial cells, we generated nephron-specific MR knockout mice (MR(Pax8/LC1)) using a doxycycline-inducible system. Under standard diet, MR(Pax8/LC1) mice exhibit inability to gain weight and significant weight loss compared to control mice. Interestingly, despite failure to thrive, MR(Pax8/LC1) mice survive but develop a severe PHA-1 phenotype with higher urinary Na(+) levels, decreased plasma Na(+), hyperkalemia, and higher levels of plasma aldosterone. This phenotype further worsens and becomes lethal under a sodium-deficient diet. Na(+)/Cl(-) co-transporter (NCC) protein expression and its phosphorylated form are downregulated in the MR(Pax8/LC1) knockouts, as well as the αENaC protein expression level, whereas the expression of glucocorticoid receptor (GR) is increased. A diet rich in Na(+) and low in K(+) does not restore plasma aldosterone to control levels but is sufficient to restore body weight, plasma, and urinary electrolytes. In conclusion, MR deletion along the nephron fully recapitulates the features of severe human PHA-1. ENaC protein expression is dependent on MR activity. Suppression of NCC under hyperkalemia predominates in a hypovolemic state.
- Published
- 2015
17. The role of TBC1D1 and TBC1D4 in contraction-induced glucose uptake in mouse skeletal muscle
- Author
-
Hadi Al-Hasani, Johannes Loffing, Dominique Loffing-Cueni, C de Wendt, Alexandra Chadt, and HG Joost
- Subjects
medicine.medical_specialty ,Contraction (grammar) ,Endocrinology ,medicine.anatomical_structure ,Chemistry ,Endocrinology, Diabetes and Metabolism ,Internal medicine ,Glucose uptake ,medicine ,TBC1D1 ,Skeletal muscle - Published
- 2015
18. Rab-GAP TBC1D4 (AS160) is dispensable for the renal control of sodium and water homeostasis but regulates GLUT4 in mouse kidney
- Author
-
Johannes Loffing, Bob Glaudemans, Nourdine Faresse, Alex Odermatt, Marianna Di Chiara, Olivier Devuyst, Hadi Al-Hasani, Dominique Loffing-Cueni, University of Zurich, and Loffing, Johannes
- Subjects
2748 Urology ,Epithelial sodium channel ,Male ,medicine.medical_specialty ,10017 Institute of Anatomy ,Genotype ,Physiology ,Glucose uptake ,Green Fluorescent Proteins ,610 Medicine & health ,030204 cardiovascular system & hematology ,Biology ,Kidney ,10052 Institute of Physiology ,Renin-Angiotensin System ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,Insulin ,Na+/K+-ATPase ,Kidney Tubules, Distal ,Promoter Regions, Genetic ,Cells, Cultured ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Water transport ,Glucose Transporter Type 4 ,urogenital system ,GTPase-Activating Proteins ,Sodium ,Glucose transporter ,1314 Physiology ,Water-Electrolyte Balance ,medicine.anatomical_structure ,Endocrinology ,Glucose ,Parvalbumins ,Phenotype ,Gene Expression Regulation ,10076 Center for Integrative Human Physiology ,Loop of Henle ,570 Life sciences ,biology ,Cotransporter ,Homeostasis - Abstract
The Rab GTPase-activating protein TBC1D4 (AS160) controls trafficking of the glucose transporter GLUT4 in adipocytes and skeletal muscle cells. TBC1D4 is also highly abundant in the renal distal tubule, although its role in this tubule is so far unknown. In vitro studies suggest that it is involved in the regulation of renal transporters and channels such as the epithelial sodium channel (ENaC), aquaporin-2 (AQP2), and the Na+-K+-ATPase. To assess the physiological role of TBC1D4 in the kidney, wild-type (TBC1D4+/+) and TBC1D4-deficient (TBC1D4−/−) mice were studied. Unexpectedly, neither under standard nor under challenging conditions (low Na+/high K+, water restriction) did TBC1D4−/−mice show any difference in urinary Na+and K+excretion, urine osmolarity, plasma ion and aldosterone levels, and blood pressure compared with TBC1D4+/+mice. Also, immunoblotting did not reveal any change in the abundance of major renal sodium- and water-transporting proteins [Na-K-2Cl cotransporter (NKCC2) NKCC2, NaCl cotransporter (NCC), ENaC, AQP2, and the Na+-K+-ATPase]. However, the abundance of GLUT4, which colocalizes with TBC1D4 along the distal nephron of TBC1D4+/+mice, was lower in whole kidney lysates of TBC1D4−/−mice than in TBC1D4+/+mice. Likewise, primary thick ascending limb (TAL) cells isolated from TBC1D4−/−mice showed an increased basal glucose uptake and an abrogated insulin response compared with TAL cells from TBC1D4+/+mice. Thus, TBC1D4 is dispensable for the regulation of renal Na+and water transport, but may play a role for GLUT4-mediated basolateral glucose uptake in distal tubules. The latter may contribute to the known anaerobic glycolytic capacity of distal tubules during renal ischemia.
- Published
- 2015
19. Severe Salt-Losing Syndrome and Hyperkalemia Induced by Adult Nephron-Specific Knockout of the Epithelial Sodium Channel α-Subunit
- Author
-
Sumedha Malsure, Robert Koesters, Bernard C. Rossier, Dominique Loffing-Cueni, Romain Perrier, Marc Maillard, Mads V. Sorensen, Emilie Boscardin, Simona Frateschi, Johannes Loffing, Edith Hummler, and Chloé Sergi
- Subjects
0301 basic medicine ,Epithelial sodium channel ,medicine.medical_specialty ,Hyperkalemia ,Sodium ,Pseudohypoaldosteronism ,chemistry.chemical_element ,Nephron ,Severity of Illness Index ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Internal medicine ,medicine ,Animals ,Epithelial Sodium Channels ,Mice, Knockout ,Aldosterone ,Reabsorption ,urogenital system ,Sodium channel ,General Medicine ,Nephrons ,medicine.disease ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Basic Research ,chemistry ,Nephrology ,medicine.symptom - Abstract
Systemic pseudohypoaldosteronism type 1 (PHA-1) is a severe salt-losing syndrome caused by loss-of-function mutations of the amiloride-sensitive epithelial sodium channel (ENaC) and characterized by neonatal life-threatening hypovolemia and hyperkalemia. The very high plasma aldosterone levels detected under hypovolemic or hyperkalemic challenge can lead to increased or decreased sodium reabsorption, respectively, through the Na(+)/Cl(-) cotransporter (NCC). However, the role of ENaC deficiency remains incompletely defined, because constitutive inactivation of individual ENaC subunits is neonatally lethal in mice. We generated adult inducible nephron-specific αENaC-knockout mice (Scnn1a(Pax8/LC1)) that exhibit hyperkalemia and body weight loss when kept on a regular-salt diet, thus mimicking PHA-1. Compared with control mice fed a regular-salt diet, knockout mice fed a regular-salt diet exhibited downregulated expression and phosphorylation of NCC protein, despite high plasma aldosterone levels. In knockout mice fed a high-sodium and reduced-potassium diet (rescue diet), although plasma aldosterone levels remained significantly increased, NCC expression returned to control levels, and body weight, plasma and urinary electrolyte concentrations, and excretion normalized. Finally, shift to a regular diet after the rescue diet reinstated the symptoms of severe PHA-1 syndrome and significantly reduced NCC phosphorylation. In conclusion, lack of ENaC-mediated sodium transport along the nephron cannot be compensated for by other sodium channels and/or transporters, only by a high-sodium and reduced-potassium diet. We further conclude that hyperkalemia becomes the determining factor in regulating NCC activity, regardless of sodium loss, in the ENaC-mediated salt-losing PHA-1 phenotype.
- Published
- 2015
20. Replication of segment-specific and intercalated cells in the mouse renal collecting system
- Author
-
Dominique Loffing-Cueni, Johannes Loffing, Brigitte Kaissling, and Philipp Wehrli
- Subjects
Male ,Cell type ,Histology ,Cellular differentiation ,Fluorescent Antibody Technique ,Mice, Inbred Strains ,Sodium Chloride ,Biology ,Potassium Chloride ,Mice ,chemistry.chemical_compound ,Sodium Potassium Chloride Symporter Inhibitors ,Anion Exchange Protein 1, Erythrocyte ,Proliferating Cell Nuclear Antigen ,Animals ,Intercalated Cell ,Kidney Tubules, Collecting ,Molecular Biology ,Bumetanide ,Cell Proliferation ,Aquaporin 2 ,Cell growth ,Stem Cells ,Body Weight ,Cell Differentiation ,DNA ,Cell Biology ,Cell cycle ,Immunohistochemistry ,Molecular biology ,Proliferating cell nuclear antigen ,Proton-Translocating ATPases ,Medical Laboratory Technology ,Bromodeoxyuridine ,chemistry ,biology.protein ,Sodium-Potassium-Exchanging ATPase ,Stem cell - Abstract
The renal collecting system (CS) is composed of segment-specific (SS) and intercalated (IC) cells. The latter comprise at least two subtypes (type A and non-type A IC). The origin and maintenance of cellular heterogeneity in the CS is unclear. Among other hypotheses, it was proposed that one subtype of IC cells represents a stem cell population from which all cell types in the CS may arise. In the present study, we tested this stem cell hypothesis for the adult kidney by assessing DNA synthesis as a marker for cell replication. SS and IC cells were identified by their characteristic expressions of sodium- (epithelial sodium channel, Na–K-ATPase), water- (aquaporin-2) and acid/base- (H⁺-ATPase, anion exchanger AE1) transporting proteins. Immunostaining for bromodeoxyuridine (BrdU) and for the proliferating cell nuclear antigen (PCNA) was used to reveal DNA synthesis in CS epithelium. BrdU- and PCNA-immunostaining as well as mitotic figures were seen in all subtypes of CS cells. Dividing cells retained the cell-type specific expression of marker molecules. Treatment of mice with bumetanide combined with a high oral salt intake, which increases the tubular salt load in the CS, profoundly increased the DNA-synthesis rate in SS and non-type A IC cells, but reduced it in type A IC cells. Thus, our data show that DNA synthesis and cell replication occur in each cell lineage of the CS and in differentiated cells. The replication rate in each cell type can be differently modulated by functional stimulation. Independent proliferation of each cell lineage might contribute to maintain the cellular heterogeneity of the CS of the adult kidney and may also add to the adaptation of the CS to altered functional requirements.
- Published
- 2006
21. Pendrin Regulation in Mouse Kidney Primarily Is Chloride-Dependent
- Author
-
Régine Chambrey, Marion Vallet, Dominique Loffing-Cueni, Nicolas Picard, May Bloch-Faure, Peter S. Aronson, Dominique Eladari, Pierre Meneton, Georges Deschênes, Marinos Fysekidis, Johannes Loffing, and Sylvie Breton
- Subjects
Male ,Epithelial sodium channel ,medicine.medical_specialty ,medicine.drug_class ,Immunoblotting ,Mice, Transgenic ,Kidney ,Mice ,chemistry.chemical_compound ,Chlorides ,Internal medicine ,otorhinolaryngologic diseases ,medicine ,Animals ,Homeostasis ,Chloride-Bicarbonate Antiporters ,Kidney Tubules, Collecting ,Epithelial Sodium Channels ,Aldosterone ,Mice, Knockout ,biology ,Reabsorption ,Sodium ,General Medicine ,Pendrin ,Immunohistochemistry ,Connecting tubule ,Mice, Inbred C57BL ,Disease Models, Animal ,Hydrochlorothiazide ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Nephrology ,Mineralocorticoid ,Knockout mouse ,biology.protein - Abstract
Recent studies indicate that pendrin, an apical Cl-/HCO3- exchanger, mediates chloride reabsorption in the connecting tubule and the cortical collecting duct and therefore is involved in extracellular fluid volume regulation. The purpose of this study was to test whether pendrin is regulated in vivo primarily by factors that are associated with changes in renal chloride transport, by aldosterone, or by the combination of both determinants. For achievement of this goal, pendrin protein abundance was studied by semiquantitative immunoblotting in different mouse models with altered aldosterone secretion or tubular chloride transport, including NaCl loading, hydrochlorothiazide administration, NaCl co-transporter knockout mice, and mice with Liddle's mutation. The parallel regulation of the aldosterone-regulated epithelial sodium channel (ENaC) was examined as a control for biologic effects of aldosterone. Major changes in pendrin protein expression were found in experimental models that are associated with altered renal chloride transport, whereas no significant changes were detected in pendrin protein abundance in models with altered aldosterone secretion. Moreover, in response to hydrochlorothiazide administration, pendrin was downregulated despite a marked secondary hyperaldosteronism. In contrast, alpha-ENaC was markedly upregulated, and the molecular weight of a large fraction of gamma-ENaC subunits was shifted from 85 to 70 kD, consistent with previous results from rat models with elevated plasma aldosterone levels. These results suggest that factors that are associated with changes in distal chloride delivery govern pendrin expression in the connecting tubule and cortical collecting duct.
- Published
- 2006
22. Dietary Sodium Intake Regulates the Ubiquitin-Protein Ligase Nedd4-2 in the Renal Collecting System
- Author
-
Johannes Loffing, Nicole Siegrist, Daniel Sauter, Dominique Loffing-Cueni, Sandra Y. Flores, Pierre Meneton, Dorothée Daidié, and Olivier Staub
- Subjects
Male ,Epithelial sodium channel ,medicine.medical_specialty ,medicine.drug_class ,Nedd4 Ubiquitin Protein Ligases ,Ubiquitin-Protein Ligases ,macromolecular substances ,Nephron ,Biology ,Rats, Sprague-Dawley ,Mice ,Random Allocation ,chemistry.chemical_compound ,Species Specificity ,Internal medicine ,medicine ,Animals ,Tissue Distribution ,Distal convoluted tubule ,Kidney Tubules, Collecting ,Cells, Cultured ,Kidney ,Aldosterone ,Endosomal Sorting Complexes Required for Transport ,urogenital system ,Sodium, Dietary ,General Medicine ,respiratory system ,Connecting tubule ,Rats ,medicine.anatomical_structure ,Endocrinology ,Gene Expression Regulation ,chemistry ,Nephrology ,Mineralocorticoid ,hormones, hormone substitutes, and hormone antagonists ,Immunostaining - Abstract
The activity of the epithelial sodium (Na(+)) channel (ENaC) in the aldosterone-sensitive distal nephron (ASDN) needs to be tightly regulated to match urinary Na(+) excretion with dietary Na(+) intake. The ubiquitin-protein ligase Nedd4-2, which in vitro interacts with ENaC subunits and reduces ENaC cell surface abundance and activity by ubiquitylation of the channel, may participate in the control of ENaC. This study confirms in vivo by reverse-transcriptase-PCR that Nedd4-2 is expressed throughout the nephron and is detectable by immunoblotting in kidney extracts. By immunohistochemistry, Nedd4-2 was found to be strongly expressed in the ASDN, with low staining intensity in the late distal convoluted tubule and early connecting tubule (where apical ENaC is high) and gradually increasing detection levels toward the collecting duct (CD; where apical ENaC is low). Compared with high-Na(+) diet (5% Na(+)), 2 wk of low-Na(+) diet (0.01% Na(+)) drastically reduces Nedd4-2 immunostaining and increases apical ENaC abundance in ASDN. Reduced Nedd4-2 immunostaining is not dependent on increased apical Na(+) entry in the CD, because it is similarly observed in mice with intact and with suppressed apical ENaC activity in the CD. Consistent with a role of mineralocorticoid hormones in the long-term regulation of Nedd4-2, 5-d treatment of cultured CD (mpkCCD(cl4)) cells with 1 microM aldosterone leads to reduction of Nedd4-2 protein expression. It is concluded that Nedd4-2 abundance is regulated by Na(+) diet, by a mechanism that likely involves aldosterone. This regulation may contribute to adaptation of apical ENaC activity to altered Na(+) intake.
- Published
- 2006
23. Aldosterone-Induced Serum and Glucocorticoid-Induced Kinase 1 Expression Is Accompanied by Nedd4-2 Phosphorylation and Increased Na+ Transport in Cortical Collecting Duct Cells
- Author
-
Dorothée Daidié, Jean Dudler, Carole Gerbex, Johannes Loffing, Olivier Staub, Elena Kamynina, Sandra Y. Flores, Sting Chabanel, and Dominique Loffing-Cueni
- Subjects
Epithelial sodium channel ,medicine.medical_specialty ,DNA, Complementary ,Time Factors ,medicine.drug_class ,Nedd4 Ubiquitin Protein Ligases ,Ubiquitin-Protein Ligases ,macromolecular substances ,Protein Serine-Threonine Kinases ,Xenopus Proteins ,Biology ,Kidney ,Adenoviridae ,Cell Line ,Immediate-Early Proteins ,Mice ,Phosphatidylinositol 3-Kinases ,Xenopus laevis ,chemistry.chemical_compound ,Internal medicine ,medicine ,Animals ,Kidney Tubules, Collecting ,Phosphorylation ,Aldosterone ,Phosphoinositide-3 Kinase Inhibitors ,Cryopreservation ,Endosomal Sorting Complexes Required for Transport ,urogenital system ,Reabsorption ,Kinase ,Sodium ,General Medicine ,Phosphoric Monoester Hydrolases ,Rats ,Retroviridae ,Endocrinology ,chemistry ,Nephrology ,Mineralocorticoid ,SGK1 ,Homeostasis ,Plasmids ,Protein Binding - Abstract
Aldosterone plays a central role in Na⁺ homeostasis by controlling Na⁺ reabsorption in the aldosterone-sensitive distal nephron involving the epithelial Na⁺ channel (ENaC). Part of the effects of aldosterone is mediated by serum and glucocorticoid-induced kinase 1 (Sgk1), a Ser/Thr kinase whose expression is rapidly induced by aldosterone and that increases in heterologous expression systems ENaC cell surface abundance and activity. Previous work in Xenopus laevis oocytes suggested that Sgk1 phosphorylates specific residues (Ser212 and Ser328) on the ubiquitin-protein ligase Nedd4-2, an enzyme that directly interacts with ENaC and negatively controls channel density at the plasma membrane. It further indicated that phosphorylation of Nedd4-2 led to impairment of ENaC/Nedd4-2 interaction and consequently to more channels at the cell surface. These data suggested a novel mode of aldosterone-dependent action, yet this was not demonstrated formally in epithelial cells that physiologically express ENaC. Here it is shown, with the use of an anti–phospho-Ser328-mNedd4-2 antibody, that 2 to 6 h of aldosterone treatment induces an increase in Nedd4-2 phosphorylation, both in a mouse cortical collecting duct cell line (mpkCCDcl4) and in kidneys of adrenalectomized rats. This augmentation, which is accompanied by a raise in Sgk1 expression and transepithelial Na⁺ transport, is sensitive to phosphatidylinositol-3 kinase inhibition, as is Sgk1 phosphorylation and Na⁺ transport. Hence, these data provide evidence in cortical collecting duct cells in vitro and in vivo that Sgk1-dependent phosphorylation of Nedd4-2 is part of the aldosterone response.
- Published
- 2005
24. Functional Cooperation of Epithelial Heteromeric Amino Acid Transporters Expressed in Madin-Darby Canine Kidney Cells
- Author
-
Dominique Loffing-Cueni, Natasha Forster, François Verrey, Christian Bauch, and Vanessa Summa
- Subjects
Cystine ,Biology ,Arginine ,Kidney ,Biochemistry ,Renal protein reabsorption ,Cell Line ,Large Neutral Amino Acid-Transporter 1 ,Kidney Tubules, Proximal ,Mice ,chemistry.chemical_compound ,Dogs ,Leucine ,medicine ,Animals ,Secretion ,Molecular Biology ,Epithelial polarity ,chemistry.chemical_classification ,Membrane Glycoproteins ,Reabsorption ,Fusion Regulatory Protein 1, Light Chains ,Biological Transport ,Transporter ,Cell Biology ,Cell biology ,Amino acid ,medicine.anatomical_structure ,chemistry ,Amino Acid Transport Systems, Basic ,Carrier Proteins - Abstract
The heteromeric amino acid transporters b(0,+)AT-rBAT (apical), y(+)LAT1-4F2hc, and possibly LAT2-4F2hc (basolateral) participate to the (re)absorption of cationic and neutral amino acids in the small intestine and kidney proximal tubule. We show now by immunofluorescence that their expression levels follow the same axial gradient along the kidney proximal tubule (S1S2S3). We reconstituted their co-expression in MDCK cell epithelia and verified their polarized localization by immunofluorescence. Expression of b(0,+)AT-rBAT alone led to a net reabsorption of l-Arg (given together with l-Leu). Coexpression of basolateral y(+)LAT1-4F2hc increased l-Arg reabsorption and reversed l-Leu transport from (re)absorption to secretion. Similarly, l-cystine was (re)absorbed when b(0,+)AT-rBAT was expressed alone. This net transport was further increased by the coexpression of 4F2hc, due to the mobilization of LAT2 (exogenous and/or endogenous) to the basolateral membrane. In summary, apical b(0,+)AT-rBAT cooperates with y(+)LAT1-4F2hc or LAT2-4F2hc for the transepithelial reabsorption of cationic amino acids and cystine, respectively. The fact that the reabsorption of l-Arg led to the secretion of l-Leu demonstrates that the implicated heteromeric amino acid transporters function in epithelia as exchangers coupled in series and supports the notion that the parallel activity of unidirectional neutral amino acid transporters is required to drive net amino acid reabsorption.
- Published
- 2003
25. SP022MOLECULAR CHARACTERIZATION OF A NOVEL MUTATION IN THE RENAL NACL COTRANSPORTER CAUSING GITELMAN'S SYNDROME BY IMPAIRING TRANSPORTER TRAFFICKING
- Author
-
Johannes Loffing, Elisa Pagnin, Dominique Loffing-Cueni, Michéle Heidemeier, Verdiana Ravarotto, Lorenzo A. Calò, and Gian Paolo Rossi
- Subjects
medicine.medical_specialty ,Population ,medicine.disease_cause ,Hypocalciuria ,Tubulopathy ,Internal medicine ,parasitic diseases ,medicine ,Distal convoluted tubule ,education ,Transplantation ,Kidney ,education.field_of_study ,Mutation ,urogenital system ,business.industry ,Point mutation ,HEK 293 cells ,medicine.disease ,Molecular biology ,Endocrinology ,medicine.anatomical_structure ,Nephrology ,embryonic structures ,medicine.symptom ,business - Abstract
Mutations affecting the sodium-chloride cotransporter (NCC) in the distal convoluted tubule of the nephron are causative of Gitelman's syndrome (GS), a rare autosomal recessive disease characterized by electrolytic alterations similar to those induced by high dose thiazide treatment. Notably, the co-existence of hypomagnesemia and hypocalciuria is a feature of GS which is a distinguish hallmark from another hypokalemic renal tubulopathy, the Bartter's syndrome (BS). Commonly GS is heterozygous compound with an estimated prevalence of 1:40000 and can be silent for years before the revealing in the early adulthood. Recognizing the genetic background is fundamental for the screening and the diagnosis of the disease, as recently studies showed that mutation affecting regulators of renal salt handling are underestimated in the general population. In a registry of BS/GS based at our University we discovered a novel point mutation (c.1204G>A which codify for an amino acid exchange G394D) in the sodium-chloride cotransporter NCC (SLC12A3) in a young woman with hypokalemia, hypomagnesemia and hypocalciuria associated to muscle pain and cramps. The present study aimed to investigate how this mutation affects NCC function by using a molecular biology approach and providing functional evidences. After a prior screening with bioinformatics tools predicting the possible pathogenicity of the mutation, were created different expression vectors with either the wild-type (wt-NCC) or the mutated G394D-NCC sequences. DNA and in-vitro transcribed RNA were afterwards transfected in a human embryonic kidney cells line (HEK293) and injected into oocytes deriving from Xenopus Laevis frog respectively. In transfected HEK 293 cells, wildtype NCC was detected by immunoblotting as two bands at approximately 130 kDa and 115 kDa corresponding to fully and core-glycosylated NCC, respectively. In contrast, G394D-NCC was seen as a single band at about 115 kDa only, suggesting an impaired maturation of the mutated protein. Similar findings were made in the oocyte expression system. Confocal microscopy on the oocytes, did also show a strong cell surface localization of wildtype NCC while mutated NCC was retained at intracellular compartments. Consistently, a decent thiazide-sensitive 22Na+ uptake into injected oocytes was only found for wildtype but not mutated NCC. Taken together all the findings in this study, a novel GS point mutation has been characterized to diminish NCC function by impairing trafficking of the protein to the cell surface. The absence of any mature glycosylation form of G394D-NCC suggests that the mutation impairs protein folding leading to a retention of NCC in the endoplasmic reticulum.
- Published
- 2017
26. 'Deletion of both Rab-GTPase–activating proteins TBC1D1 and TBC1D4 in mice eliminates insulin- and AICAR-stimulated glucose transport [corrected]
- Author
-
Johannes Loffing, Anja Immisch, Christian de Wendt, Torben Stermann, Christian Springer, Dominique Loffing-Cueni, Hans-Georg Joost, Hadi Al-Hasani, Alexandra Chadt, Zhou Zhou, Geoffrey D. Holman, University of Zurich, and Al-Hasani, Hadi
- Subjects
Male ,medicine.medical_specialty ,10017 Institute of Anatomy ,Genotyping Techniques ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Glucose uptake ,Adipose tissue ,610 Medicine & health ,030209 endocrinology & metabolism ,White adipose tissue ,Carbohydrate metabolism ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Internal medicine ,Internal Medicine ,medicine ,Animals ,Insulin ,030304 developmental biology ,0303 health sciences ,Glucose Transporter Type 4 ,biology ,Body Weight ,GTPase-Activating Proteins ,Glucose transporter ,Skeletal muscle ,Nuclear Proteins ,Biological Transport ,Calorimetry, Indirect ,Ribonucleotides ,Aminoimidazole Carboxamide ,2712 Endocrinology, Diabetes and Metabolism ,Endocrinology ,medicine.anatomical_structure ,Glucose ,2724 Internal Medicine ,biology.protein ,Body Composition ,570 Life sciences ,GLUT4 - Abstract
The Rab-GTPase–activating proteins TBC1D1 and TBC1D4 (AS160) were previously shown to regulate GLUT4 translocation in response to activation of AKT and AMP-dependent kinase. However, knockout mice lacking either Tbc1d1 or Tbc1d4 displayed only partially impaired insulin-stimulated glucose uptake in fat and muscle tissue. The aim of this study was to determine the impact of the combined inactivation of Tbc1d1 and Tbc1d4 on glucose metabolism in double-deficient (D1/4KO) mice. D1/4KO mice displayed normal fasting glucose concentrations but had reduced tolerance to intraperitoneally administered glucose, insulin, and AICAR. D1/4KO mice showed reduced respiratory quotient, indicating increased use of lipids as fuel. These mice also consistently showed elevated fatty acid oxidation in isolated skeletal muscle, whereas insulin-stimulated glucose uptake in muscle and adipose cells was almost completely abolished. In skeletal muscle and white adipose tissue, the abundance of GLUT4 protein, but not GLUT4 mRNA, was substantially reduced. Cell surface labeling of GLUTs indicated that RabGAP deficiency impairs retention of GLUT4 in intracellular vesicles in the basal state. Our results show that TBC1D1 and TBC1D4 together play essential roles in insulin-stimulated glucose uptake and substrate preference in skeletal muscle and adipose cells.
- Published
- 2014
27. Analyse der komplementären Funktion von TBC1D1 und TBC1D4 im Glukosestoffwechsel des Skelettmuskels
- Author
-
Torben Stermann, Johannes Loffing, HG Joost, Alexandra Chadt, Hadi Al-Hasani, Anja Immisch, Dominique Loffing-Cueni, C de Wendt, and S Osthold
- Subjects
Endocrinology, Diabetes and Metabolism - Published
- 2014
28. Protein phosphatase 1 Inhibitor‐1 deficiency causes hypotension and reduces the phosphorylation of renal NaCl cotransporter (892.18)
- Author
-
Johannes Loffing, R L Miller, Chao-Ling Yang, Dominique Loffing-Cueni, Monique Carrel, Nicolas Picard, Robert A. Fenton, Katja Trompf, and David H. Ellison
- Subjects
medicine.medical_specialty ,urogenital system ,Chemistry ,Kinase ,Protein phosphatase 1 ,Biochemistry ,medicine.anatomical_structure ,Endocrinology ,Ion homeostasis ,Blood pressure ,Internal medicine ,parasitic diseases ,embryonic structures ,Genetics ,medicine ,Phosphorylation ,Distal convoluted tubule ,Cotransporter ,Molecular Biology ,Biotechnology - Abstract
The thiazide-sensitive NaCl cotransporter (NCC) of the renal distal convoluted tubule (DCT) controls ion homeostasis and blood pressure (BP). Gain-of-function in NCC-regulating kinases or loss-of-f...
- Published
- 2014
29. Trafficking of GFP-tagged ΔF508-CFTR to the plasma membrane in a polarized epithelial cell line
- Author
-
Collin Shaw, Malu Govindan, Bruce A. Stanton, Dominique Loffing-Cueni, Caitlin R. Stanton, Jan Loffing, and Amilyn M. Taplin
- Subjects
Nystatin ,Physiology ,Recombinant Fusion Proteins ,Green Fluorescent Proteins ,Cystic Fibrosis Transmembrane Conductance Regulator ,Cell Line ,Green fluorescent protein ,Cell polarity ,Cyclic AMP ,medicine ,Animals ,Humans ,ortho-Aminobenzoates ,Transgenes ,Enzyme Inhibitors ,Ion transporter ,Epithelial polarity ,Microscopy, Confocal ,Ionophores ,biology ,Chemistry ,Cell Membrane ,Temperature ,Cell Polarity ,Epithelial Cells ,Cell Biology ,Membrane transport ,Calcium Channel Blockers ,Genistein ,Epithelium ,Cystic fibrosis transmembrane conductance regulator ,Transport protein ,Cell biology ,Electrophysiology ,Butyrates ,Luminescent Proteins ,Protein Transport ,medicine.anatomical_structure ,Biochemistry ,biology.protein ,Indicators and Reagents - Abstract
The ΔF508 mutation reduces the amount of cystic fibrosis transmembrane conductance regulator (CFTR) expressed in the plasma membrane of epithelial cells. However, a reduced temperature, butyrate compounds, and “chemical chaperones” allow ΔF508-CFTR to traffic to the plasma membrane and increase Cl−permeability in heterologous and nonpolarized cells. Because trafficking is affected by the polarized state of epithelial cells and is cell-type dependent, our goal was to determine whether these maneuvers induce ΔF508-CFTR trafficking to the apical plasma membrane in polarized epithelial cells. To this end, we generated and characterized a line of polarized Madin-Darby canine kidney (MDCK) cells stably expressing ΔF508-CFTR tagged with green fluorescent protein (GFP). A reduced temperature, glycerol, butyrate, or DMSO had no effect on 8-(4-chlorophenylthio)-cAMP (CPT-cAMP)-stimulated transepithelial Cl−secretion across polarized monolayers. However, when the basolateral membrane was permeabilized, butyrate, but not the other experimental maneuvers, increased the CPT-cAMP-stimulated Cl−current across the apical plasma membrane. Thus butyrate increased the amount of functional ΔF508-CFTR in the apical plasma membrane. Butyrate failed to stimulate transepithelial Cl−secretion because of inhibitory effects on Cl−uptake across the basolateral membrane. These observations suggest that studies on heterologous and nonpolarized cells should be interpreted cautiously. The GFP tag on ΔF508-CFTR will allow investigation of ΔF508-CFTR trafficking in living, polarized MDCK epithelial cells in real time.
- Published
- 2001
30. Luminal Heterodimeric Amino Acid Transporter Defective in Cystinuria
- Author
-
François Verrey, Grégoire Rossier, Jan Loffing, Rahel Pfeiffer, Thomas Eggermann, Christian Bauch, Christian Meier, Lukas C. Kühn, and Dominique Loffing-Cueni
- Subjects
Male ,Amino Acid Transport Systems ,Brush border ,Protein subunit ,Molecular Sequence Data ,Fluorescent Antibody Technique ,Sequence alignment ,Biology ,Kidney ,Article ,Mice ,Xenopus laevis ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Amino acid transporter ,Amino Acids ,Cloning, Molecular ,Molecular Biology ,Peptide sequence ,In Situ Hybridization ,chemistry.chemical_classification ,Cystinuria ,Membrane Glycoproteins ,Microvilli ,Biological Transport ,Cell Biology ,medicine.disease ,Molecular biology ,Amino acid ,Biochemistry ,chemistry ,Organ Specificity ,Oocytes ,Amino Acid Transport Systems, Basic ,Carrier Proteins ,Glycoprotein ,Chromosomes, Human, Pair 19 ,Sequence Alignment - Abstract
Mutations of the glycoprotein rBAT cause cystinuria type I, an autosomal recessive failure of dibasic amino acid transport (b0,+type) across luminal membranes of intestine and kidney cells. Here we identify the permease-like protein b0,+AT as the catalytic subunit that associates by a disulfide bond with rBAT to form a hetero-oligomeric b0,+amino acid transporter complex. We demonstrate its b0,+-type amino acid transport kinetics using a heterodimeric fusion construct and show its luminal brush border localization in kidney proximal tubule. These biochemical, transport, and localization characteristics as well as the chromosomal localization on 19q support the notion that the b0,+AT protein is the product of the gene defective in non-type I cystinuria.
- Published
- 1999
31. Molecular Cloning and Tissue Expression of the Insulin-like Growth Factor II Prohormone in the Bony FishCottus scorpius
- Author
-
Manfred Reinecke, Annette C. Schmid, and Dominique Loffing-Cueni
- Subjects
Molecular Sequence Data ,Prohormone ,Scorpaeniformes ,Biology ,Molecular cloning ,Endocrinology ,Insulin-Like Growth Factor II ,Complementary DNA ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Cloning, Molecular ,Southern blot ,chemistry.chemical_classification ,Messenger RNA ,Base Sequence ,Reverse Transcriptase Polymerase Chain Reaction ,Fishes ,biology.organism_classification ,Molecular biology ,Amino acid ,Blotting, Southern ,Liver ,chemistry ,Sculpin ,Animal Science and Zoology ,medicine.drug - Abstract
The cDNA encoding pro-IGF-II of an advanced teleost fish, Cottus scorpius (Scorpaeniformes), the daddy sculpin, was isolated from liver by RT-PCR and molecular cloning. Like other IGFs, the deduced 168 amino acid peptide contains B-, C-, A-, D-, and E-domains and six cysteine residues (CysB9, CysB21, CysA6, CysA7, CysA11, and CysA20) necessary for the maintenance of tertiary structure. At the amino acid level, the sculpin IGF-II prohormone exhibits 85-92% homology to pro-IGF-II of other bony fish but only 51% homology to human. The mature sculpin IGF-II peptide comprises 70 amino acids. Its A-, B-, and D-domains exhibit homologies as high as 91, 91, and 100%, respectively, when compared with the other bony fish species studied. The high sequence homologies may indicate a particular physiological impact of IGF-II in bony fish. RT-PCR followed by Southern blotting revealed an IGF-II mRNA transcript of the expected size in liver, pyloric and splenic islets, stomach, small and large intestine, kidney, gill, testis, ovary, brain, and heart. The local production of IGF-II in many organs indicates that IGF-II is involved in organ-specific functions in a paracrine/autocrine manner. Furthermore, the results show that all bony fish organs which have been demonstrated to express IGF-I mRNA also express IGF-II mRNA.
- Published
- 1999
32. Subunit Stoichiometry of a Core Conduction Element in a Cloned Epithelial Amiloride-Sensitive Na+ Channel
- Author
-
Bakhrom K. Berdiev, Pierre-Jean Ripoll, Iskander I. Ismailov, Biljana Jovov, Patricia A. Halpin, Thomas R. Kleyman, Dominique Loffing-Cueni, Bruce A. Stanton, Ryan G. Morris, and Katherine H. Karlson
- Subjects
Epithelial sodium channel ,Proteolipids ,Xenopus ,Protein subunit ,Lipid Bilayers ,Mutant ,Kinetics ,Biophysics ,Plasma protein binding ,Biology ,Sodium Channels ,Amiloride ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Cloning, Molecular ,Lipid bilayer ,Sequence Deletion ,030304 developmental biology ,0303 health sciences ,Sodium channel ,Molecular biology ,Recombinant Proteins ,Electrophysiology ,030217 neurology & neurosurgery ,Research Article ,Protein Binding ,medicine.drug - Abstract
The molecular composition of a core conduction element formed by the alpha-subunit of cloned epithelial Na+ channels (ENaC) was studied in planar lipid bilayers. Two pairs of in vitro translated proteins were employed in combinatorial experiments: 1) wild-type (WT) and an N-terminally truncated alphaDeltaN-rENaC that displays accelerated kinetics (tauo = 32 +/- 13 ms, tauc = 42 +/- 11 ms), as compared with the WT channel (tauc1 = 18 +/- 8 ms, tauc2 = 252 +/- 31 ms, and tauo = 157 +/- 43 ms); and 2) WT and an amiloride binding mutant, alphaDelta278-283-rENaC. The channels that formed in a alphaWT:alphaDeltaN mixture fell into two groups: one with tauo and tauc that corresponded to those exhibited by the alphaDeltaN-rENaC alone, and another with a double-exponentially distributed closed time and a single-exponentially distributed open time that corresponded to the alphaWT-rENaC alone. Five channel subtypes with distinct sensitivities to amiloride were found in a 1alphaWT:1alphaDelta278-283 protein mixture. Statistical analyses of the distributions of channel phenotypes observed for either set of the WT:mutant combinations suggest a tetrameric organization of alpha-subunits as a minimal model for the core conduction element in ENaCs.
- Published
- 1998
33. IGF-I in the bony fish Cottus scorpius: cDNA, expression and differential localization in brain and islets1The sequence reported has been deposited in the EMBL Nucleotide Sequence Database (Accession No. Y12583).1
- Author
-
Manfred Reinecke, Dominique Loffing-Cueni, Hannes Graf, and Annette C. Schmid
- Subjects
Signal peptide ,medicine.medical_specialty ,Insulin ,medicine.medical_treatment ,Pancreatic islets ,Biology ,Biochemistry ,Paracrine signalling ,Endocrinology ,medicine.anatomical_structure ,Internal medicine ,Complementary DNA ,Gene expression ,medicine ,Autocrine signalling ,Molecular Biology ,Peptide sequence - Abstract
The cDNA encoding prepro-insulin-like growth factor (IGF)-I of a teleost, Cottus scorpius, (Scorpaeniformes) was established from liver by RT-PCR and molecular cloning. Typically, the deduced 184 amino acid protein contains a signal peptide, B-, C-, A-, D- and E-domains and all residues necessary for maintenance of tertiary structure. C. scorpius IGF-I shares only ∼57% identity with C. scorpius insulin in the A-domain and 7% in the B-domain. RT-PCR followed by Southern blotting revealed a transcript in liver, pancreatic islets, stomach, small and large intestine, kidney, gill, testis, ovary, heart and brain indicating paracrine/autocrine actions of locally produced IGF-I. IGF-I- and insulin-immunoreactivities coexisted in the islets, but did not in other sites such as brain. Thus, in contrast to other bony fish, sculpin insulin cells most probably produce IGF-I. The results also challenge the current model of the IGF/insulin evolution.
- Published
- 1998
34. Insulin-Like Growth Factor I in the Teleost Oreochromis mossambicus, the Tilapia: Gene Sequence, Tissue Expression, and Cellular Localization*
- Author
-
Annette C. Schmid, Manfred Reinecke, Rebekka Ermatinger, and Dominique Loffing-Cueni
- Subjects
endocrine system ,medicine.medical_specialty ,Oreochromis mossambicus ,DNA, Complementary ,food.ingredient ,medicine.medical_treatment ,Molecular Sequence Data ,Fluorescent Antibody Technique ,Sequence Homology ,Biology ,Polymerase Chain Reaction ,Insulin-like growth factor ,Paracrine signalling ,Endocrinology ,food ,Internal medicine ,medicine ,Animals ,Tissue Distribution ,Amino Acid Sequence ,RNA, Messenger ,Insulin-Like Growth Factor I ,Cellular localization ,Head Kidney ,Base Sequence ,Pancreatic islets ,Growth factor ,RNA-Directed DNA Polymerase ,Tilapia ,Sequence Analysis, DNA ,biology.organism_classification ,Immunohistochemistry ,Molecular biology ,medicine.anatomical_structure - Abstract
Using reverse transcription-PCR and molecular cloning, the complementary DNA sequence encoding preproinsulin-like growth factor I (IGF-I) of a teleost, the tilapia (Oreochromis mossambicus) was established from liver. At the amino acid level, tilapia IGF-I shows all residues necessary for the maintenance of tertiary structure and shares about 80% identity with IGF-I from other teleosts. The B and A domains of tilapia IGF-I show more than 90% homology to those of other teleosts and 86 ‐93% to those of human. However, in contrast to salmonids, the C domain of tilapia is truncated. Reverse transcription-PCR analysis followed by Southern blotting with an internal probe specific for tilapia IGF-I indicated a transcript in liver, pancreas, gut, kidney, head kidney, gill, ovary, testis, eye, and brain. In correlation, parenchymal cells were identified as likely local production sites by the use of immunohistochemistry. IGF-I immunoreactivity was confined to D cells in pancreatic islets, gastroentero-endocrine cells, cells of renal proximal tubules, interrenal cells of the head kidney, gill chondrocytes, chloride cells of the gill epithelium, granulosa cells in the ovary, spermatocytes and Sertoli cells in testis, and neurons in retina and brain. The local production of IGF-I in multiple organs of the tilapia indicates paracrine/autocrine actions of IGF-I involved in organ-specific functions. The results further demonstrate that the primary structure of IGF-I, especially in the B and A domains, is highly conserved during phylogeny. (Endocrinology 138: 3613‐3619, 1997)
- Published
- 1997
35. Protein phosphatase 1 inhibitor-1 deficiency reduces phosphorylation of renal NaCl cotransporter and causes arterial hypotension
- Author
-
Johannes Loffing, Monique Carrel, Nicolas Picard, R. Lance Miller, Robert A. Fenton, David H. Ellison, Chao Ling Yang, Katja Trompf, Dominique Loffing-Cueni, University of Zurich, and Loffing, Johannes
- Subjects
Male ,10017 Institute of Anatomy ,Xenopus ,Blood Pressure ,030204 cardiovascular system & hematology ,Mice ,0302 clinical medicine ,Solute Carrier Family 12, Member 3 ,Phosphorylation ,Kidney Tubules, Distal ,Solute Carrier Family 12, Member 1 ,0303 health sciences ,2727 Nephrology ,biology ,General Medicine ,Up-Regulation ,medicine.anatomical_structure ,Nephrology ,Sulfate Transporters ,10076 Center for Integrative Human Physiology ,Knockout mouse ,embryonic structures ,Female ,Hypotension ,medicine.medical_specialty ,Anion Transport Proteins ,610 Medicine & health ,Mice, Transgenic ,Protein Serine-Threonine Kinases ,Gene product ,03 medical and health sciences ,Internal medicine ,parasitic diseases ,medicine ,Animals ,Humans ,Distal convoluted tubule ,030304 developmental biology ,urogenital system ,Proteins ,Protein phosphatase 1 ,Pendrin ,Gitelman syndrome ,medicine.disease ,Ion homeostasis ,Endocrinology ,Basic Research ,biology.protein ,Loop of Henle ,570 Life sciences - Abstract
The thiazide-sensitive NaCl cotransporter (NCC) of the renal distal convoluted tubule (DCT) controls ion homeostasis and arterial BP. Loss-of-function mutations of NCC cause renal salt wasting with arterial hypotension (Gitelman syndrome). Conversely, mutations in the NCC-regulating WNK kinases or kelch-like 3 protein cause familial hyperkalemic hypertension. Here, we performed automated sorting of mouse DCTs and microarray analysis for comprehensive identification of novel DCT-enriched gene products, which may potentially regulate DCT and NCC function. This approach identified protein phosphatase 1 inhibitor-1 (I-1) as a DCT-enriched transcript, and immunohistochemistry revealed I-1 expression in mouse and human DCTs and thick ascending limbs. In heterologous expression systems, coexpression of NCC with I-1 increased thiazide-dependent Na(+) uptake, whereas RNAi-mediated knockdown of endogenous I-1 reduced NCC phosphorylation. Likewise, levels of phosphorylated NCC decreased by approximately 50% in I-1 (I-1(-/-)) knockout mice without changes in total NCC expression. The abundance and phosphorylation of other renal sodium-transporting proteins, including NaPi-IIa, NKCC2, and ENaC, did not change, although the abundance of pendrin increased in these mice. The abundance, phosphorylation, and subcellular localization of SPAK were similar in wild-type (WT) and I-1(-/-) mice. Compared with WT mice, I-1(-/-) mice exhibited significantly lower arterial BP but did not display other metabolic features of NCC dysregulation. Thus, I-1 is a DCT-enriched gene product that controls arterial BP, possibly through regulation of NCC activity.
- Published
- 2013
36. Role of the renal mineralocorticoid receptor for potassium homeostasis
- Author
-
Dominique Loffing-Cueni, Anne Debonneville, Edith Hummler, Jérémie Canonica, Caroline Ronzaud, Marian Roesinger, Olivier Staub, and Johannes Loffing
- Subjects
medicine.medical_specialty ,Mineralocorticoid receptor ,Endocrinology ,Chemistry ,Internal medicine ,Potassium ,Genetics ,medicine ,chemistry.chemical_element ,Molecular Biology ,Biochemistry ,Homeostasis ,Biotechnology - Published
- 2013
37. Renal tubular NEDD4-2 deficiency causes NCC-mediated salt-dependent hypertension
- Author
-
Caroline Ronzaud, Natasha A. Boase, Johannes Loffing, Baoli Yang, Romain Perrier, Nicole Fowler-Jaeger, Edith Hummler, Dominique Loffing-Cueni, Sumedha Malsure, Marc Maillard, Sharad Kumar, Pierrette Hausel, Olivier Staub, Robert Koesters, Anne Debonneville, John B. Stokes, Ronzaud, Caroline, Loffing-Cueni, Dominique, Hausel, Pierrette, Debonneville, Anne, Malsure, Sumedha Ram, Fowler-Jaeger, Nicole, Boase, Natasha Anne, Perrier, Romain, Maillard, Marc, Yang, Baoli, Stokes, John B, Koesters, Robert, Kumar, Sharad, Hummler, Edith, Loffing, Johannes, Staub, Olivier, and University of Zurich
- Subjects
Epithelial sodium channel ,10017 Institute of Anatomy ,Animals ,Blood Pressure ,Disease Models, Animal ,Endosomal Sorting Complexes Required for Transport/deficiency ,Endosomal Sorting Complexes Required for Transport/genetics ,Epithelial Sodium Channels/metabolism ,Humans ,Hypertension/etiology ,Hypertension/genetics ,Kidney Tubules/physiopathology ,Liddle Syndrome/etiology ,Liddle Syndrome/genetics ,Mice ,Mice, Knockout ,Potassium/blood ,Potassium/urine ,Potassium Channels, Inwardly Rectifying/metabolism ,Receptors, Drug/metabolism ,Sodium/blood ,Sodium/urine ,Sodium, Dietary/administration & dosage ,Sodium, Dietary/adverse effects ,Symporters/metabolism ,Ubiquitin-Protein Ligases/deficiency ,Ubiquitin-Protein Ligases/genetics ,Nedd4 Ubiquitin Protein Ligases ,Receptors, Drug ,knockout ,2700 General Medicine ,030204 cardiovascular system & hematology ,chemistry.chemical_compound ,0302 clinical medicine ,Solute Carrier Family 12, Member 3 ,humans ,0303 health sciences ,Aldosterone ,Symporters ,Chemistry ,General Medicine ,animals ,Kidney Tubules ,kidney tubules ,Hypertension ,Research Article ,medicine.medical_specialty ,mice ,hypertension ,Ubiquitin-Protein Ligases ,610 Medicine & health ,macromolecular substances ,03 medical and health sciences ,Liddle Syndrome ,Internal medicine ,medicine ,Potassium Channels, Inwardly Rectifying ,Epithelial Sodium Channels ,030304 developmental biology ,NEDD4L ,disease models ,Endosomal Sorting Complexes Required for Transport ,urogenital system ,Sodium ,Sodium, Dietary ,Endocrinology ,Symporter ,Potassium ,Commentary ,ROMK ,570 Life sciences ,biology ,Cotransporter ,Homeostasis - Abstract
The E3 ubiquitin ligase NEDD4-2 (encoded by the Nedd4L gene) regulates the amiloride-sensitive epithelial Na+ channel (ENaC/SCNN1) to mediate Na+ homeostasis. Mutations in the human β/γENaC subunits that block NEDD4-2 binding or constitutive ablation of exons 6-8 of Nedd4L in mice both result in salt-sensitive hypertension and elevated ENaC activity (Liddle syndrome). To determine the role of renal tubular NEDD4-2 in adult mice, we generated tetracycline-inducible, nephron-specific Nedd4L KO mice. Under standard and high- Na+ diets, conditional KO mice displayed decreased plasma aldosterone but normal Na+/K+ balance. Under a high-Na+ diet, KO mice exhibited hypercalciuria and increased blood pressure, which were reversed by thiazide treatment. Protein expression of βENaC, γENaC, the renal outer medullary K+ channel (ROMK), and total and phosphorylated thiazide-sensitive Na+Cl- cotransporter (NCC) levels were increased in KO kidneys. Unexpectedly, Scnn1a mRNA, which encodes the αENaC subunit, was reduced and proteolytic cleavage of αENaC decreased. Taken together, these results demonstrate that loss of NEDD4-2 in adult renal tubules causes a new form of mild, salt-sensitive hypertension without hyperkalemia that is characterized by upregulation of NCC, elevation of β/γENaC, but not αENaC, and a normal Na+/K+ balance maintained by downregulation of ENaC activity and upregulation of ROMK. Refereed/Peer-reviewed
- Published
- 2013
38. Aldosterone deficiency adversely affects pregnancy outcome in mice
- Author
-
Dominique Loffing-Cueni, Carla Bettoni, Carsten A. Wagner, Markus Mohaupt, Marianna Di Chiara, Johannes Loffing, Abhijeet Todkar, University of Zurich, and Wagner, Carsten A
- Subjects
Aldosterone synthase ,Male ,10017 Institute of Anatomy ,Physiology ,Placenta ,Clinical Biochemistry ,Blood Pressure ,030204 cardiovascular system & hematology ,Sodium Chloride ,1308 Clinical Biochemistry ,10052 Institute of Physiology ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,2737 Physiology (medical) ,Pre-Eclampsia ,Pregnancy ,Lymphocytes ,Aldosterone ,reproductive and urinary physiology ,0303 health sciences ,Fetal Growth Retardation ,Homozygote ,Pregnancy Outcome ,Proteinuria ,medicine.anatomical_structure ,10076 Center for Integrative Human Physiology ,Gestation ,Female ,medicine.medical_specialty ,Heterozygote ,Gestational Age ,610 Medicine & health ,Biology ,Preeclampsia ,03 medical and health sciences ,Necrosis ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Cytochrome P-450 CYP11B2 ,Fetal Death ,030304 developmental biology ,Fetus ,1314 Physiology ,medicine.disease ,Diet ,Disease Models, Animal ,Blood pressure ,Endocrinology ,chemistry ,Mutation ,biology.protein ,570 Life sciences ,biology - Abstract
Circulating aldosterone levels are increased in human pregnancy. Inadequately low aldosterone levels as present in preeclampsia, a life-threatening disease for both mother and child, are discussed to be involved in its pathogenesis or severity. Moreover, inactivating polymorphisms in the aldosterone synthase gene have been detected in preeclamptic women. Here, we used aldosterone synthase-deficient (AS(-/-)) mice to test whether the absence of aldosterone is sufficient to impair pregnancy or even to cause preeclampsia. AS(-/-) and AS(+/+) females were mated with AS(+/+) and AS(-/-) males, respectively, always generating AS(+/-) offspring. With maternal aldosterone deficiency in AS(-/-) mice, systolic blood pressure was low before and further reduced during pregnancy with no increase in proteinuria. Yet, AS(-/-) had smaller litters due to loss of fetuses as indicated by a high number of necrotic placentas with massive lymphocyte infiltrations at gestational day 18. Surviving fetuses and their placentas from AS(-/-) females were smaller. High-salt diet before and during pregnancy increased systolic blood pressure only before pregnancy in both genotypes and abolished the difference in blood pressure during late pregnancy. Litter size from AS(-/-) was slightly improved and the differences in placental and fetal weights between AS(+/+) and AS(-/-) mothers disappeared. Overall, an increased placental efficiency was observed in both groups paralleled by a normalization of elevated HIF1α levels in the AS(-/-) placentas. Our results demonstrate that aldosterone deficiency has profound adverse effects on placental function. High dietary salt intake improved placental function. In this animal model, aldosterone deficiency did not cause preeclampsia.
- Published
- 2012
- Full Text
- View/download PDF
39. In vivo nuclear translocation of mineralocorticoid and glucocorticoid receptors in rat kidney: differential effect of corticosteroids along the distal tubule
- Author
-
Dominique Loffing-Cueni, Johannes Loffing, Daniel Ackermann, Celso E. Gomez-Sanchez, Daniel Habermehl, Nikolay Gresko, Bernard C. Rossier, Monique Carrel, University of Zurich, and Loffing, J
- Subjects
2748 Urology ,Male ,medicine.medical_specialty ,10017 Institute of Anatomy ,Physiology ,medicine.drug_class ,610 Medicine & health ,030204 cardiovascular system & hematology ,Biology ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Glucocorticoid receptor ,Mineralocorticoid receptor ,Receptors, Glucocorticoid ,Corticosterone ,Antibody Specificity ,Internal medicine ,11-beta-Hydroxysteroid Dehydrogenase Type 2 ,medicine ,Animals ,Distal convoluted tubule ,RNA, Messenger ,Receptor ,Kidney Tubules, Distal ,Aldosterone ,030304 developmental biology ,0303 health sciences ,Adrenalectomy ,Sodium, Dietary ,1314 Physiology ,Nephrons ,Connecting tubule ,Rats ,Protein Transport ,medicine.anatomical_structure ,Endocrinology ,Receptors, Mineralocorticoid ,chemistry ,Mineralocorticoid ,570 Life sciences ,biology - Abstract
Aldosterone and corticosterone bind to mineralocorticoid (MR) and glucocorticoid receptors (GR), which, upon ligand binding, are thought to translocate to the cell nucleus to act as transcription factors. Mineralocorticoid selectivity is achieved by the 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2) that inactivates 11β-hydroxy glucocorticoids. High expression levels of 11β-HSD2 characterize the aldosterone-sensitive distal nephron (ASDN), which comprises the segment-specific cells of late distal convoluted tubule (DCT2), connecting tubule (CNT), and collecting duct (CD). We used MR- and GR-specific antibodies to study localization and regulation of MR and GR in kidneys of rats with altered plasma aldosterone and corticosterone levels. In control rats, MR and GR were found in cell nuclei of thick ascending limb (TAL), DCT, CNT, CD cells, and intercalated cells (IC). GR was also abundant in cell nuclei and the subapical compartment of proximal tubule (PT) cells. Dietary NaCl loading, which lowers plasma aldosterone, caused a selective removal of GR from cell nuclei of 11β-HSD2-positive ASDN. The nuclear localization of MR was unaffected. Adrenalectomy (ADX) resulted in removal of MR and GR from the cell nuclei of all epithelial cells. Aldosterone replacement rapidly relocated the receptors in the cell nuclei. In ASDN cells, low-dose corticosterone replacement caused nuclear localization of MR, but not of GR. The GR was redistributed to the nucleus only in PT, TAL, early DCT, and IC that express no or very little 11β-HSD2. In ASDN cells, nuclear GR localization was only achieved when corticosterone was replaced at high doses. Thus ligand-induced nuclear translocation of MR and GR are part of MR and GR regulation in the kidney and show remarkable segment- and cell type-specific characteristics. Differential regulation of MR and GR may alter the level of heterodimerization of the receptors and hence may contribute to the complexity of corticosteroid effects on ASDN function.
- Published
- 2011
40. Aldosterone‐dependent and ‐independent effects on renal potassium excretion
- Author
-
Carsten A. Wagner, Oliver Smithies, Nicolas Picard, Johannes Loffing, Dominique Loffing-Cueni, Marija Mihailova, and Natalia Makhanova
- Subjects
endocrine system ,medicine.medical_specialty ,Aldosterone ,Hyperkalemia ,Potassium ,Renal K(+) excretion ,chemistry.chemical_element ,urologic and male genital diseases ,Biochemistry ,chemistry.chemical_compound ,Endocrinology ,chemistry ,Renal potassium excretion ,Internal medicine ,Genetics ,medicine ,ROMK ,medicine.symptom ,Molecular Biology ,Biotechnology - Abstract
Aldosterone (aldo) is released from adrenal glands upon potassium (K) loading and hyperkalemia. It stimulates renal K excretion via the ROMK K-channel and via activation of the apical epithelial so...
- Published
- 2008
41. Mouse model of type II Bartter's syndrome. II. Altered expression of renal sodium- and water-transporting proteins
- Author
-
Qingshang Yan, Johannes Loffing, John P. Geibel, Gerhard Giebisch, François Verrey, Monique Carrel, Nicole Schulz, Tong Wang, Carsten A. Wagner, Panagiotis Fakitsas, Dominique Loffing-Cueni, Steven C. Hebert, and University of Zurich
- Subjects
2748 Urology ,medicine.medical_specialty ,Sodium-Hydrogen Exchangers ,10017 Institute of Anatomy ,Physiology ,Sodium-Potassium-Chloride Symporters ,Sodium ,chemistry.chemical_element ,610 Medicine & health ,Bartter syndrome ,Sodium-Phosphate Cotransporter Proteins, Type IIa ,Dinoprostone ,10052 Institute of Physiology ,Kidney Tubules, Proximal ,chemistry.chemical_compound ,Mice ,Tubulopathy ,Internal medicine ,Benzamil ,medicine ,Renal medulla ,Animals ,Distal convoluted tubule ,Potassium Channels, Inwardly Rectifying ,Epithelial Sodium Channels ,Kidney Tubules, Distal ,Cation Transport Proteins ,Solute Carrier Family 12, Member 1 ,Kidney ,urogenital system ,Reverse Transcriptase Polymerase Chain Reaction ,Sodium-Hydrogen Exchanger 3 ,Bartter Syndrome ,Water ,1314 Physiology ,medicine.disease ,Adaptation, Physiological ,Mice, Mutant Strains ,Up-Regulation ,Bartter's syndrome ,Disease Models, Animal ,medicine.anatomical_structure ,Endocrinology ,chemistry ,10076 Center for Integrative Human Physiology ,Loop of Henle ,570 Life sciences ,biology ,Carrier Proteins - Abstract
Bartter's syndrome represents a group of hereditary salt- and water-losing renal tubulopathies caused by loss-of-function mutations in proteins mediating or regulating salt transport in the thick ascending limb (TAL) of Henle's loop. Mutations in the ROMK channel cause type II antenatal Bartter's syndrome that presents with maternal polyhydramnios and postnatal life-threatening volume depletion. We have developed a colony of Romk null mice showing a Bartter-like phenotype and with increased survival to adulthood, suggesting the activation of compensatory mechanisms. To test the hypothesis that upregulation of Na+-transporting proteins in segments distal to the TAL contributes to compensation, we studied expression of salt-transporting proteins in ROMK-deficient ( Romk−/−) mice. Plasma aldosterone was 40% higher and urinary PGE2 excretion was 1.5-fold higher in Romk−/− compared with wild-type littermates. Semiquantitative immunoblotting of kidney homogenates revealed decreased abundances of proximal tubule Na+/H+ exchanger (NHE3) and Na+-Pi cotransporter (NaPi-IIa) and TAL-specific Na+-K+-2Cl−-cotransporter (NKCC2/BSC1) in Romk−/− mice, while the distal convoluted tubule (DCT)-specific Na+-Cl− cotransporter (NCC/TSC) was markedly increased. The abundance of the α-,β-, and γ-subunits of the epithelial Na+ channel (ENaC) was slightly increased, although only differences for γ-ENaC reached statistical significance. Morphometry revealed a fourfold increase in the fractional volume of DCT but not of connecting tubule (CNT) and collecting duct (CCD). Consistently, CNT and CD of Romk−/− mice revealed no apparent increase in the luminal abundance of the ENaC compared with those of wild-type mice. These data suggest that the loss of ROMK-dependent Na+ absorption in the TAL is compensated predominately by upregulation of Na+ transport in downstream DCT cells. These adaptive changes in Romk−/− mice may help to limit renal Na+ loss, and thereby, contribute to survival of these mice.
- Published
- 2008
- Full Text
- View/download PDF
42. Deletion of Both Rab-GTPase–Activating Proteins TBC14KO and TBC1D4 in Mice Eliminates Insulin- and AICAR-Stimulated Glucose Transport. Diabetes 2015;64:746–759
- Author
-
Anja Immisch, Christian de Wendt, Christian Springer, Johannes Loffing, Dominique Loffing-Cueni, Hadi Al-Hasani, Alexandra Chadt, Hans-Georg Joost, Torben Stermann, Zhou Zhou, and Geoffrey D. Holman
- Subjects
Biochemistry ,GTPase-activating protein ,Endocrinology, Diabetes and Metabolism ,Diabetes mellitus ,Insulin ,medicine.medical_treatment ,Internal Medicine ,Glucose transporter ,medicine ,Rab ,Biology ,medicine.disease - Abstract
During production, two errors were introduced to the article listed above. The first is in the title, …
- Published
- 2015
43. Mineralocorticoid versus glucocorticoid receptor occupancy mediating aldosterone-stimulated sodium transport in a novel renal cell line
- Author
-
Johannes Loffing, Jean-Daniel Horisberger, Rikke Nørregaard, Nicole Fowler Jaeger, Bernard C. Rossier, Elena Gonzalez-Rodriguez, Hans-Peter Gaeggeler, and Dominique Loffing-Cueni
- Subjects
Epithelial sodium channel ,medicine.medical_specialty ,medicine.drug_class ,Sodium ,Carbenoxolone ,chemistry.chemical_element ,Biology ,Binding, Competitive ,Dexamethasone ,Mice ,chemistry.chemical_compound ,Receptors, Glucocorticoid ,Glucocorticoid receptor ,Corticosterone ,Internal medicine ,medicine ,Animals ,Kidney Tubules, Collecting ,Aldosterone ,Glucocorticoids ,Cell Line, Transformed ,Dose-Response Relationship, Drug ,Biological Transport ,General Medicine ,Receptors, Mineralocorticoid ,Endocrinology ,chemistry ,Nephrology ,Mineralocorticoid ,11-beta-Hydroxysteroid Dehydrogenases ,Glucocorticoid ,medicine.drug - Abstract
Aldosterone controls sodium balance by regulating an epithelial sodium channel (ENaC)-mediated sodium transport along the aldosterone-sensitive distal nephron, which expresses both mineralocorticoid (MR) and glucocorticoid receptors (GR). Mineralocorticoid specificity is ensured by 11beta-hydroxysteroid dehydrogenase type 2, which metabolizes cortisol or corticosterone into inactive metabolites that are unable to bind MR and/or GR. The fractional occupancy of MR and GR by aldosterone mediating the sodium transport response in the aldosterone-sensitive distal nephron cannot be studied in vivo. For answering this question, a novel mouse cortical collecting duct cell line (mCCD(cl1)), which expresses significant levels of MR and GR and a robust aldosterone sodium transport response, was used. Aldosterone elicited a biphasic response: Low doses (K(1/2) = approximately 0.5 nM) induced a transient and early increase of sodium transport (peaking at 3 h), whereas high doses (K(1/2) = approximately 90 nM) entailed an approximately threefold larger, long-lasting response. At 3 h, the corticosterone dose-response curve was shifted to the right compared with that of aldosterone by more than two log concentrations, an effect that was fully reverted in the presence of the 11beta-hydroxysteroid dehydrogenase type 2 inhibitor carbenoxolone. Low doses of dexamethasone (0.1 to 1 nM) failed to induce an early response, but high doses elicited a long-lasting response (K(1/2) = approximately 8 nM), similar to that observed for high aldosterone concentrations. Equilibrium binding assays showed that both aldosterone and corticosterone bind to a high-affinity, low-capacity site, whereas dexamethasone binds to one site. Within the physiologic range of aldosterone concentrations, sodium transport is predicted to be controlled by MR occupancy during circadian cycles and by MR and GR occupancy during salt restriction or acute stress.
- Published
- 2005
44. Altered renal distal tubule structure and renal Na(+) and Ca(2+) handling in a mouse model for Gitelman's syndrome
- Author
-
Gary E. Shull, Kerstin Richter, Johannes Loffing, Dominique Loffing-Cueni, Volker Vallon, Brigitte Kaissling, May Bloch-Faure, Joost G. J. Hoenderop, Pierre Meneton, Fintan Aregger, and Laurence Pietri
- Subjects
Epithelial sodium channel ,medicine.medical_specialty ,Receptors, Drug ,Hypocalciuria ,Mice ,Tubulopathy ,Internal medicine ,medicine ,Animals ,Magnesium ,Solute Carrier Family 12, Member 3 ,Distal convoluted tubule ,Kidney Tubules, Distal ,Ion transporter ,Symporters ,Reabsorption ,Chemistry ,urogenital system ,Sodium ,Alkalosis ,General Medicine ,Syndrome ,Gitelman syndrome ,medicine.disease ,Sodium Chloride Symporters ,Connecting tubule ,Renal disorders [UMCN 5.4] ,Disease Models, Animal ,medicine.anatomical_structure ,Endocrinology ,Nephrology ,Calcium ,Kidney Diseases ,medicine.symptom ,Carrier Proteins - Abstract
Item does not contain fulltext Gitelman's syndrome, an autosomal recessive renal tubulopathy caused by loss-of-function mutations in the thiazide-sensitive NaCl co-transporter (NCC) of the distal convoluted tubule (DCT), is characterized by mild renal Na(+) wasting, hypocalciuria, hypomagnesemia, and hypokalemic alkalosis. For gaining further insights into the pathophysiology of Gitelman's syndrome, the impact of NCC ablation on the morphology of the distal tubule, on the distribution and abundance of ion transport proteins along its length, and on renal tubular Na(+) and Ca(2+) handling in a gene-targeted mouse model was studied. NCC-deficient mice had significantly elevated plasma aldosterone levels and exhibited hypocalciuria, hypomagnesemia, and compensated alkalosis. Immunofluorescent detection of distal tubule marker proteins and ultrastructural analysis revealed that the early DCT, which physiologically lacks epithelial Na(+) (ENaC) and Ca(2+) (TRPV5) channels, was virtually absent in NCC-deficient mice. In contrast, the late DCT seemed intact and retained expression of the apical ENaC and TRPV5 as well as basolateral Na(+)-Ca(2+) exchanger. The connecting tubule exhibited a marked epithelial hypertrophy accompanied by an increased apical abundance of ENaC. Ca(2+) reabsorption seemed unaltered in the distal convolution (i.e., the DCT and connecting tubule) as indicated by real-time reverse transcription-PCR, Western blotting, and immunohistochemistry for TRPV5 and Na(+)-Ca(2+) exchanger and micropuncture experiments. The last experiments further indicated that reduced glomerular filtration and enhanced fractional reabsorption of Na(+) and Ca(2+) upstream and of Na(+) downstream of the DCT provide some compensation for the Na(+) transport defect in the DCT and contribute to the hypocalciuria. Thus, loss of NCC leads to major structural remodeling of the renal distal tubule that goes along with marked changes in glomerular and tubular function, which may explain some of the clinical features of Gitelman's syndrome.
- Published
- 2004
45. Distribution of transcellular calcium and sodium transport pathways along mouse distal nephron
- Author
-
Victor Valderrabano, Bernard C. Rossier, Johannes Loffing, Dominique Loffing-Cueni, L. E. A. Kläusli, René J. M. Bindels, Brigitte Kaissling, Steven C. Hebert, and Joost G. J. Hoenderop
- Subjects
Calbindins ,TRPV5 ,Physiology ,Sodium ,Renal cortex ,Receptors, Drug ,Regulation of salt and water reabsorption in the renal collecting duct ,chemistry.chemical_element ,TRPV Cation Channels ,Nephron ,Calcium-Transporting ATPases ,Calcium ,Models, Biological ,Sodium Channels ,Sodium-Calcium Exchanger ,Mice ,Plasma Membrane Calcium-Transporting ATPases ,S100 Calcium Binding Protein G ,Calcium-binding protein ,medicine ,Animals ,Solute Carrier Family 12, Member 3 ,Transcellular ,Epithelial Sodium Channels ,Kidney Tubules, Distal ,Cation Transport Proteins ,Kidney ,Ion Transport ,Symporters ,Immunohistochemistry ,Sodium Chloride Symporters ,Cell biology ,medicine.anatomical_structure ,Parvalbumins ,chemistry ,Biochemistry ,Regulatie water en zouttransport in de verzamelbuis van de nier ,Female ,Calcium Channels ,Carrier Proteins - Abstract
First published August 15, 2001; 10.1152/ajprenal. 00085.2001.—The organization of Na+and Ca2+transport pathways along the mouse distal nephron is incompletely known. We revealed by immunohistochemistry a set of Ca2+and Na+transport proteins along the mouse distal convolution. The thiazide-sensitive Na+-Cl−cotransporter (NCC) characterized the distal convoluted tubule (DCT). The amiloride-sensitive epithelial Na+channel (ENaC) colocalized with NCC in late DCT (DCT2) and extended to the downstream connecting tubule (CNT) and collecting duct (CD). In early DCT (DCT1), the basolateral Ca2+-extruding proteins [Na+/Ca2+exchanger (NCX), plasma membrane Ca2+-ATPase (PCMA)] and the cytoplasmic Ca2+-binding protein calbindin D28K(CB) were found at very low levels, whereas the cytoplasmic Ca2+/Mg2+-binding protein parvalbumin was highly abundant. NCX, PMCA, and CB prevailed in DCT2 and CNT, where we located the apical epithelial Ca2+channel (ECaC1). Its subcellular localization changed from apical in DCT2 to exclusively cytoplasmic at the end of CNT. NCX and PMCA decreased in parallel with the fading of ECaC1 in the apical membrane. All three of them were undetectable in CD. These findings disclose DCT2 and CNT as major sites for transcellular Ca2+transport in the mouse distal nephron. Cellular colocalization of Ca2+and Na+transport pathways suggests their mutual interactions in transport regulation.
- Published
- 2001
46. Localization of organic cation transporters OCT1 and OCT2 in rat kidney
- Author
-
Sebastian Bachmann, Valentin Gorboulev, Dominique Loffing-Cueni, Jörn Kricke, Ulrich Karbach, Brigitte Kaissling, Christopher Volk, Friederike Meyer-Wentrup, and Hermann Koepsell
- Subjects
Organic Cation Transport Proteins ,Physiology ,Blotting, Western ,Nephron ,Biology ,Kidney ,Cell membrane ,Rats, Sprague-Dawley ,medicine ,Animals ,Tissue Distribution ,RNA, Messenger ,In Situ Hybridization ,Organic cation transport proteins ,Reabsorption ,Cell Membrane ,Organic Cation Transporter 1 ,Kidney metabolism ,Membrane Proteins ,Organic Cation Transporter 2 ,Transporter ,Immunohistochemistry ,Rats ,medicine.anatomical_structure ,Biochemistry ,Renal physiology ,biology.protein ,Carrier Proteins - Abstract
Renal excretion and reabsorption of organic cations are mediated by electrogenic and electroneutral organic cation transporters, which belong to a recently discovered family of polyspecific transporters. These transporters are electrogenic and exhibit differences in substrate specificity. In rat, the renal expression of the polyspecific cation transporters rOCT1 and rOCT2 was investigated. By in situ hybridization, significant amounts of both rOCT1 and rOCT2 mRNA were detected in S1, S2, and S3 segments of proximal tubules. By immunohistochemistry, expression of the rOCT1 protein was mainly observed in S1 and S2 segments of proximal tubules, with lower expression levels in the S3 segments. At variance, rOCT2 protein was mainly expressed in the S2 and S3 segments. Both transporters were localized to the basolateral cell membrane. Neither rOCT1 nor rOCT2 was detected in the vasculature, the glomeruli, and nephron segments other than proximal tubules. The data suggest that rOCT1 and rOCT2 are responsible for basolateral cation uptake in the proximal tubule, which represents the first step in cation secretion.
- Published
- 2000
47. Localization of epithelial sodium channel and aquaporin-2 in rabbit kidney cortex
- Author
-
Bernard C. Rossier, Dominique Loffing-Cueni, Johannes Loffing, Brigitte Kaissling, Mark A. Knepper, Steven C. Hebert, Beatriz R. Olson, and Andreas Macher
- Subjects
Epithelial sodium channel ,Male ,medicine.medical_specialty ,Calbindins ,Kidney Cortex ,Physiology ,Sodium-Potassium-Chloride Symporters ,Renal cortex ,Sodium Chloride Symporter Inhibitors ,Aquaporin ,Nephron ,Biology ,urologic and male genital diseases ,Aquaporins ,Benzothiadiazines ,Sodium Channels ,S100 Calcium Binding Protein G ,Internal medicine ,medicine ,Animals ,Tissue Distribution ,Diuretics ,Epithelial Sodium Channels ,Bumetanide ,Aquaporin 2 ,urogenital system ,Reabsorption ,respiratory system ,Apical membrane ,Connecting tubule ,Aquaporin 6 ,Cell biology ,Rats ,Proton-Translocating ATPases ,Endocrinology ,medicine.anatomical_structure ,Female ,Rabbits ,Carrier Proteins ,hormones, hormone substitutes, and hormone antagonists - Abstract
The amiloride-sensitive epithelial sodium channel (ENaC) and the vasopressin-dependent water channel aquaporin-2 (AQP2) mediate mineralocorticoid-regulated sodium- and vasopressin-regulated water reabsorption, respectively. Distributions of ENaC and AQP2 have been shown by immunohistochemistry in rats. Functional data from rabbits suggest a different distribution pattern of these channels than in rats. We studied, by immunohistochemistry in the rabbit kidney cortex, the distributions of ENaC and AQP2, in conjunction with marker proteins for distal segments. In rabbit cortex ENaC is restricted to the connecting tubule (CNT) cells and cortical collecting duct (CCD) cells. The intracellular distribution of ENaC shifts from the apical membrane in the most upstream CNT cells to a cytoplasmic location further downstream in the CNT and in the CCD cells. AQP2 is detected in the CCD cells exclusively. The anatomic subdivisions in the rabbit distal nephron coincide exactly with distributions of apical transport systems. The differences between rabbits and rats in the distribution patterns of ENaC and AQP2 may explain functional differences in renal salt and water handling between these species.
- Published
- 2000
48. Butyrate increases apical membrane CFTR but reduces chloride secretion in MDCK cells
- Author
-
Bruce A. Stanton, Jan Loffing, Dominique Loffing-Cueni, Bryan D. Moyer, and Donna Reynolds
- Subjects
Physiology ,Recombinant Fusion Proteins ,Cell ,Green Fluorescent Proteins ,Cystic Fibrosis Transmembrane Conductance Regulator ,Butyrate ,Kidney ,Cystic fibrosis ,Cell Line ,Cell membrane ,chemistry.chemical_compound ,Dogs ,Chlorides ,medicine ,Electrochemistry ,Animals ,biology ,Cell Membrane ,Electric Conductivity ,Sodium butyrate ,Intracellular Membranes ,Apical membrane ,medicine.disease ,Molecular biology ,Cystic fibrosis transmembrane conductance regulator ,Butyrates ,Luminescent Proteins ,medicine.anatomical_structure ,chemistry ,Biochemistry ,Urea cycle ,biology.protein ,Indicators and Reagents ,Sodium-Potassium-Exchanging ATPase ,Carrier Proteins - Abstract
Sodium butyrate and its derivatives are useful therapeutic agents for the treatment of genetic diseases including urea cycle disorders, sickle cell disease, thalassemias, and possibly cystic fibrosis (CF). Butyrate partially restores cAMP-activated Cl−secretion in CF epithelial cells by stimulating ΔF508 cystic fibrosis transmembrane conductance regulator (ΔF508-CFTR) gene expression and increasing the amount of ΔF508-CFTR in the plasma membrane. Because the effect of butyrate on Cl−secretion by renal epithelial cells has not been reported, we examined the effects of chronic butyrate treatment (15–18 h) on the function, expression, and localization of CFTR fused to the green fluorescent protein (GFP-CFTR) in stably transfected MDCK cells. We report that sodium butyrate reduced Cl−secretion across MDCK cells, yet increased apical membrane GFP-CFTR expression 25-fold and increased apical membrane Cl−currents 30-fold. Although butyrate also increased Na-K-ATPase protein expression twofold, the drug reduced the activity of the Na-K-ATPase by 55%. Our findings suggest that butyrate inhibits cAMP-stimulated Cl−secretion across MDCK cells in part by reducing the activity of the Na-K-ATPase.
- Published
- 1999
49. Thiazide treatment of rats provokes apoptosis in distal tubule cells
- Author
-
M. R. Kaplan, B. Kaissling, Ivan Hegyi, Johannes Loffing, Dominique Loffing-Cueni, Michel Le Hir, and Steven C. Hebert
- Subjects
Male ,Calbindins ,ATPase ,Receptors, Drug ,Sodium Chloride Symporter Inhibitors ,Apoptosis ,030204 cardiovascular system & hematology ,Kidney ,chemistry.chemical_compound ,0302 clinical medicine ,Solute Carrier Family 12, Member 3 ,Diuretics ,Fluorescent Antibody Technique, Indirect ,Kidney Tubules, Distal ,In Situ Hybridization ,0303 health sciences ,biology ,Symporters ,Immunohistochemistry ,Sodium Chloride Symporters ,3. Good health ,medicine.anatomical_structure ,Nephrology ,Calbindin 1 ,Metolazone ,medicine.drug ,medicine.medical_specialty ,In situ hybridization ,Benzothiadiazines ,03 medical and health sciences ,S100 Calcium Binding Protein G ,Internal medicine ,medicine ,Animals ,Distal convoluted tubule ,Rats, Wistar ,Thiazide ,030304 developmental biology ,Analysis of Variance ,Epithelium ,Rats ,body regions ,Microscopy, Electron ,Endocrinology ,chemistry ,biology.protein ,RNA ,Ca(2+) Mg(2+)-ATPase ,Cotransporter ,Carrier Proteins - Abstract
Thiazide treatment of rats provokes apoptosis in distal tubule cells. We studied the effects of inhibition of apical NaCl entry on the structural correlates for electrolyte transport in the distal convoluted tubule (DCT) of rats. Thiazide diuretics were used to block NaCl entry specifically in the DCT. Metolazone or hydrochlorothiazide (HCTZ) were applied for three days subcutaneously via osmotic minipumps. The renal epithelial structure of control and treated rats was studied by light and electron microscopy. Distribution of the thiazide-sensitive NaCl cotransporter (rTSC1), calbindin D 28K and Ca 2+ -Mg 2+ -ATPase was examined by immunohistochemistry, and the content of rTSC1 transcripts by Northern blot and in situ hybridization. In treated rats the DCT epithelium had lost the structural characteristics of electrolyte transporting epithelia and the cells were in different stages of apoptosis. In damaged cells calbindin D 28K and Ca 2+ -Mg 2+ -ATPase were strongly decreased; the rTSC1 was shifted from the luminal membrane to the basal cell half and was found additionally in small membrane vesicles in intercellular and peritubular spaces. Transcripts of rTSC1 were drastically reduced in homogenates of kidney cortex and almost absent in damaged DCT cells. All other tubular segments were unaffected by the treatment. Focal inflammatory infiltrates were found to be specifically surrounding DCT profiles. Thus, inhibition by thiazides of apical NaCl entry into DCT cells is associated with apoptosis of DCT cells and focal peritubular inflammation.
- Published
- 1996
50. OS058. Aldosterone deficiency adversely affects pregnancy outcome in mice
- Author
-
M. Di Chiara, Abhijeet Todkar, Markus G. Mohaupt, Carsten A. Wagner, Dominique Loffing-Cueni, Carla Bettoni, and Jan Loffing
- Subjects
Aldosterone synthase ,medicine.medical_specialty ,Fetus ,Pregnancy ,Aldosterone ,biology ,Offspring ,business.industry ,Obstetrics and Gynecology ,medicine.disease ,Preeclampsia ,chemistry.chemical_compound ,Endocrinology ,Blood pressure ,chemistry ,Internal medicine ,Internal Medicine ,medicine ,biology.protein ,Gestation ,business - Abstract
Introduction In pregnancy, plasma volume is expanded due to high aldosterone levels to support placental perfusion and fetal nutrition. Inadequately low aldosterone levels as present in preeclampsia, a life-threatening disease for both mother and child, are discussed to be involved in its pathogenesis or severity. Objectives We used aldosterone synthase deficient ( AS −/− ) mice to test whether the absence of aldosterone is sufficient to impair pregnancy or even to cause preeclampsia. Methods AS −/− and AS +/+ females were mated with AS +/+ and AS −/− males, respectively, always generating AS +/− offspring. Blood pressure was measured by tail cuff, fetal and placental number and size as well as placental histology were assessed. Placental expression of HIF-1 α and angiogenic factors was assessed by semiquantitative RT-PCR. Results With maternal aldosterone deficiency in AS −/− mice, systolic blood pressure was low before and further reduced during pregnancy and with no increase in proteinuria. Yet, AS −/− had smaller litters due to loss of fetuses as indicated by a high number of necrotic placentas with massive lymphocyte infiltrations at gestational day 18. Surviving fetuses and their placentas from AS −/− females were smaller. High salt diet before and during pregnancy increased systolic blood pressure only before pregnancy in both genotypes and reduced blood pressure during late pregnancy as compared to normal salt controls. Litter size from AS −/− was slightly improved and the differences in placental and fetal weights between AS +/+ and AS −/− mothers disappeared. Overall an increased placental efficiency was observed in both groups. Conclusion Our results demonstrate that aldosterone deficiency has profound adverse effects on placental function. High dietary salt intake improved placental function and lowered blood pressure in wild-type mice. In this animal model, aldosterone deficiency did not cause preeclampsia.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.