Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Pflugers Arch ; 476(4): 533-543, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38110744

ABSTRACT

Pendrin (SLC26A4) is an anion exchanger from the SLC26 transporter family which is mutated in human patients affected by Pendred syndrome, an autosomal recessive disease characterized by sensoneurinal deafness and hypothyroidism. Pendrin is also expressed in the kidney where it mediates the exchange of internal HCO3- for external Cl- at the apical surface of renal type B and non-A non-B-intercalated cells. Studies using pendrin knockout mice have first revealed that pendrin is essential for renal base excretion. However, subsequent studies have demonstrated that pendrin also controls chloride absorption by the distal nephron and that this mechanism is critical for renal NaCl balance. Furthermore, pendrin has been shown to control vascular volume and ultimately blood pressure. This review summarizes the current knowledge about how pendrin is linking renal acid-base regulation to blood pressure control.


Subject(s)
Kidney , Nephrons , Animals , Mice , Humans , Blood Pressure/physiology , Sulfate Transporters , Kidney/metabolism , Nephrons/metabolism , Sodium Chloride , Chlorides/metabolism , Anion Transport Proteins/genetics
2.
Handb Exp Pharmacol ; 283: 35-58, 2024.
Article in English | MEDLINE | ID: mdl-36811727

ABSTRACT

The molecular basis of chloride transport varies all along the nephron depending on the tubular segments especially in the apical entry of the cell. The major chloride exit pathway during reabsorption is provided by two kidney-specific ClC chloride channels ClC-Ka and ClC-Kb (encoded by CLCNKA and CLCNKB gene, respectively) corresponding to rodent ClC-K1 and ClC-K2 (encoded by Clcnk1 and Clcnk2). These channels function as dimers and their trafficking to the plasma membrane requires the ancillary protein Barttin (encoded by BSND gene). Genetic inactivating variants of the aforementioned genes lead to renal salt-losing nephropathies with or without deafness highlighting the crucial role of ClC-Ka, ClC-Kb, and Barttin in the renal and inner ear chloride handling. The purpose of this chapter is to summarize the latest knowledge on renal chloride structure peculiarity and to provide some insight on the functional expression on the segments of the nephrons and on the related pathological effects.


Subject(s)
Chlorides , Kidney , Chlorides/metabolism , Cell Membrane/metabolism , Chloride Channels/genetics , Chloride Channels/metabolism
3.
J Med Genet ; 59(11): 1035-1043, 2022 11.
Article in English | MEDLINE | ID: mdl-35115415

ABSTRACT

BACKGROUND: Nephrolithiasis (NL) is a complex multifactorial disease affecting up to 10%-20% of the human population and causing a significant burden on public health systems worldwide. It results from a combination of environmental and genetic factors. Hyperoxaluria is a major risk factor for NL. METHODS: We used a whole exome-based approach in a patient with calcium oxalate NL. The effects of the mutation were characterised using cell culture and in silico analyses. RESULTS: We identified a rare heterozygous missense mutation (c.1519C>T/p.R507W) in the SLC26A6 gene that encodes a secretory oxalate transporter. This mutation cosegregated with hyperoxaluria in the family. In vitro characterisation of mutant SLC26A6 demonstrated that Cl--dependent oxalate transport was dramatically reduced because the mutation affects both SLC26A6 transport activity and membrane surface expression. Cotransfection studies demonstrated strong dominant-negative effects of the mutant on the wild-type protein indicating that the phenotype of patients heterozygous for this mutation may be more severe than predicted by haploinsufficiency alone. CONCLUSION: Our study is in line with previous observations made in the mouse showing that SLC26A6 inactivation can cause inherited enteric hyperoxaluria with calcium oxalate NL. Consistent with an enteric form of hyperoxaluria, we observed a beneficial effect of increasing calcium in the patient's diet to reduce urinary oxalate excretion.


Subject(s)
Antiporters , Hyperoxaluria , Nephrolithiasis , Sulfate Transporters , Humans , Antiporters/genetics , Calcium/metabolism , Calcium Oxalate/metabolism , Hyperoxaluria/complications , Hyperoxaluria/genetics , Mutation , Nephrolithiasis/genetics , Nephrolithiasis/complications , Nephrolithiasis/metabolism , Oxalates/metabolism , Sulfate Transporters/genetics
4.
Kidney Int ; 97(3): 452-455, 2020 03.
Article in English | MEDLINE | ID: mdl-32087886

ABSTRACT

Young onset distal tubular acidosis is a rare genetic disorder that can lead, if untreated, to many complications. Mutations in few genes account for almost half of the cases, whereas the molecular mechanisms accounting for the remaining cases are still unknown. The present study reports the use of whole-exome sequencing to identify new dRTA-causing genes and demonstrates that inactivating mutations in the ATP6V1C2 gene impair renal proton pump function.


Subject(s)
Acidosis, Renal Tubular , Vacuolar Proton-Translocating ATPases/genetics , Humans , Mutation , Exome Sequencing
7.
Kidney Int ; 94(3): 514-523, 2018 09.
Article in English | MEDLINE | ID: mdl-30146013

ABSTRACT

Pseudohypoaldosteronism type II (PHAII) is a genetic disease characterized by association of hyperkalemia, hyperchloremic metabolic acidosis, hypertension, low renin, and high sensitivity to thiazide diuretics. It is caused by mutations in the WNK1, WNK4, KLHL3 or CUL3 gene. There is strong evidence that excessive sodium chloride reabsorption by the sodium chloride cotransporter NCC in the distal convoluted tubule is involved. WNK4 is expressed not only in distal convoluted tubule cells but also in ß-intercalated cells of the cortical collecting duct. These latter cells exchange intracellular bicarbonate for external chloride through pendrin, and therefore, account for renal base excretion. However, these cells can also mediate thiazide-sensitive sodium chloride absorption when the pendrin-dependent apical chloride influx is coupled to apical sodium influx by the sodium-driven chloride/bicarbonate exchanger. Here we determine whether this system is involved in the pathogenesis of PHAII. Renal pendrin activity was markedly increased in a mouse model carrying a WNK4 missense mutation (Q562E) previously identified in patients with PHAII. The upregulation of pendrin led to an increase in thiazide-sensitive sodium chloride absorption by the cortical collecting duct, and it caused metabolic acidosis. The function of apical potassium channels was altered in this model, and hyperkalemia was fully corrected by pendrin genetic ablation. Thus, we demonstrate an important contribution of pendrin in renal regulation of sodium chloride, potassium and acid-base homeostasis and in the pathophysiology of PHAII. Furthermore, we identify renal distal bicarbonate secretion as a novel mechanism of renal tubular acidosis.


Subject(s)
Acidosis, Renal Tubular/physiopathology , Kidney Tubules, Collecting/physiopathology , Protein Serine-Threonine Kinases/genetics , Pseudohypoaldosteronism/complications , Sulfate Transporters/metabolism , Acidosis, Renal Tubular/blood , Acidosis, Renal Tubular/etiology , Animals , Disease Models, Animal , Gene Knockout Techniques , Humans , Kidney Tubules, Collecting/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation, Missense , Potassium/blood , Potassium/metabolism , Pseudohypoaldosteronism/genetics , Pseudohypoaldosteronism/physiopathology , Renal Elimination , Sodium Chloride/metabolism , Sodium-Bicarbonate Symporters/metabolism , Sulfate Transporters/genetics , Up-Regulation
8.
J Am Soc Nephrol ; 28(1): 130-139, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27151921

ABSTRACT

We recently described a novel thiazide-sensitive electroneutral NaCl transport mechanism resulting from the parallel operation of the Cl-/HCO3- exchanger pendrin and the Na+-driven Cl-/2HCO3- exchanger (NDCBE) in ß-intercalated cells of the collecting duct. Although a role for pendrin in maintaining Na+ balance, intravascular volume, and BP is well supported, there is no in vivo evidence for the role of NDCBE in maintaining Na+ balance. Here, we show that deletion of NDCBE in mice caused only subtle perturbations of Na+ homeostasis and provide evidence that the Na+/Cl- cotransporter (NCC) compensated for the inactivation of NDCBE. To unmask the role of NDCBE, we generated Ndcbe/Ncc double-knockout (dKO) mice. On a normal salt diet, dKO and single-knockout mice exhibited similar activation of the renin-angiotensin-aldosterone system, whereas only dKO mice displayed a lower blood K+ concentration. Furthermore, dKO mice displayed upregulation of the epithelial sodium channel (ENaC) and the Ca2+-activated K+ channel BKCa. During NaCl depletion, only dKO mice developed marked intravascular volume contraction, despite dramatically increased renin activity. Notably, the increase in aldosterone levels expected on NaCl depletion was attenuated in dKO mice, and single-knockout and dKO mice had similar blood K+ concentrations under this condition. In conclusion, NDCBE is necessary for maintaining sodium balance and intravascular volume during salt depletion or NCC inactivation in mice. Furthermore, NDCBE has an important role in the prevention of hypokalemia. Because NCC and NDCBE are both thiazide targets, the combined inhibition of NCC and the NDCBE/pendrin system may explain thiazide-induced hypokalemia in some patients.


Subject(s)
Blood Volume , Chloride-Bicarbonate Antiporters/physiology , Hypokalemia/etiology , Animals , Mice , Mice, Knockout , Up-Regulation
9.
J Am Soc Nephrol ; 28(1): 209-217, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27335120

ABSTRACT

Chloride transport by the renal tubule is critical for blood pressure (BP), acid-base, and potassium homeostasis. Chloride uptake from the urinary fluid is mediated by various apical transporters, whereas basolateral chloride exit is thought to be mediated by ClC-Ka/K1 and ClC-Kb/K2, two chloride channels from the ClC family, or by KCl cotransporters from the SLC12 gene family. Nevertheless, the localization and role of ClC-K channels is not fully resolved. Because inactivating mutations in ClC-Kb/K2 cause Bartter syndrome, a disease that mimics the effects of the loop diuretic furosemide, ClC-Kb/K2 is assumed to have a critical role in salt handling by the thick ascending limb. To dissect the role of this channel in detail, we generated a mouse model with a targeted disruption of the murine ortholog ClC-K2. Mutant mice developed a Bartter syndrome phenotype, characterized by renal salt loss, marked hypokalemia, and metabolic alkalosis. Patch-clamp analysis of tubules isolated from knockout (KO) mice suggested that ClC-K2 is the main basolateral chloride channel in the thick ascending limb and in the aldosterone-sensitive distal nephron. Accordingly, ClC-K2 KO mice did not exhibit the natriuretic response to furosemide and exhibited a severely blunted response to thiazide. We conclude that ClC-Kb/K2 is critical for salt absorption not only by the thick ascending limb, but also by the distal convoluted tubule.


Subject(s)
Anion Transport Proteins/physiology , Chloride Channels/physiology , Nephrons/metabolism , Sodium Chloride/metabolism , Animals , Diuretics/pharmacology , Furosemide/pharmacology , Mice , Mice, Knockout , Nephrons/drug effects , Sodium Chloride Symporter Inhibitors/pharmacology
10.
J Am Soc Nephrol ; 28(5): 1507-1520, 2017 May.
Article in English | MEDLINE | ID: mdl-27932475

ABSTRACT

Distal nephron acid secretion is mediated by highly specialized type A intercalated cells (A-ICs), which contain vacuolar H+-ATPase (V-type ATPase)-rich vesicles that fuse with the apical plasma membrane on demand. Intracellular bicarbonate generated by luminal H+ secretion is removed by the basolateral anion-exchanger AE1. Chronically reduced renal acid excretion in distal renal tubular acidosis (dRTA) may lead to nephrocalcinosis and renal failure. Studies in MDCK monolayers led to the proposal of a dominant-negative trafficking mechanism to explain AE1-associated dominant dRTA. To test this hypothesis in vivo, we generated an Ae1 R607H knockin mouse, which corresponds to the most common dominant dRTA mutation in human AE1, R589H. Compared with wild-type mice, heterozygous and homozygous R607H knockin mice displayed incomplete dRTA characterized by compensatory upregulation of the Na+/HCO3- cotransporter NBCn1. Red blood cell Ae1-mediated anion-exchange activity and surface polypeptide expression did not change. Mutant mice expressed far less Ae1 in A-ICs, but basolateral targeting of the mutant protein was preserved. Notably, mutant mice also exhibited reduced expression of V-type ATPase and compromised targeting of this proton pump to the plasma membrane upon acid challenge. Accumulation of p62- and ubiquitin-positive material in A-ICs of knockin mice suggested a defect in the degradative pathway, which may explain the observed loss of A-ICs. R607H knockin did not affect type B intercalated cells. We propose that reduced basolateral anion-exchange activity in A-ICs inhibits trafficking and regulation of V-type ATPase, compromising luminal H+ secretion and possibly lysosomal acidification.


Subject(s)
Acidosis, Renal Tubular/enzymology , Anion Exchange Protein 1, Erythrocyte/physiology , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/enzymology , Vacuolar Proton-Translocating ATPases/physiology , Animals , Anion Exchange Protein 1, Erythrocyte/genetics , Male , Mice , Models, Biological
11.
Nephrol Dial Transplant ; 32(7): 1137-1145, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28064162

ABSTRACT

BACKGROUND: Pendrin, the chloride/bicarbonate exchanger of ß-intercalated cells of the renal connecting tubule and the collecting duct, plays a key role in NaCl reabsorption by the distal nephron. Therefore, pendrin may be important for the control of extracellular fluid volume and blood pressure. METHODS: Here, we have used a genetic mouse model in which the expression of pendrin can be switched-on in vivo by the administration of doxycycline. Pendrin can also be rapidly removed when doxycycline administration is discontinued. Therefore, our genetic strategy allows us to test selectively the acute effects of loss of pendrin function. RESULTS: We show that acute loss of pendrin leads to a significant decrease of blood pressure. In addition, acute ablation of pendrin did not alter significantly the acid-base status or blood K + concentration. CONCLUSION: By using a transgenic mouse model, avoiding off-target effects related to pharmacological compounds, this study suggests that pendrin could be a novel target to treat hypertension.


Subject(s)
Anion Transport Proteins/physiology , Blood Pressure/physiology , Hypertension/etiology , Animals , Hypertension/metabolism , Hypertension/pathology , Male , Mice , Mice, Transgenic , Sulfate Transporters
12.
Eur Radiol ; 27(2): 651-659, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27221559

ABSTRACT

OBJECTIVES: Screening of living kidney donors may require scintigraphy to split glomerular filtration rate (GFR). To determine the usefulness of computed tomography (CT) to split GFR, we compared scintigraphy-split GFR to CT-split GFR. We evaluated CT-split GFR as a screening test to detect scintigraphy-split GFR lower than 40 mL/min/1.73 m2/kidney. METHODS: This was a monocentric retrospective study on 346 potential living donors who had GFR measurement, renal scintigraphy, and CT. We predicted GFR for each kidney by splitting GFR using the following formula: Volume-split GFR for a given kidney = measured GFR*[volume of this kidney/(volume of this kidney + volume of the opposite kidney)]. The same formula was used for length-split GFR. We compared length- and volume-split GFR to scintigraphy-split GFR at donation and with a 4-year follow-up. RESULTS: A better correlation was observed between length-split GFR and scintigraphy-split GFR (r = 0.92) than between volume-split GFR and scintigraphy-split GFR (r = 0.89). A length-split GFR threshold of 45 mL/min/1.73 m2/kidney had a sensitivity of 100 % and a specificity of 75 % to detect scintigraphy-split GFR less than 40 mL/min/1.73 m2/kidney. Both techniques with their respective thresholds detected living donors with similar eGFR evolution during follow-up. CONCLUSION: Length-split GFR can be used to detect patients requiring scintigraphy. KEY POINTS: • Excellent correlation between kidney length and scintigraphy predicted GFR • Kidney length screening detects all donors with GFR lower than 40 mL/min/1.73 m 2 • Kidney length screening can replace scintigraphy screening.


Subject(s)
Glomerular Filtration Rate/physiology , Kidney Transplantation , Kidney/diagnostic imaging , Kidney/physiology , Living Donors , Tomography, X-Ray Computed/methods , Female , Follow-Up Studies , Humans , Male , Middle Aged , Radionuclide Imaging , Retrospective Studies , Sensitivity and Specificity
13.
Transpl Int ; 30(8): 799-806, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28152216

ABSTRACT

Two end-stage renal disease (ESRD) risk calculators were recently developed by Grams et al., and Ibrahim et al. to calculate ESRD risk before donation among living kidney donors. However, those calculators have never been studied among potential donors for whom donation was refused due to medical contraindications and compared to a group of donors. We compared 15-year and lifetime ESRD risk of donors and nondonors due to medical cause as estimated by those two calculators. Nondonors due to medical cause (n = 27) had a significantly higher 15-year ESRD risk compared to donors (n = 288) with both calculators (0.25 vs. 0.14, P < 0.001 for that developed by Grams et al. and 2.21 vs. 1.43, P = 0.002 for that developed by Ibrahim et al.). On the contrary, lifetime ESRD risk was not significantly different between the two groups. At both times (15 years and lifetime), we observed a significant overlap of ESRD risk between the two groups. ESRD risk calculators could be complementary to standard screening strategy but cannot be used alone to accept or decline donation.


Subject(s)
Kidney Failure, Chronic/etiology , Kidney Transplantation , Living Donors , Nephrectomy/adverse effects , Adult , Contraindications, Procedure , Donor Selection , Female , Humans , Male , Middle Aged , Risk , Tissue and Organ Harvesting/adverse effects , Tissue and Organ Procurement
14.
J Am Soc Nephrol ; 27(12): 3511-3520, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27468975

ABSTRACT

Metabolic acidosis is associated with increased urinary calcium excretion and related sequelae, including nephrocalcinosis and nephrolithiasis. The increased urinary calcium excretion induced by metabolic acidosis predominantly results from increased mobilization of calcium out of bone and inhibition of calcium transport processes within the renal tubule. The mechanisms whereby acid alters the integrity and stability of bone have been examined extensively in the published literature. Here, after briefly reviewing this literature, we consider the effects of acid on calcium transport in the renal tubule and then discuss why not all gene defects that cause renal tubular acidosis are associated with hypercalciuria and nephrocalcinosis.


Subject(s)
Acidosis/genetics , Acidosis/urine , Calcium/urine , Kidney Tubules , Acid-Base Imbalance/complications , Acid-Base Imbalance/etiology , Acid-Base Imbalance/metabolism , Acidosis/classification , Bone Diseases/etiology , Calcium/metabolism , Humans , Hypercalciuria/etiology , Kidney Tubules/metabolism , Nephrocalcinosis/etiology
15.
J Am Soc Nephrol ; 27(11): 3320-3330, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27044666

ABSTRACT

ATPase H+-transporting lysosomal accessory protein 2 (Atp6ap2), also known as the (pro)renin receptor, is a type 1 transmembrane protein and an accessory subunit of the vacuolar H+-ATPase (V-ATPase) that may also function within the renin-angiotensin system. However, the contribution of Atp6ap2 to renin-angiotensin-dependent functions remains unconfirmed. Using mice with an inducible conditional deletion of Atp6ap2 in mouse renal epithelial cells, we found that decreased V-ATPase expression and activity in the intercalated cells of the collecting duct impaired acid-base regulation by the kidney. In addition, these mice suffered from marked polyuria resistant to desmopressin administration. Immunoblotting revealed downregulation of the medullary Na+-K+-2Cl- cotransporter NKCC2 in these mice compared with wild-type mice, an effect accompanied by a hypotonic medullary interstitium and impaired countercurrent multiplication. This phenotype correlated with strong autophagic defects in epithelial cells of medullary tubules. Notably, cells with high accumulation of the autophagosomal substrate p62 displayed the strongest reduction of NKCC2 expression. Finally, nephron-specific Atp6ap2 depletion did not affect angiotensin II production, angiotensin II-dependent BP regulation, or sodium handling in the kidney. Taken together, our results show that nephron-specific deletion of Atp6ap2 does not affect the renin-angiotensin system but causes a combination of renal concentration defects and distal renal tubular acidosis as a result of impaired V-ATPase activity.


Subject(s)
Kidney/enzymology , Proton-Translocating ATPases/physiology , Receptors, Cell Surface/physiology , Renin-Angiotensin System/physiology , Vacuolar Proton-Translocating ATPases/physiology , Animals , Female , Male , Mice
16.
Am J Physiol Renal Physiol ; 311(5): F901-F906, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27582101

ABSTRACT

The distal nephron is a heterogeneous part of the nephron composed by six different cell types, forming the epithelium of the distal convoluted (DCT), connecting, and collecting duct. To dissect the function of these cells, knockout models specific for their unique cell marker have been created. However, since this part of the nephron develops at the border between the ureteric bud and the metanephric mesenchyme, the specificity of the single cell markers has been recently questioned. Here, by mapping the fate of the aquaporin 2 (AQP2) and Na+-Cl- cotransporter (NCC)-positive cells using transgenic mouse lines expressing the yellow fluorescent protein fluorescent marker, we showed that the origin of the distal nephron is extremely composite. Indeed, AQP2-expressing precursor results give rise not only to the principal cells, but also to some of the A- and B-type intercalated cells and even to cells of the DCT. On the other hand, some principal cells and B-type intercalated cells can develop from NCC-expressing precursors. In conclusion, these results demonstrate that the origin of different cell types in the distal nephron is not as clearly defined as originally thought. Importantly, they highlight the fact that knocking out a gene encoding for a selective functional marker in the adult does not guarantee cell specificity during the overall kidney development. Tools allowing not only cell-specific but also time-controlled recombination will be useful in this sense.


Subject(s)
Kidney Tubules, Collecting/metabolism , Kidney Tubules, Distal/metabolism , Nephrons/metabolism , Animals , Aquaporin 2/metabolism , Mice , Mice, Transgenic , Models, Biological , Sodium Chloride Symporters/metabolism
17.
Proc Natl Acad Sci U S A ; 110(19): 7928-33, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23610411

ABSTRACT

The Na(+) concentration of the intracellular milieu is very low compared with the extracellular medium. Transport of Na(+) along this gradient is used to fuel secondary transport of many solutes, and thus plays a major role for most cell functions including the control of cell volume and resting membrane potential. Because of a continuous leak, Na(+) has to be permanently removed from the intracellular milieu, a process that is thought to be exclusively mediated by the Na(+)/K(+)-ATPase in animal cells. Here, we show that intercalated cells of the mouse kidney are an exception to this general rule. By an approach combining two-photon imaging of isolated renal tubules, physiological studies, and genetically engineered animals, we demonstrate that inhibition of the H(+) vacuolar-type ATPase (V-ATPase) caused drastic cell swelling and depolarization, and also inhibited the NaCl absorption pathway that we recently discovered in intercalated cells. In contrast, pharmacological blockade of the Na(+)/K(+)-ATPase had no effects. Basolateral NaCl exit from ß-intercalated cells was independent of the Na(+)/K(+)-ATPase but critically relied on the presence of the basolateral ion transporter anion exchanger 4. We conclude that not all animal cells critically rely on the sodium pump as the unique bioenergizer, but can be replaced by the H(+) V-ATPase in renal intercalated cells. This concept is likely to apply to other animal cell types characterized by plasma membrane expression of the H(+) V-ATPase.


Subject(s)
Kidney/metabolism , Sodium-Potassium-Exchanging ATPase/physiology , Sodium/metabolism , Absorption , Animals , Cell Membrane/metabolism , Cells, Cultured , Chloride-Bicarbonate Antiporters/genetics , Immunohistochemistry , Ions , Membrane Potentials , Mice , Mice, Knockout , Perfusion , Proton Pumps/physiology , Sodium Chloride/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Vacuolar Proton-Translocating ATPases/metabolism
18.
PLoS Genet ; 9(7): e1003641, 2013.
Article in English | MEDLINE | ID: mdl-23874234

ABSTRACT

Mutations of SLC26A4 are a common cause of human hearing loss associated with enlargement of the vestibular aqueduct. SLC26A4 encodes pendrin, an anion exchanger expressed in a variety of epithelial cells in the cochlea, the vestibular labyrinth and the endolymphatic sac. Slc26a4 (Δ/Δ) mice are devoid of pendrin and develop a severe enlargement of the membranous labyrinth, fail to acquire hearing and balance, and thereby provide a model for the human phenotype. Here, we generated a transgenic mouse line that expresses human SLC26A4 controlled by the promoter of ATP6V1B1. Crossing this transgene into the Slc26a4 (Δ/Δ) line restored protein expression of pendrin in the endolymphatic sac without inducing detectable expression in the cochlea or the vestibular sensory organs. The transgene prevented abnormal enlargement of the membranous labyrinth, restored a normal endocochlear potential, normal pH gradients between endolymph and perilymph in the cochlea, normal otoconia formation in the vestibular labyrinth and normal sensory functions of hearing and balance. Our study demonstrates that restoration of pendrin to the endolymphatic sac is sufficient to restore normal inner ear function. This finding in conjunction with our previous report that pendrin expression is required for embryonic development but not for the maintenance of hearing opens the prospect that a spatially and temporally limited therapy will restore normal hearing in human patients carrying a variety of mutations of SLC26A4.


Subject(s)
Ear, Inner/metabolism , Endolymphatic Sac/metabolism , Hearing Loss/genetics , Membrane Transport Proteins/genetics , Animals , Anion Transport Proteins/metabolism , Ear, Inner/pathology , Endolymph/metabolism , Endolymphatic Sac/pathology , Female , Hearing Loss/pathology , Humans , Mice , Mice, Transgenic , Mutation , Pregnancy , Sulfate Transporters , Vacuolar Proton-Translocating ATPases/genetics , Vestibular Aqueduct/metabolism , Vestibular Aqueduct/physiopathology
19.
Annu Rev Physiol ; 74: 325-49, 2012.
Article in English | MEDLINE | ID: mdl-21888509

ABSTRACT

The distal nephron plays a critical role in the renal control of homeostasis. Until very recently most studies focused on the control of Na(+), K(+), and water balance by principal cells of the collecting duct and the regulation of solute and water by hormones from the renin-angiotensin-aldosterone system and by antidiuretic hormone. However, recent studies have revealed the unexpected importance of renal intercalated cells, a subtype of cells present in the connecting tubule and collecting ducts. Such cells were thought initially to be involved exclusively in acid-base regulation. However, it is clear now that intercalated cells absorb NaCl and K(+) and hence may participate in the regulation of blood pressure and potassium balance. The second paradigm-challenging concept we highlight is the emerging importance of local paracrine factors that play a critical role in the renal control of water and electrolyte balance.


Subject(s)
Electrolytes/metabolism , Kidney Tubules, Distal/metabolism , Adenosine Triphosphate/metabolism , Animals , Biological Transport/physiology , Blood Pressure/physiology , Chlorides/metabolism , Colon/metabolism , Diuretics/pharmacology , H(+)-K(+)-Exchanging ATPase/metabolism , Homeostasis/physiology , Humans , Kallikreins/metabolism , Kallikreins/physiology , Kidney Tubules, Distal/cytology , Potassium/metabolism , Renin-Angiotensin System/physiology , Sodium Channels/physiology , Sodium Chloride/metabolism , Thiazides/pharmacology
20.
Pflugers Arch ; 467(8): 1623-41, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25515081

ABSTRACT

Because majority of biological processes are dependent on pH, maintaining systemic acid-base balance is critical. The kidney contributes to systemic acid-base regulation, by reabsorbing HCO3 (-) (both filtered by glomeruli and generated within a nephron) and acidifying urine. Abnormalities in those processes will eventually lead to a disruption in systemic acid-base balance and provoke metabolic acid-base disorders. Research over the past 30 years advanced our understanding on cellular and molecular mechanisms responsible for those processes. In particular, a variety of transgenic animal models, where target genes are deleted either globally or conditionally, provided significant insights into how specific transporters are contributing to the renal acid-base regulation. Here, we broadly overview the mechanisms of renal ion transport participating to acid-base regulation, with emphasis on data obtained from transgenic mice models.


Subject(s)
Kidney/metabolism , Membrane Transport Proteins/metabolism , Water-Electrolyte Balance , Water-Electrolyte Imbalance/metabolism , Animals , Disease Models, Animal , Genotype , Humans , Kidney/physiopathology , Membrane Transport Proteins/genetics , Mice, Transgenic , Models, Biological , Phenotype , Renal Elimination , Renal Reabsorption , Water-Electrolyte Imbalance/genetics , Water-Electrolyte Imbalance/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL