Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
Acta Physiol (Oxf) ; 239(2): e14046, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37665159

ABSTRACT

OBJECTIVE: To understand the mechanisms involved in the response to a low-K+ diet (LK), we investigated the role of the growth factor GDF15 and the ion pump H,K-ATPase type 2 (HKA2) in this process. METHODS: Male mice of different genotypes (WT, GDF15-KO, and HKA2-KO) were fed an LK diet for different periods of time. We analyzed GDF15 levels, metabolic and physiological parameters, and the cellular composition of collecting ducts. RESULTS: Mice fed an LK diet showed a 2-4-fold increase in plasma and urine GDF15 levels. Compared to WT mice, GDF15-KO mice rapidly developed hypokalemia due to impaired renal adaptation. This is related to their 1/ inability to increase the number of type A intercalated cells (AIC) and 2/ absence of upregulation of H,K-ATPase type 2 (HKA2), the two processes responsible for K+ retention. Interestingly, we showed that the GDF15-mediated proliferative effect on AIC was dependent on the ErbB2 receptor and required the presence of HKA2. Finally, renal leakage of K+ induced a reduction in muscle mass in GDF15-KO mice fed LK diet. CONCLUSIONS: In this study, we showed that GDF15 and HKA2 are linked and play a central role in the response to K+ restriction by orchestrating the modification of the cellular composition of the collecting duct.

2.
JCI Insight ; 6(15)2021 08 09.
Article in English | MEDLINE | ID: mdl-34166227

ABSTRACT

Idiopathic nephrotic syndrome (INS) is characterized by proteinuria and renal sodium retention leading to edema. This sodium retention is usually attributed to epithelial sodium channel (ENaC) activation after plasma aldosterone increase. However, most nephrotic patients show normal aldosterone levels. Using a corticosteroid-clamped (CC) rat model of INS (CC-PAN), we showed that the observed electrogenic and amiloride-sensitive Na retention could not be attributed to ENaC. We then identified a truncated variant of acid-sensing ion channel 2b (ASIC2b) that induced sustained acid-stimulated sodium currents when coexpressed with ASIC2a. Interestingly, CC-PAN nephrotic ASIC2b-null rats did not develop sodium retention. We finally showed that the expression of the truncated ASIC2b in the kidney was dependent on the presence of albumin in the tubule lumen and activation of ERK in renal cells. Finally, the presence of ASIC2 mRNA was also detected in kidney biopsies from patients with INS but not in any of the patients with other renal diseases. We have therefore identified a variant of ASIC2b responsible for the renal Na retention in the pathological context of INS.


Subject(s)
Acid Sensing Ion Channels/metabolism , Kidney , MAP Kinase Signaling System , Nephrotic Syndrome , Sodium Channels/metabolism , Sodium , Albumins/metabolism , Animals , Disease Models, Animal , Gene Expression Profiling , Homeostasis , Kidney/metabolism , Kidney/pathology , Nephrotic Syndrome/blood , Nephrotic Syndrome/metabolism , Proteinuria/metabolism , Rats , Sodium/blood , Sodium/metabolism
3.
Acta Physiol (Oxf) ; 232(3): e13661, 2021 07.
Article in English | MEDLINE | ID: mdl-33840159

ABSTRACT

AIM: Type A intercalated cells of the renal collecting duct participate in the maintenance of the acid/base balance through their capacity to adapt proton secretion to homeostatic requirements. We previously showed that increased proton secretion stems in part from the enlargement of the population of proton secreting cells in the outer medullary collecting duct through division of fully differentiated cells, and that this response is triggered by growth/differentiation factor 15. This study aimed at deciphering the mechanism of acid load-induced secretion of Gdf15 and its mechanism of action. METHODS: We developed an original method to evaluate the proliferation of intercalated cells and applied it to genetically modified or pharmacologically treated mice under basal and acid-loaded conditions. RESULTS: Gdf15 is secreted by principal cells of the collecting duct in response to the stimulation of vasopressin receptors. Vasopressin-induced production of cAMP triggers activation of AMP-stimulated kinases and of Na,K-ATPase, and induction of p53 and Gdf15. Gdf15 action on intercalated cells is mediated by ErbB2 receptors, the activation of which triggers the expression of cyclin d1, of p53 and anti-proliferative genes, and of Egr1. CONCLUSION: Acidosis-induced proliferation of intercalated cells results from a cross talk with principal cells which secrete Gdf15 in response to their stimulation by vasopressin. Thus, vasopressin is a major determinant of the collecting duct cellular homeostasis as it promotes proliferation of intercalated cells under acidosis conditions and of principal cells under normal acid-base status.


Subject(s)
Acidosis , Kidney Tubules, Collecting , Animals , Cell Proliferation , Mice , Nephrons , Sodium-Potassium-Exchanging ATPase
4.
EMBO Rep ; 22(5): e50766, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33749979

ABSTRACT

SIRT7 is a NAD+ -dependent deacetylase that controls important aspects of metabolism, cancer, and bone formation. However, the molecular targets and functions of SIRT7 in the kidney are currently unknown. In silico analysis of kidney transcripts of the BXD murine genetic reference population revealed a positive correlation between Sirt7 and Slc12a7 mRNA expression, suggesting a link between the corresponding proteins that these transcripts encode, SIRT7, and the K-Cl cotransporter KCC4, respectively. Here, we find that protein levels and activity of heterologously expressed KCC4 are significantly modulated depending on its acetylation status in Xenopus laevis oocytes. Moreover, SIRT7 interacts with KCC4 in a NAD+ -dependent manner and increases its stability and activity in HEK293 cells. Interestingly, metabolic acidosis increases SIRT7 expression in kidney, as occurs with KCC4. In contrast, total SIRT7-deficient mice present lower KCC4 expression and an exacerbated metabolic acidosis than wild-type mice during an ammonium chloride challenge. Altogether, our data suggest that SIRT7 interacts with, stabilizes and modulates KCC4 activity through deacetylation, and reveals a novel role for SIRT7 in renal physiology.


Subject(s)
Sirtuins , Symporters , Acetylation , Animals , HEK293 Cells , Humans , Kidney , Mice , Sirtuins/genetics , Sirtuins/metabolism , Symporters/genetics , Symporters/metabolism , K Cl- Cotransporters
5.
Dis Model Mech ; 14(5)2021 05 01.
Article in English | MEDLINE | ID: mdl-33737325

ABSTRACT

Heterozygous mutations in HNF1B cause the complex syndrome renal cysts and diabetes (RCAD), characterized by developmental abnormalities of the kidneys, genital tracts and pancreas, and a variety of renal, pancreas and liver dysfunctions. The pathogenesis underlying this syndrome remains unclear as mice with heterozygous null mutations have no phenotype, while constitutive/conditional Hnf1b ablation leads to more severe phenotypes. We generated a novel mouse model carrying an identified human mutation at the intron-2 splice donor site. Unlike heterozygous mice previously characterized, mice heterozygous for the splicing mutation exhibited decreased HNF1B protein levels and bilateral renal cysts from embryonic day 15, originated from glomeruli, early proximal tubules (PTs) and intermediate nephron segments, concurrently with delayed PT differentiation, hydronephrosis and rare genital tract anomalies. Consistently, mRNA sequencing showed that most downregulated genes in embryonic kidneys were primarily expressed in early PTs and the loop of Henle and involved in ion/drug transport, organic acid and lipid metabolic processes, while the expression of previously identified targets upon Hnf1b ablation, including cystic disease genes, was weakly or not affected. Postnatal analyses revealed renal abnormalities, ranging from glomerular cysts to hydronephrosis and, rarely, multicystic dysplasia. Urinary proteomics uncovered a particular profile predictive of progressive decline in kidney function and fibrosis, and displayed common features with a recently reported urine proteome in an RCAD pediatric cohort. Altogether, our results show that reduced HNF1B levels lead to developmental disease phenotypes associated with the deregulation of a subset of HNF1B targets. They further suggest that this model represents a unique clinical/pathological viable model of the RCAD disease.


Subject(s)
Central Nervous System Diseases/genetics , Dental Enamel/abnormalities , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/genetics , Genes, Developmental , Haploinsufficiency/genetics , Hepatocyte Nuclear Factor 1-beta/genetics , Kidney Diseases, Cystic/genetics , Animals , Animals, Newborn , Cell Polarity , Central Nervous System Diseases/pathology , Cilia/pathology , Dental Enamel/pathology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Embryo, Mammalian/pathology , Gene Dosage , Gene Expression Profiling , Heterozygote , Humans , Hydronephrosis/complications , Kidney Diseases, Cystic/pathology , Kidney Glomerulus/pathology , Kidney Tubules/pathology , Mice, Inbred C57BL , Mutation/genetics , Nephrons/pathology , RNA Splicing/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Severity of Illness Index
6.
Healthc Q ; 22(SP): 82-95, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32049618

ABSTRACT

Senior healthcare leaders are the difference makers as key influencers in ushering in an organizational culture committed to patient safety. Although leaders at all levels are champions of transformation, leaders at the "top" have a unique opportunity - and a responsibility - to foster a culture that supports an organization on its journey to zero harm. Through a literature review of more than 60 resources and validation with thought leaders, national and provincial partners have developed a patient safety culture bundle for CEOs and senior healthcare leaders. The bundle is based on a set of evidence-based practices that must be applied collectively to establish and sustain a culture of quality and safety in order to deliver safe care.


Subject(s)
Organizational Culture , Patient Safety , Safety Management/organization & administration , Evidence-Based Practice , Humans , Leadership , Medical Errors/prevention & control
7.
J Am Soc Nephrol ; 30(12): 2322-2336, 2019 12.
Article in English | MEDLINE | ID: mdl-31558682

ABSTRACT

BACKGROUND: Lithium, mainstay treatment for bipolar disorder, causes nephrogenic diabetes insipidus and hypercalcemia in about 20% and 10% of patients, respectively, and may lead to acidosis. These adverse effects develop in only a subset of patients treated with lithium, suggesting genetic factors play a role. METHODS: To identify susceptibility genes for lithium-induced adverse effects, we performed a genome-wide association study in mice, which develop such effects faster than humans. On day 8 and 10 after assigning female mice from 29 different inbred strains to normal chow or lithium diet (40 mmol/kg), we housed the animals for 48 hours in metabolic cages for urine collection. We also collected blood samples. RESULTS: In 17 strains, lithium treatment significantly elevated urine production, whereas the other 12 strains were not affected. Increased urine production strongly correlated with lower urine osmolality and elevated water intake. Lithium caused acidosis only in one mouse strain, whereas hypercalcemia was found in four strains. Lithium effects on blood pH or ionized calcium did not correlate with effects on urine production. Using genome-wide association analyses, we identified eight gene-containing loci, including a locus containing Acer2, which encodes a ceramidase and is specifically expressed in the collecting duct. Knockout of Acer2 led to increased susceptibility for lithium-induced diabetes insipidus development. CONCLUSIONS: We demonstrate that genome-wide association studies in mice can be used successfully to identify susceptibility genes for development of lithium-induced adverse effects. We identified Acer2 as a first susceptibility gene for lithium-induced diabetes insipidus in mice.


Subject(s)
Alkaline Ceramidase/genetics , Diabetes Insipidus, Nephrogenic/genetics , Lithium Chloride/toxicity , Acid-Base Equilibrium/physiology , Acidosis/chemically induced , Acidosis/genetics , Animals , Diabetes Insipidus, Nephrogenic/chemically induced , Dinoprostone/urine , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Hematocrit , Hypercalcemia/chemically induced , Hypercalcemia/genetics , Kidney Tubules, Collecting/metabolism , Mice , Mice, Inbred Strains , Mice, Knockout , Nephrons/metabolism , RNA, Messenger/biosynthesis , Sodium/blood , Species Specificity
8.
Physiol Rep ; 7(15): e14177, 2019 08.
Article in English | MEDLINE | ID: mdl-31397090

ABSTRACT

The mineralocorticoid hormone aldosterone plays a crucial role in the control of Na+ and K+ balance, blood volume, and arterial blood pressure, by acting in the aldosterone-sensitive distal nephron (ASDN) and stimulating a complex transcriptional, translational, and cellular program. Because the complexity of the aldosterone response is still not fully appreciated, we aimed at identifying new elements in this pathway. Here, we demonstrate that the expression of the proto-oncogene PIM3 (Proviral Integration Site of Moloney Murine Leukemia Virus 3), a serine/threonine kinase belonging to the calcium/calmodulin-regulated group of kinases, is stimulated by aldosterone in vitro (mCCDcl1 cells), ex vivo (mouse kidney slices), and in vivo in mice. Characterizing a germline Pim3-/- mouse model, we found that these mice have an upregulated Renin-Angiotensin-Aldosterone System (RAAS), with high circulating aldosterone and plasma renin activity levels on both standard or Na+ -deficient diet. Surprisingly, we did not observe any obvious salt-losing phenotype in Pim3 KO mice as shown by normal blood pressure, plasma and urinary electrolytes, as well as unchanged expression levels of the major Na+ transport proteins. These observations suggest that the potential effects of the loss of the Pim3 gene are physiologically compensated. Indeed, the 2 other family members of the PIM kinase family, PIM1 and PIM2 are upregulated in the kidney of Pim3-/- mice, and may therefore be involved in such compensation. In conclusion, our data demonstrate that the PIM3 kinase is a novel aldosterone-induced protein, but its precise role in aldosterone-dependent renal homeostasis remains to be determined.


Subject(s)
Aldosterone/pharmacology , Kidney/drug effects , Nephrons/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Kidney/metabolism , Male , Mice, Inbred C57BL , Nephrons/drug effects , Nuclear Proteins/genetics , Phenotype , Sodium/metabolism , Transcription Factors/genetics
9.
Am J Physiol Renal Physiol ; 317(2): F435-F443, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31188029

ABSTRACT

We have recently reported that type A intercalated cells of the collecting duct secrete Na+ by a mechanism coupling the basolateral type 1 Na+-K+-2Cl- cotransporter with apical type 2 H+-K+-ATPase (HKA2) functioning under its Na+/K+ exchange mode. The first aim of the present study was to evaluate whether this secretory pathway is a target of atrial natriuretic peptide (ANP). Despite hyperaldosteronemia, metabolic acidosis is not associated with Na+ retention. The second aim of the present study was to evaluate whether ANP-induced stimulation of Na+ secretion by type A intercalated cells might account for mineralocorticoid escape during metabolic acidosis. In Xenopus oocytes expressing HKA2, cGMP, the second messenger of ANP, increased the membrane expression, activity, and Na+-transporting rate of HKA2. Feeding mice with a NH4Cl-enriched diet increased urinary excretion of aldosterone and induced a transient Na+ retention that reversed within 3 days. At that time, expression of ANP mRNA in the collecting duct and urinary excretion of cGMP were increased. Reversion of Na+ retention was prevented by treatment with an inhibitor of ANP receptors and was absent in HKA2-null mice. In conclusion, paracrine stimulation of HKA2 by ANP is responsible for the escape of the Na+-retaining effect of aldosterone during metabolic acidosis.


Subject(s)
Acid-Base Equilibrium , Acidosis/enzymology , Atrial Natriuretic Factor/metabolism , H(+)-K(+)-Exchanging ATPase/metabolism , Kidney Tubules, Collecting/enzymology , Sodium/urine , Acidosis/genetics , Acidosis/physiopathology , Acidosis/urine , Adaptation, Physiological , Aldosterone/urine , Animals , Cyclic GMP/urine , Female , H(+)-K(+)-Exchanging ATPase/deficiency , H(+)-K(+)-Exchanging ATPase/genetics , Hydrogen-Ion Concentration , Mice, Inbred C57BL , Mice, Knockout , Paracrine Communication , Rats , Signal Transduction , Xenopus laevis
10.
PLoS One ; 12(9): e0183774, 2017.
Article in English | MEDLINE | ID: mdl-28931009

ABSTRACT

Regulation of our water homeostasis is fine-tuned by dynamic translocation of Aquaporin-2 (AQP2)-bearing vesicles to and from the plasma membrane of renal principal cells. Whereas binding of vasopressin to its type-2 receptor initiates a cAMP-protein kinase A cascade and AQP2 translocation to the apical membrane, this is counteracted by protein kinase C-activating hormones, resulting in ubiquitination-dependent internalization of AQP2. The proteins targeting AQP2 for ubiquitin-mediated degradation are unknown. In collecting duct mpkCCD cells, siRNA knockdown of NEDD4 and NEDD4L E3 ligases yielded increased AQP2 abundance, but they did not bind AQP2. Membrane Yeast Two-Hybrid assays using full-length AQP2 as bait, identified NEDD4 family interacting protein 2 (NDFIP2) to bind AQP2. NDFIP2 and its homologue NDFIP1 have PY motifs by which they bind NEDD4 family members and bring them close to target proteins. In HEK293 cells, NDFIP1 and NDFIP2 bound AQP2 and were essential for NEDD4/NEDD4L-mediated ubiquitination and degradation of AQP2, an effect not observed with PY-lacking NDFIP1/2 proteins. In mpkCCD cells, downregulation of NDFIP1, NEDD4 and NEDD4L, but not NDFIP2, increased AQP2 abundance. In mouse kidney, Ndfip1 and Ndfip2 mRNA distribution was similar and high in proximal tubules and collecting ducts, which was also found for NDFIP1 proteins. Our results reveal that NEDD4/NEDD4L mediate ubiquitination and degradation of AQP2, but that NDFIP proteins are needed to connect NEDD4/NEDD4L to AQP2. As NDFIP1/2 bind many NEDD4 family E3 ligases, which are implicated in several cellular processes, NDFIP1/2 may be the missing link for AQP2 ubiquitination and degradation from different subcellular locations.


Subject(s)
Aquaporin 2/metabolism , Carrier Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Membrane Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Carrier Proteins/genetics , Cell Line , Down-Regulation , Endosomal Sorting Complexes Required for Transport/antagonists & inhibitors , Endosomal Sorting Complexes Required for Transport/genetics , HEK293 Cells , Humans , Immunoprecipitation , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Nedd4 Ubiquitin Protein Ligases , Nephrons/metabolism , Protein Binding , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitination
11.
Plant Biotechnol J ; 15(3): 285-296, 2017 03.
Article in English | MEDLINE | ID: mdl-27483398

ABSTRACT

Influenza virus-like particles (VLPs) have been shown to induce a safe and potent immune response through both humoral and cellular responses. They represent promising novel influenza vaccines. Plant-based biotechnology allows for the large-scale production of VLPs of biopharmaceutical interest using different model organisms, including Nicotiana benthamiana plants. Through this platform, influenza VLPs bud from the plasma membrane and accumulate between the membrane and the plant cell wall. To design and optimize efficient production processes, a better understanding of the plant cell wall composition of infiltrated tobacco leaves is a major interest for the plant biotechnology industry. In this study, we have investigated the alteration of the biochemical composition of the cell walls of N. benthamiana leaves subjected to abiotic and biotic stresses induced by the Agrobacterium-mediated transient transformation and the resulting high expression levels of influenza VLPs. Results show that abiotic stress due to vacuum infiltration without Agrobacterium did not induce any detectable modification of the leaf cell wall when compared to non infiltrated leaves. In contrast, various chemical changes of the leaf cell wall were observed post-Agrobacterium infiltration. Indeed, Agrobacterium infection induced deposition of callose and lignin, modified the pectin methylesterification and increased both arabinosylation of RG-I side chains and the expression of arabinogalactan proteins. Moreover, these modifications were slightly greater in plants expressing haemagglutinin-based VLP than in plants infiltrated with the Agrobacterium strain containing only the p19 suppressor of silencing.


Subject(s)
Agrobacterium/metabolism , Biotechnology/methods , Cell Wall/metabolism , Hemagglutinins/metabolism , Nicotiana/metabolism , Agrobacterium/genetics , Hemagglutinins/genetics , Influenza Vaccines/genetics , Influenza Vaccines/metabolism , Plants, Genetically Modified/genetics , Plants, Genetically Modified/metabolism , Nicotiana/genetics
12.
Sci Rep ; 6: 35215, 2016 10 14.
Article in English | MEDLINE | ID: mdl-27739476

ABSTRACT

Olfactory receptors (ORs) are G protein-coupled receptors which serve important sensory functions beyond their role as odorant detectors in the olfactory epithelium. Here we describe a novel role for one of these ORs, Olfr1393, as a regulator of renal glucose handling. Olfr1393 is specifically expressed in the kidney proximal tubule, which is the site of renal glucose reabsorption. Olfr1393 knockout mice exhibit urinary glucose wasting and improved glucose tolerance, despite euglycemia and normal insulin levels. Consistent with this phenotype, Olfr1393 knockout mice have a significant decrease in luminal expression of Sglt1, a key renal glucose transporter, uncovering a novel regulatory pathway involving Olfr1393 and Sglt1. In addition, by utilizing a large scale screen of over 1400 chemicals we reveal the ligand profile of Olfr1393 for the first time, offering new insight into potential pathways of physiological regulation for this novel signaling pathway.


Subject(s)
Glucose/metabolism , Kidney Tubules, Proximal/metabolism , Olfactory Receptor Neurons/metabolism , Animals , Cell Line , Dogs , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Olfactory Mucosa/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Odorant/metabolism , Signal Transduction/physiology , Sodium-Glucose Transporter 1/metabolism
13.
J Physiol ; 594(20): 5991-6008, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27412964

ABSTRACT

KEY POINTS: The cortical collecting duct (CCD) plays an essential role in sodium homeostasis by fine-tuning the amount of sodium that is excreted in the urine. Ex vivo, the microperfused CCD reabsorbs sodium in the absence of lumen-to-bath concentration gradients. In the present study, we show that, in the presence of physiological lumen-to-bath concentration gradients, and in the absence of endocrine, paracrine and neural regulation, the mouse CCD secretes sodium, which represents a paradigm shift. This secretion occurs via the paracellular route, as well as a transcellular pathway that is energized by apical H+ /K+ -ATPase type 2 pumps operating as Na+ /K+ exchangers. The newly identified transcellular secretory pathway represents a physiological target for the regulation of sodium handling and for anti-hypertensive therapeutic agents. ABSTRACT: In vitro microperfusion experiments have demonstrated that cortical collecting ducts (CCDs) reabsorb sodium via principal and type B intercalated cells under sodium-depleted conditions and thereby contribute to sodium and blood pressure homeostasis. However, these experiments were performed in the absence of the transepithelial ion concentration gradients that prevail in vivo and determine paracellular transport. The present study aimed to characterize Na+ , K+ and Cl- fluxes in the mouse CCD in the presence of physiological transepithelial concentration gradients. For this purpose, we combined in vitro measurements of ion fluxes across microperfused CCDs of sodium-depleted mice with the predictions of a mathematical model. When NaCl transport was inhibited in all cells, CCDs secreted Na+ and reabsorbed K+ ; Cl- transport was negligible. Removing inhibitors of type A and B intercalated cells increased Na+ secretion in wild-type (WT) mice but not in H+ /K+ -ATPase type 2 (HKA2) knockout mice. Further inhibition of basolateral NaCl entry via the Na+ -K+ -2Cl- cotransporter in type A intercalated cells reduced Na+ secretion in WT mice to the levels observed in HKA2-/- mice. With no inhibitors, WT mouse CCDs still secreted Na+ and reabsorbed K+ . In vivo, HKA2-/- mice excreted less Na+ than WT mice after switching to a high-salt diet. Taken together, our results indicate that type A intercalated cells secrete Na+ via basolateral Na+ -K+ -2Cl- cotransporters in tandem with apical HKA2 pumps. They also suggest that the CCD can mediate overall Na+ secretion, and that its ability to reabsorb NaCl in vivo depends on the presence of acute regulatory factors.


Subject(s)
Epithelium/metabolism , Kidney Tubules, Collecting/metabolism , Animals , Biological Transport/physiology , Chlorides/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Potassium/metabolism , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism
14.
J Biol Chem ; 291(21): 11105-13, 2016 May 20.
Article in English | MEDLINE | ID: mdl-27033704

ABSTRACT

Water accumulation in the interstitium (edema) and the peritoneum (ascites) of nephrotic patients is classically thought to stem from the prevailing low plasma albumin concentration and the decreased transcapillary oncotic pressure gradient. However, several clinical and experimental observations suggest that it might also stem from changes in capillary permeability. We addressed this hypothesis by studying the peritoneum permeability of rats with puromycin aminonucleoside-induced nephrotic syndrome. The peritoneum of puromycin aminonucleoside rats displayed an increase in the water filtration coefficient of paracellular and transcellular pathways, and a decrease in the reflection coefficient to proteins. It also displayed oxidative stress and subsequent activation of NF-κB. Scavenging of reactive oxygen species and inhibition of NF-κB prevented the changes in the water permeability and reflection coefficient to proteins and reduced the volume of ascites by over 50%. Changes in water permeability were associated with the overexpression of the water channel aquaporin 1, which was prevented by reactive oxygen species scavenging and inhibition of NF-κB. In conclusion, nephrotic syndrome is associated with an increased filtration coefficient of the peritoneum and a decreased reflection coefficient to proteins. These changes, which account for over half of ascite volume, are triggered by oxidative stress and subsequent activation of NF-κB.


Subject(s)
Ascites , NF-kappa B/metabolism , Nephrotic Syndrome , Oxidative Stress/drug effects , Peritoneum , Puromycin Aminonucleoside/adverse effects , Animals , Aquaporin 1/metabolism , Ascites/chemically induced , Ascites/metabolism , Ascites/pathology , Humans , Male , Nephrotic Syndrome/chemically induced , Nephrotic Syndrome/metabolism , Nephrotic Syndrome/pathology , Peritoneum/metabolism , Peritoneum/pathology , Puromycin Aminonucleoside/pharmacology , Rats , Rats, Sprague-Dawley
15.
J Am Soc Nephrol ; 25(2): 250-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24179170

ABSTRACT

In relation to dietary Na(+) intake and aldosterone levels, collecting duct principal cells are exposed to large variations in Na(+) transport. In these cells, Na(+) crosses the apical membrane via epithelial Na(+) channels (ENaC) and is extruded into the interstitium by Na,K-ATPase. The activity of ENaC and Na,K-ATPase must be highly coordinated to accommodate variations in Na(+) transport and minimize fluctuations in intracellular Na(+) concentration. We hypothesized that, independent of hormonal stimulus, cross-talk between ENaC and Na,K-ATPase coordinates Na(+) transport across apical and basolateral membranes. By varying Na(+) intake in aldosterone-clamped rats and overexpressing γ-ENaC or modulating apical Na(+) availability in cultured mouse collecting duct cells, enhanced apical Na(+) entry invariably led to increased basolateral Na,K-ATPase expression and activity. In cultured collecting duct cells, enhanced apical Na(+) entry increased the basolateral cell surface expression of Na,K-ATPase by inhibiting p38 kinase-mediated endocytosis of Na,K-ATPase. Our results reveal a new role for p38 kinase in mediating cross-talk between apical Na(+) entry via ENaC and its basolateral exit via Na,K-ATPase, which may allow principal cells to maintain intracellular Na(+) concentrations within narrow limits.


Subject(s)
Epithelial Sodium Channels/physiology , Kidney Tubules, Collecting/metabolism , MAP Kinase Signaling System/physiology , Sodium-Potassium-Exchanging ATPase/physiology , Sodium/metabolism , p38 Mitogen-Activated Protein Kinases/physiology , AMP-Activated Protein Kinases/physiology , Aldosterone/physiology , Animals , Basement Membrane/metabolism , Biological Transport, Active/physiology , Cell Line, Transformed , Cell Membrane/metabolism , Cell Polarity , Endocytosis/physiology , Enzyme Induction , Epithelial Sodium Channels/biosynthesis , Epithelial Sodium Channels/genetics , Homeostasis/physiology , Intracellular Fluid/metabolism , Ion Transport/physiology , Kidney Tubules, Collecting/cytology , Lysosomes/metabolism , MAP Kinase Signaling System/drug effects , Male , Mice , Rats , Rats, Sprague-Dawley , Sodium-Potassium-Exchanging ATPase/biosynthesis , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
16.
Proc Natl Acad Sci U S A ; 110(35): 14366-71, 2013 Aug 27.
Article in English | MEDLINE | ID: mdl-23940364

ABSTRACT

Large deletions in the first intron of the With No lysine (K) 1 (WNK1) gene are responsible for Familial Hyperkalemic Hypertension (FHHt), a rare form of human hypertension associated with hyperkalemia and hyperchloremic metabolic acidosis. We generated a mouse model of WNK1-associated FHHt to explore the consequences of this intronic deletion. WNK1(+/FHHt) mice display all clinical and biological signs of FHHt. This phenotype results from increased expression of long WNK1 (L-WNK1), the ubiquitous kinase isoform of WNK1, in the distal convoluted tubule, which in turn, stimulates the activity of the Na-Cl cotransporter. We also show that the activity of the epithelial sodium channel is not altered in FHHt mice, suggesting that other mechanisms are responsible for the hyperkalemia and acidosis in this model. Finally, we observe a decreased expression of the renal outer medullary potassium channel in the late distal convoluted tubule of WNK1(+/FHHt) mice, which could contribute to the hyperkalemia. In summary, our study provides insights into the in vivo mechanisms underlying the pathogenesis of WNK1-mediated FHHt and further corroborates the importance of WNK1 in ion homeostasis and blood pressure.


Subject(s)
Kidney Tubules, Distal/metabolism , Protein Serine-Threonine Kinases/genetics , Pseudohypoaldosteronism/genetics , Animals , Epithelial Sodium Channels/metabolism , Gene Deletion , Mice , Mice, Transgenic , Minor Histocompatibility Antigens , Potassium Channels, Inwardly Rectifying/genetics , Pseudohypoaldosteronism/metabolism , WNK Lysine-Deficient Protein Kinase 1
17.
J Clin Invest ; 123(7): 3166-71, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23934124

ABSTRACT

Paracrine communication between different parts of the renal tubule is increasingly recognized as an important determinant of renal function. Previous studies have shown that changes in dietary acid-base load can reverse the direction of apical α-ketoglutarate (αKG) transport in the proximal tubule and Henle's loop from reabsorption (acid load) to secretion (base load). Here we show that the resulting changes in the luminal concentrations of αKG are sensed by the αKG receptor OXGR1 expressed in the type B and non-A-non-B intercalated cells of the connecting tubule (CNT) and the cortical collecting duct (CCD). The addition of 1 mM αKG to the tubular lumen strongly stimulated Cl(-)-dependent HCO(3)(-) secretion and electroneutral transepithelial NaCl reabsorption in microperfused CCDs of wild-type mice but not Oxgr1(-/-) mice. Analysis of alkali-loaded mice revealed a significantly reduced ability of Oxgr1(-/-) mice to maintain acid-base balance. Collectively, these results demonstrate that OXGR1 is involved in the adaptive regulation of HCO(3)(-) secretion and NaCl reabsorption in the CNT/CCD under acid-base stress and establish αKG as a paracrine mediator involved in the functional coordination of the proximal and the distal parts of the renal tubule.


Subject(s)
Acid-Base Equilibrium , Ketoglutaric Acids/urine , Kidney Tubules, Collecting/physiology , Paracrine Communication , Animals , Bicarbonates/metabolism , In Vitro Techniques , Ketoglutaric Acids/blood , Male , Mice , Mice, Knockout , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/metabolism , Sodium Chloride/metabolism
18.
Am J Physiol Renal Physiol ; 305(7): F1053-63, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23884139

ABSTRACT

Albuminuria is strongly associated with progressive kidney tubulo-interstitial damage and chronic kidney disease (CKD) progression. In proteinuric nephropathies, albumin reabsorption by the proximal tubule is saturated and the distal nephron is exposed to high concentrations of luminal albumin that may produce adverse effects. Since proximal tubular cells exposed to albuminuria exhibit a proinflammatory and profibrotic response, we assessed the effect of albuminuria in the collecting duct (CD). With the use of kidney sections and isolated cortical CDs (CCDs) from puromycin-aminonucleoside-induced nephrotic rats (PAN rats) exhibiting proteinuria, immunofluorescence microscopy revealed internalized albumin in CD cells. In these proteinuric rats, increased expression levels of cytokines and profibrotic signaling markers were detected in isolated CCDs and bands of inflammatory fibrosis could be observed around CDs. Albumin endocytosis was confirmed by FITC-albumin uptake in cultured murine CCD (mCCDcl1) cells. Exposure of mCCDcl1 cells to albumin induced NF-κB activation as assessed by luciferase reporter gene assay, nuclear translocation of NF-κB p65 subunit, and increased NF-κB target gene expression. Moreover, albuminuria-like condition results in transforming growth factor-ß1 (TGF-ß1) overexpression and the upregulation of profibrotic signaling markers such as Snail or vimentin via an autocrine mechanism. In mCCDcl1 cells, neutrophil gelatinase-associated lipocalin (NGAL)/lipocalin-2/24p3 receptor (24p3R) mediates albumin endocytosis as well as activation of NF-κB and TGF-ß1 signaling pathways. Therefore, CD may play a key role in initiation and/or progression of inflammation and fibrosis in response to proteinuria.


Subject(s)
Acute-Phase Proteins/physiology , Albumins/metabolism , Albuminuria/metabolism , Albuminuria/pathology , Kidney Tubules, Collecting/pathology , Lipocalins/physiology , Oncogene Proteins/physiology , Albuminuria/complications , Animals , Cell Line , Endocytosis/physiology , Kidney Tubules, Collecting/metabolism , Lipocalin-2 , Male , Mice , NF-kappa B/metabolism , Nephritis/etiology , Nephritis/metabolism , Nephrosclerosis/etiology , Nephrosclerosis/metabolism , Rats , Rats, Wistar , Transforming Growth Factor beta1/metabolism
19.
J Biol Chem ; 288(20): 13936-13949, 2013 May 17.
Article in English | MEDLINE | ID: mdl-23519473

ABSTRACT

Dipeptidyl peptidases (DP) 8 and 9 are homologous, cytoplasmic N-terminal post-proline-cleaving enzymes that are anti-targets for the development of DP4 (DPPIV/CD26) inhibitors for treating type II diabetes. To date, DP8 and DP9 have been implicated in immune responses and cancer biology, but their pathophysiological functions and substrate repertoire remain unknown. This study utilizes terminal amine isotopic labeling of substrates (TAILS), an N-terminal positional proteomic approach, for the discovery of in vivo DP8 and DP9 substrates. In vivo roles for DP8 and DP9 in cellular metabolism and homeostasis were revealed via the identification of more than 29 candidate natural substrates and pathways affected by DP8/DP9 overexpression. Cleavage of 14 substrates was investigated in vitro; 9/14 substrates for both DP8 and DP9 were confirmed by MALDI-TOF MS, including two of high confidence, calreticulin and adenylate kinase 2. Adenylate kinase 2 plays key roles in cellular energy and nucleotide homeostasis. These results demonstrate remarkable in vivo substrate overlap between DP8/DP9, suggesting compensatory roles for these enzymes. This work provides the first global investigation into DP8 and DP9 substrates, providing a number of leads for future investigations into the biological roles and significance of DP8 and DP9 in human health and disease.


Subject(s)
Adenylate Kinase/metabolism , Calreticulin/metabolism , Dipeptidases/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Proteomics/methods , Amino Acid Sequence , Cations , Cell Line, Tumor , Cell Separation , Cytoplasm/metabolism , Energy Metabolism , Flow Cytometry , Homeostasis , Humans , Isotope Labeling , Mass Spectrometry , Molecular Sequence Data , Protein Structure, Tertiary , Substrate Specificity
20.
Pflugers Arch ; 465(8): 1149-58, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23440459

ABSTRACT

Renal K(+) retention is activated during pregnancy through a mechanism unknown to date. Here, we showed that the renal stimulation of H,K-ATPase type 2 (HKA2), whose expression was recently identified to be progesterone-dependent, is part of the mechanism favoring K(+) accumulation during gestation. Moreover, investigation of the gestational phenotype of HKA2-null mice compared to their wild-type (WT) littermate revealed a decrease in fertility (gestation was successful in 33 % of HKA2-null mice vs. 83 % of WT mice) and in litter size (6.5 ± 0.6 and 7.8 ± 0.4 fetuses per litter, respectively). We also observed that urinary K(+) excretion decreased by 20 % and plasma K(+) concentration rose slightly (11 %) in WT mice during gestation (relative to basal conditions). In contrast, the renal excretion of K(+) and plasma K(+) levels in HKA2-null mice remained constant during gestation, whereas fecal K(+) excretion increased. As a consequence, HKA2-null mice did not accumulate K(+) in their extracellular compartment as efficiently as WT mice did. Finally, the link between inefficient K(+) balance adaptations and gestational complications was established when we observed that these complications could be reversed with an increased K(+) uptake. Altogether, these results define a novel physiological role for the HKA2 transporter and uncover a link between K(+) metabolism and fertility.


Subject(s)
Fertility/physiology , H(+)-K(+)-Exchanging ATPase/metabolism , Homeostasis/physiology , Kidney/metabolism , Animals , Female , Kidney/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Potassium/metabolism , Potassium/urine , Pregnancy , Pregnancy Complications/metabolism , Pregnancy Complications/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...