Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 133(21)2023 11 01.
Article in English | MEDLINE | ID: mdl-37676724

ABSTRACT

Consumption of low dietary potassium, common with ultraprocessed foods, activates the thiazide-sensitive sodium chloride cotransporter (NCC) via the with no (K) lysine kinase/STE20/SPS1-related proline-alanine-rich protein kinase (WNK/SPAK) pathway to induce salt retention and elevate blood pressure (BP). However, it remains unclear how high-potassium "DASH-like" diets (dietary approaches to stop hypertension) inactivate the cotransporter and whether this decreases BP. A transcriptomics screen identified Ppp1Ca, encoding PP1A, as a potassium-upregulated gene, and its negative regulator Ppp1r1a, as a potassium-suppressed gene in the kidney. PP1A directly binds to and dephosphorylates NCC when extracellular potassium is elevated. Using mice genetically engineered to constitutively activate the NCC-regulatory kinase SPAK and thereby eliminate the effects of the WNK/SPAK kinase cascade, we confirmed that PP1A dephosphorylated NCC directly in a potassium-regulated manner. Prior adaptation to a high-potassium diet was required to maximally dephosphorylate NCC and lower BP in constitutively active SPAK mice, and this was associated with potassium-dependent suppression of Ppp1r1a and dephosphorylation of its cognate protein, inhibitory subunit 1 (I1). In conclusion, potassium-dependent activation of PP1A and inhibition of I1 drove NCC dephosphorylation, providing a mechanism to explain how high dietary K+ lowers BP. Shifting signaling of PP1A in favor of activation of WNK/SPAK may provide an improved therapeutic approach for treating salt-sensitive hypertension.


Subject(s)
Hypertension , Protein Serine-Threonine Kinases , Animals , Mice , Blood Pressure/physiology , Solute Carrier Family 12, Member 3/genetics , Solute Carrier Family 12, Member 3/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Sodium Chloride/metabolism , Sodium Chloride/pharmacology , Potassium, Dietary/metabolism , Potassium, Dietary/pharmacology , Kidney/metabolism , Hypertension/genetics , Hypertension/metabolism , Potassium/metabolism , Potassium/pharmacology , Phosphorylation
2.
Am J Physiol Renal Physiol ; 325(3): F377-F393, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37498547

ABSTRACT

The urinary potassium (K+) excretion machinery is upregulated with increasing dietary K+, but the role of accompanying dietary anions remains inadequately characterized. Poorly absorbable anions, including [Formula: see text], are thought to increase K+ secretion through a transepithelial voltage effect. Here, we tested if they also influence the K+ secretion machinery. Wild-type mice, aldosterone synthase (AS) knockout (KO) mice, or pendrin KO mice were randomized to control, high-KCl, or high-KHCO3 diets. The K+ secretory capacity was assessed in balance experiments. Protein abundance, modification, and localization of K+-secretory transporters were evaluated by Western blot analysis and confocal microscopy. Feeding the high-KHCO3 diet increased urinary K+ excretion and the transtubular K+ gradient significantly more than the high-KCl diet, coincident with more pronounced upregulation of epithelial Na+ channels (ENaC) and renal outer medullary K+ (ROMK) channels and apical localization in the distal nephron. Experiments in AS KO mice revealed that the enhanced effects of [Formula: see text] were aldosterone independent. The high-KHCO3 diet also uniquely increased the large-conductance Ca2+-activated K+ (BK) channel ß4-subunit, stabilizing BKα on the apical membrane, the Cl-/[Formula: see text] exchanger, pendrin, and the apical KCl cotransporter (KCC3a), all of which are expressed specifically in pendrin-positive intercalated cells. Experiments in pendrin KO mice revealed that pendrin was required to increase K+ excretion with the high-KHCO3 diet. In summary, [Formula: see text] stimulates K+ excretion beyond a poorly absorbable anion effect, upregulating ENaC and ROMK in principal cells and BK, pendrin, and KCC3a in pendrin-positive intercalated cells. The adaptive mechanism prevents hyperkalemia and alkalosis with the consumption of alkaline ash-rich diets but may drive K+ wasting and hypokalemia in alkalosis.NEW & NOTEWORTHY Dietary anions profoundly impact K+ homeostasis. Here, we found that a K+-rich diet, containing [Formula: see text] as the counteranion, enhances the electrogenic K+ excretory machinery, epithelial Na+ channels, and renal outer medullary K+ channels, much more than a high-KCl diet. It also uniquely induces KCC3a and pendrin, in B-intercalated cells, providing an electroneutral KHCO3 secretion pathway. These findings reveal new K+ balance mechanisms that drive adaption to alkaline and K+-rich foods, which should guide new treatment strategies for K+ disorders.


Subject(s)
Alkalosis , Potassium , Animals , Mice , Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Anions/metabolism , Diet , Mice, Knockout , Potassium/metabolism , Potassium, Dietary/metabolism , Sodium/metabolism , Sulfate Transporters/genetics
3.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 40(4): 415-20, 2015 Apr.
Article in Chinese | MEDLINE | ID: mdl-25931220

ABSTRACT

OBJECTIVE: To explore the impact of pre-pregnancy body mass index on baby's physical growth and nutritional status. METHODS: A total of 491 pairs of mother-infant were divided into 3 groups according to mother's pre-pregnancy body mass index (BMI): a pre-pregnancy low BMI group (BMI<18.5 kg/m², n=93), a pre-pregnancy normal BMI group (18.5 kg/m² ≤ BMI<24.0 kg/m², n=326), and a pre-pregnancy high BMI group (BMI ≥ 24.0 kg/m², n=72). Analysis of variance of repeated measurement data and the median percentage methods were used to compare the physical growth and nutritional status of babies in different groups. RESULTS: Baby's weight in the high BMI group were higher than that in the normal BMI and the low BMI group (F=3.958, P=0.020). The incidence of malnutrition in the low BMI group showed a tendency to decline along with the months (χ²=5.611, P=0.018), the incidence of overweight and obesity in the high and the normal BMI groups displayed a tendency to decline along with the months (χ²=18.773, 53.248, all P<0.001). Baby in the low BMI group had higher incidence of malnutrition while baby in the high BMI group had higher incidence of overweight and obesity. CONCLUSION: Pregnancy BMI was correlated with the growth of baby. Too high or too low prepregnancy BMI exerts harmful effect on baby's weight and nutritional status. Medical workers should strengthen the education on women's pre-pregnancy to remind them keeping BMI at normal level.


Subject(s)
Body Mass Index , Infant Nutritional Physiological Phenomena , Nutritional Status , Birth Weight , Female , Humans , Infant , Obesity , Overweight , Pregnancy , Weight Gain
4.
Physiol Rep ; 1(6): e00160, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24400161

ABSTRACT

A multiplex family was identified with biochemical and clinical features suggestive of Bartter's syndrome (BS). The eldest sibling presented with developmental delay and rickets at 4 years of age with evidence of hypercalciuria and hypokalemia. The second sibling presented at 1 year of age with urinary tract infections, polyuria, and polydipsia. The third child was born after a premature delivery with a history of polyhydramnios and neonatal hypocalcemia. Following corrective treatment she also developed hypercalciuria and a hypokalemic metabolic alkalosis. There was evidence of secondary hyperreninemia and hyperaldosteronism in all three siblings consistent with BS. Known BS genes were screened and functional assays of ROMK (alias KCNJ1, Kir1.1) were carried out in Xenopus oocytes. We detected compound heterozygous missense changes in KCNJ1, encoding the potassium channel ROMK. The S219R/L220F mutation was segregated from father and mother, respectively. In silico modeling of the missense mutations suggested deleterious changes. Studies in Xenopus oocytes revealed that both S219R and L220F had a deleterious effect on ROMK-mediated potassium currents. Coinjection to mimic the compound heterozygosity produced a synergistic decrease in channel function and revealed a loss of PKA-dependent stabilization of PIP2 binding. In conclusion, in a multiplex family with BS, we identified compound heterozygous mutations in KCNJ1. Functional studies of ROMK confirmed the pathogenicity of these mutations and defined the mechanism of channel dysfunction.

5.
Am J Physiol Renal Physiol ; 299(6): F1359-64, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20926634

ABSTRACT

The renal outer medullary K(+) (ROMK) channel plays a critical role in renal sodium handling. Recent genome sequencing efforts in the Framingham Heart Study offspring cohort (Ji W, Foo JN, O'Roak BJ, Zhao H, Larson MG, Simon DB, Newton-Cheh C, State MW, Levy D, and Lifton RP. Nat Genet 40: 592-599, 2008) recently revealed an association between suspected loss-of-function polymorphisms in the ROMK channel and resistance to hypertension, suggesting that ROMK activity may also be a determinant of blood pressure control in the general population. Here we examine whether these sequence variants do, in fact, alter ROMK channel function and explore the mechanisms. As assessed by two-microelectrode voltage clamp in Xenopus oocytes, 3/5 of the variants (R193P, H251Y, and T313FS) displayed an almost complete attenuation of whole cell ROMK channel activity. Surface antibody binding measurements of external epitope-tagged channels and analysis of glycosylation-state maturation revealed that these variants prevent channel expression at the plasmalemma, likely as a consequence of retention in the endoplasmic reticulum. The other variants (P166S, R169H) had no obvious effects on the basal channel activity or surface expression but, instead, conferred a gain in regulated-inhibitory gating. As assessed in giant excised patch-clamp studies, apparent phosphotidylinositol 4,5-bisphosphate (PIP(2)) binding affinity of the variants was reduced, causing channels to be more susceptible to inhibition upon PIP(2) depletion. Unlike the protein product of the major ROMK allele, these two variants are sensitive to the inhibitory affects of a G protein-coupled receptor, which stimulates PIP(2) hydrolysis. In summary, we have found that hypertension resistance sequence variants inhibit ROMK channel function by different mechanisms, providing new insights into the role of the channel in the maintenance of blood pressure.


Subject(s)
Potassium Channels, Inwardly Rectifying/genetics , Animals , Humans , Hypertension/genetics , Ion Channel Gating/genetics , Kidney Medulla , Mice , Phosphatidylinositol 4,5-Diphosphate/metabolism , Polymorphism, Genetic , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/physiology , Proteostasis Deficiencies , Xenopus laevis
6.
J Med Entomol ; 45(1): 68-74, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18283944

ABSTRACT

A cDNA encoding a putative aquaporin was cloned from the ovaries of the American dog tick, Dermacentor variabilis (Say) (Acari: Ixodidae). The encoded protein is most similar to the vertebrate aquaporin 9 protein family. Localization by reverse transcription-polymerase chain reaction (RT-PCR) shows expression in the gut and ovaries of adult females but not in the synganglion, Malpighian tubules, or salivary glands. Quantitative RT-PCR indicates that it is primarily expressed in the ovaries, with approximately 146 times more transcript than in the gut. When expressed in Xenopus oocytes, the aquaporin-like protein localized to the plasma membrane.


Subject(s)
Aquaporins/isolation & purification , Gastrointestinal Tract/chemistry , Insect Proteins/isolation & purification , Ixodidae/chemistry , Ovary/chemistry , Amino Acid Sequence , Animals , Aquaporins/analysis , Aquaporins/classification , Base Sequence , Female , Gene Expression Regulation , Insect Proteins/analysis , Insect Proteins/classification , Ixodidae/metabolism , Molecular Sequence Data , Oocytes , Protein Transport , Xenopus
7.
Proc Natl Acad Sci U S A ; 103(51): 19569-74, 2006 Dec 19.
Article in English | MEDLINE | ID: mdl-17151195

ABSTRACT

The kidney plays a key role in maintaining potassium (K) homeostasis. K excretion is determined by the balance between K secretion and absorption in distal tubule segments such as the connecting tubule and cortical collecting duct. K secretion takes place by K entering principal cells (PC) from blood side through Na+, K+ -ATPase and being secreted into the lumen via both ROMK-like small-conductance K (SK) channels and Ca2+ -activated big-conductance K (BK) channels. K reabsorption occurs by stimulation of apical K/H-ATPase and inhibition of K recycling across the apical membrane in intercalated cells (IC). The role of ROMK channels in K secretion is well documented. However, the importance of BK channels in mediating K secretion is incompletely understood. It has been shown that their activity increases with high tubule flow rate and augmented K intake. However, BK channels have a low open probability and are mainly located in IC, which lack appropriate transporters for effective K secretion. Here we demonstrate that inhibition of ERK and P38 MAPKs stimulates BK channels in both PC and IC in the cortical collecting duct and that changes in K intake modulate their activity. Under control conditions, BK channel activity in PC was low but increased significantly by inhibition of both ERK and P38. Blocking MAPKs also increased channel open probability of BK in IC and thereby it may affect K backflux and net K absorption Thus, modulation of ERK and P38 MAPK activity is involved in controlling net K secretion in the distal nephron.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Kidney Tubules, Collecting/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Potassium/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Benzoquinones/pharmacology , Biophysical Phenomena , Biophysics , Blotting, Western , Female , Flavonoids/pharmacology , Lactams, Macrocyclic/pharmacology , Male , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Rifabutin/analogs & derivatives
8.
J Am Soc Nephrol ; 17(10): 2687-96, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16971657

ABSTRACT

It was demonstrated previously that low dietary potassium (K) intake stimulates Src family protein tyrosine kinase (PTK) expression via a superoxide-dependent signaling. This study explored the role of mitogen-activated protein kinase (MAPK) in mediating the effect of superoxide anions on PTK expression and ROMK (Kir 1.1) channel activity. Western blot analysis demonstrated that low K intake significantly increased the phosphorylation of P38 MAPK (P38) and extracellular signal-regulated kinase (ERK) but had no effect on phosphorylation of c-JUN N-terminus kinase in renal cortex and outer medulla. The stimulatory effect of low K intake on P38 and ERK was abolished by treatment of rats with tempol. The possibility that increases in superoxide and related products that are induced by low K intake were responsible for stimulating phosphorylation of P38 and ERK also was supported by the finding that application of H(2)O(2) increased the phosphorylation of ERK and P38 in the cultured mouse collecting duct cells. Simultaneous blocking of ERK and P38 completely abolished the effect of H(2)O(2) on c-Src expression in mouse collecting duct cells. For determination of the role of P38 and ERK in the regulation of ROMK-like small-conductance K (SK) channels, the patch-clamp technique was used to study the effect of inhibiting P38 and ERK on SK channels in the cortical collecting duct from rats that were on a control K diet (1.1%) and on a K-deficient diet for 1 d. Inhibition of ERK, c-JUN N-terminus kinase, or P38 alone had no effect on SK channels. In contrast, simultaneous inhibition of P38 and ERK significantly increased channel activity. The effect of inhibiting MAPK on SK channels was not affected in the presence of herbimycin A, a PTK inhibitor, and was larger in rats that were on a K-deficient diet than in rats that were on a normal-K diet. However, the stimulatory effect of inhibiting ERK and P38 on SK was absent in the cortical collecting duct that was treated with colchicine. It is concluded that low K intake-induced increases in superoxide levels are responsible for stimulation of P38 and ERK and that MAPK inhibit the SK channels by stimulating PTK expression and via a PTK-independent mechanism.


Subject(s)
Adrenal Cortex/metabolism , Kidney Tubules, Collecting/drug effects , Mitogen-Activated Protein Kinases/pharmacology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium Channels/metabolism , Potassium/metabolism , Animals , Blotting, Western , Female , Immunoprecipitation , Kidney Tubules, Collecting/metabolism , Male , Patch-Clamp Techniques , Phosphorylation , Potassium Channels, Inwardly Rectifying/metabolism , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Superoxides/metabolism
9.
Proc Natl Acad Sci U S A ; 103(22): 8558-63, 2006 May 30.
Article in English | MEDLINE | ID: mdl-16709664

ABSTRACT

Members of the WNK family of serine/threonine kinases have been implicated as important modulators of salt homeostasis, regulating the balance between renal sodium reabsorption and potassium excretion. Gain-of-expression mutations in the WNK1 gene uncouple Na(+) and K(+) balance and cause a familial disorder of diminished renal potassium excretion, excessive sodium retention, and hypertension (pseudohypoaldosteronism type II or Gordon's syndrome). Alternative splicing of the WNK1 gene produces a kidney-specific short form of WNK1 (KS-WNK1) and a more ubiquitous long form (L-WNK1), but it is not clear how either of these isoforms influence renal potassium excretion. Here we demonstrate that KS-WNK1 and L-WNK1 converge in a pathway to regulate the renal outer-medullary K(+) channel, Kir1.1. Reconstitution studies in Xenopus oocytes reveal that L-WNK1 significantly inhibits Kir1.1 by reducing cell surface localization of the channel. A catalytically inactive L-WNK1 mutant has no inhibitory effect on Kir1.1, indicating that channel inhibition depends on kinase activity. KS-WNK1, lacking an intact kinase domain, does not directly alter Kir1.1. Instead, KS-WNK1 negatively regulates L-WNK1 to release Kir1.1 from inhibition. Acute dietary potassium loading increases the relative abundance of KS-WNK1 to L-WNK1 transcript and protein in the kidney, indicating that physiologic up-regulation of Kir1.1 activity involves a WNK1 isoform switch and KS-WNK1-mediated release from L-WNK1 inhibition. Thus, these observations provide evidence for the physiological regulation of Na(+) and K(+) balance by a kinase isoform switch mechanism.


Subject(s)
Kidney/metabolism , Potassium/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Electrophysiology , Gene Expression Regulation , Isoenzymes/genetics , Isoenzymes/metabolism , Kidney/drug effects , Male , Oocytes/metabolism , Patch-Clamp Techniques , Potassium/pharmacology , Potassium Channels, Inwardly Rectifying/metabolism , Protein Serine-Threonine Kinases/genetics , Rats , Rats, Sprague-Dawley , Xenopus laevis
10.
Am J Physiol Renal Physiol ; 290(4): F806-12, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16204406

ABSTRACT

We used Western blotting to examine the expression of phosphatidylinositol 3-kinase (PI3K) in the renal cortex and outer medulla and employed the patch-clamp technique to study the effect of PI3K on the ROMK-like small-conductance K (SK) channels in the cortical collecting duct (CCD). Low K intake increased the expression of the 110-kDa alpha-subunit (p110alpha) of PI3K compared with rats on a normal-K diet. Because low K intake increases superoxide levels (2), the possibility that increases in superoxide anions may be responsible for the effect of low K intake on the expression of PI3K is supported by finding that addition of H(2)O(2) stimulates the expression of p110alpha in M1 cells. Inhibition of PI3K with either wortmannin or LY-294002 significantly increased channel activity in the CCD from rats on a K-deficient (KD) diet or on a normal-K diet. The stimulatory effect of wortmannin on ROMK channel activity cannot be mimicked by inhibition of phospholipase C with U-73122. This suggests that the effect of inhibiting PI3K was not the result of increasing the phosphatidylinositol 4,5-bisphosphate level. Moreover, application of the exogenous phosphatidylinositol 3,4,5-trisphosphate analog had no effect on channel activity in excised patches. Because low K intake has been shown to increase the activity of protein tyrosine kinase (PTK), we explored the role of the interaction between PTK and PI3K in the regulation of the SK channel activity. Inhibition of PTK increased SK channel activity in the CCD from rats on a KD diet. However, addition of wortmannin did not further increase ROMK channel activity. Also, the effect of wortmannin was abolished by treatment of CCD with phalloidin. We conclude that PI3K is involved in mediating the effect of low K intake on ROMK channel activity in the CCD and that the effect of PI3K on SK channels requires the involvement of PTK and the cytoskeleton.


Subject(s)
Kidney Tubules, Collecting/physiology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Small-Conductance Calcium-Activated Potassium Channels/physiology , Animals , Blotting, Western , Diet , Enzyme Inhibitors/pharmacology , Female , Male , Patch-Clamp Techniques , Potassium/physiology , Potassium Channels, Inwardly Rectifying , Rats , Rats, Sprague-Dawley , Superoxides
11.
Am J Physiol Renal Physiol ; 287(5): F954-9, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15265768

ABSTRACT

We used the patch-clamp technique to study the effect of adenosine on the apical 70-pS K channel in the thick ascending limb (TAL) of the rat kidney. Application of 1 microM cyclohexyladenosine (CHA), an adenosine analog, stimulated apical 70-pS K channel activity and increased the product of channel open probability and channel number (NP(o)) from 0.34 to 0.7. Also, addition of CGS-21680, a specific A(2a) adenosine receptor agonist, mimicked the effect of CHA and increased NP(o) from 0.33 to 0.77. The stimulatory effect of CHA and CGS-21680 was completely blocked by H89, an inhibitor of protein kinase A (PKA), or by inhibition of adenylate cyclase with SQ-22536. This suggests that the stimulatory effect of adenosine analogs is mediated by a PKA-dependent pathway. The effect of adenosine analog was almost absent in the TAL from rats on a K-deficient (KD) diet for 7 days. Application of DDMS, an agent that inhibits cytochrome P-450 hydrolase, not only significantly increased the activity of the 70-pS K channel but also restored the stimulatory effect of CHA on the 70-pS K channel in the TAL from rats on a KD diet. Also, the effect of CHA was absent in the presence of 20-HETE. Inhibition of PKA blocked the stimulatory effect of CHA on the apical 70-pS K channel in the presence of DDMS in the TAL from rats on a KD diet. We conclude that stimulation of adenosine receptor increases the apical 70-pS K channel activity via a PKA-dependent pathway and that the effect of adenosine on the apical 70-pS K channel is suppressed by low-K intake. Moreover, the diminished response to adenosine is the result of increase in 20-HETE formation, which inhibits the cAMP-dependent pathway in the TAL from rats on a KD diet.


Subject(s)
Adenosine/analogs & derivatives , Adenosine/pharmacology , Kidney/metabolism , Potassium Channels/drug effects , Potassium, Dietary/pharmacology , Sulfonamides , Adenosine A2 Receptor Agonists , Adenylyl Cyclase Inhibitors , Animals , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Hydroxyeicosatetraenoic Acids/pharmacology , Isoquinolines/pharmacology , Kidney/drug effects , Membrane Potentials/drug effects , Patch-Clamp Techniques , Phenethylamines/pharmacology , Rats , Rats, Sprague-Dawley
12.
Am J Physiol Cell Physiol ; 286(6): C1258-63, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15151916

ABSTRACT

We used the patch-clamp technique to study the effect of insulin-like growth factor I (IGF-I) on the apical 70-pS K channel in the isolated thick ascending limb (TAL) of the rat kidney. The isolated TAL was cut open to gain access to the apical membrane. Addition of 25 nM IGF-I stimulates the apical 70-pS K channel and increases channel activity, defined by the product of channel open probability and channel number, from 0.31 to 1.21. The stimulatory effect of IGF-I is not mediated by nitric oxide- or protein tyrosine phosphatase-dependent mechanisms, because inhibition of nitric oxide synthase or blocking protein tyrosine phosphatase did not abolish the stimulatory effect of IGF-I on the 70-pS K channel. In contrast, inhibition of mitogen-activated protein (MAP) kinase with PD-98059 or U0126 abolished the stimulatory effect of IGF-I. This suggests that MAP kinase is responsible for mediating the effect of IGF-I on the apical K channels. Moreover, the effect of IGF-I on the apical 70-pS K channel is biphasic because high concentrations (>200 nM) inhibit apical 70-pS K channels. Application of 400 nM IGF-I decreased channel activity from 1.45 to 0.2. The inhibitory effect of IGF-I is not blocked by calphostin C (an inhibitor of PKC), but inhibition of protein tyrosine kinase with herbimycin A abolished the IGF-induced inhibition. We conclude that IGF-I has a dual effect on the apical 70-pS K channel in the TAL: low concentrations of IGF-I stimulate, whereas high concentrations inhibit the channel activity. The stimulatory effect of IGF-I is mediated by a MAP kinase-dependent pathway, whereas the inhibitory effect is the result of stimulation of protein tyrosine kinase.


Subject(s)
Cell Membrane/metabolism , Insulin-Like Growth Factor I/pharmacology , Loop of Henle/metabolism , Potassium Channels/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/physiology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Insulin-Like Growth Factor I/metabolism , Loop of Henle/cytology , Loop of Henle/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Patch-Clamp Techniques , Potassium/metabolism , Potassium Channels/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...