1
|
Vera E, Cornejo I, Henao JC, Tribiños F, Burgos J, Sepúlveda FV, Cid LP. Normal vision and development in mice with low functional expression of Kir7.1 in heterozygosis for a blindness-producing mutation inactivating the channel. Am J Physiol Cell Physiol 2024; 326:C1178-C1192. [PMID: 38406825 DOI: 10.1152/ajpcell.00597.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/18/2024] [Accepted: 02/19/2024] [Indexed: 02/27/2024]
Abstract
K+ channel Kir7.1 expressed at the apical membrane of the retinal pigment epithelium (RPE) plays an essential role in retinal function. An isoleucine-to-threonine mutation at position 120 of the protein is responsible for blindness-causing vitreo-retinal dystrophy. We have studied the molecular mechanism of action of Kir7.1-I120T in vitro by heterologous expression and in vivo in CRISPR-generated knockin mice. Full-size Kir7.1-I120T reaches the plasma membrane but lacks any activity. Analysis of Kir7.1 and the I120T mutant in mixed transfection experiments, and that of tandem tetrameric constructs made by combining wild type (WT) and mutant protomers, leads us to conclude that they do not form heterotetramers in vitro. Homozygous I120T/I120T mice show cleft palate and tracheomalacia and do not survive beyond P0, whereas heterozygous WT/I120T develop normally. Membrane conductance of RPE cells isolated from WT/WT and heterozygous WT/I120T mice is dominated by Kir7.1 current. Using Rb+ as a charge carrier, we demonstrate that the Kir7.1 current of WT/I120T RPE cells corresponds to approximately 50% of that in cells from WT/WT animals, in direct proportion to WT gene dosage. This suggests a lack of compensatory effects or interference from the mutated allele product, an interpretation consistent with results obtained using WT/- hemizygous mouse. Electroretinography and behavioral tests also show normal vision in WT/I120T animals. The hypomorphic ion channel phenotype of heterozygous Kir7.1-I120T mutants is therefore compatible with normal development and retinal function. The lack of detrimental effect of this degree of functional deficit might explain the recessive nature of Kir7.1 mutations causing human eye disease.NEW & NOTEWORTHY Human retinal pigment epithelium K+ channel Kir7.1 is affected by generally recessive mutations leading to blindness. We investigate one such mutation, isoleucine-to-threonine at position 120, both in vitro and in vivo in knockin mice. The mutated channel is inactive and in heterozygosis gives a hypomorphic phenotype with normal retinal function. Mutant channels do not interfere with wild-type Kir7.1 channels which are expressed concomitantly without hindrance, providing an explanation for the recessive nature of the disease.
Collapse
Affiliation(s)
- Erwin Vera
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Universidad Austral de Chile, Valdivia, Chile
| | - Isabel Cornejo
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Valdivia, Chile
| | - Juan Carlos Henao
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Universidad Austral de Chile, Valdivia, Chile
| | | | | | - Francisco V Sepúlveda
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia, Chile
| | - L Pablo Cid
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia, Chile
| |
Collapse
|
2
|
Zietara A, Palygin O, Levchenko V, Dissanayake LV, Klemens CA, Geurts A, Denton JS, Staruschenko A. K ir7.1 knockdown and inhibition alter renal electrolyte handling but not the development of hypertension in Dahl salt-sensitive rats. Am J Physiol Renal Physiol 2023; 325:F177-F187. [PMID: 37318990 PMCID: PMC10393338 DOI: 10.1152/ajprenal.00059.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/22/2023] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
High K+ supplementation is correlated with a lower risk of the composite of death, major cardiovascular events, and ameliorated blood pressure, but the exact mechanisms have not been established. Inwardly rectifying K+ (Kir) channels expressed in the basolateral membrane of the distal nephron play an essential role in maintaining electrolyte homeostasis. Mutations in this channel family have been shown to result in strong disturbances in electrolyte homeostasis, among other symptoms. Kir7.1 is a member of the ATP-regulated subfamily of Kir channels. However, its role in renal ion transport and its effect on blood pressure have yet to be established. Our results indicate the localization of Kir7.1 to the basolateral membrane of aldosterone-sensitive distal nephron cells. To examine the physiological implications of Kir7.1, we generated a knockout of Kir7.1 (Kcnj13) in Dahl salt-sensitive (SS) rats and deployed chronic infusion of a specific Kir7.1 inhibitor, ML418, in the wild-type Dahl SS strain. Knockout of Kcnj13 (Kcnj13-/-) resulted in embryonic lethality. Heterozygous Kcnj13+/- rats revealed an increase in K+ excretion on a normal-salt diet but did not exhibit a difference in blood pressure development or plasma electrolytes after 3 wk of a high-salt diet. Wild-type Dahl SS rats exhibited increased renal Kir7.1 expression when dietary K+ was increased. K+ supplementation also demonstrated that Kcnj13+/- rats excreted more K+ on normal salt. The development of hypertension was not different when rats were challenged with high salt for 3 wk, although Kcnj13+/- rats excrete less Na+. Interestingly, chronic infusion of ML418 significantly increased Na+ and Cl- excretion after 14 days of high salt but did not alter salt-induced hypertension development. Here, we found that reduction of Kir7.1 function, either through genetic ablation or pharmacological inhibition, can influence renal electrolyte excretion but not to a sufficient degree to impact the development of SS hypertension.NEW & NOTEWORTHY To investigate the role of the Kir7.1 channel in salt-sensitive hypertension, its function was examined using complementary genetic and pharmacological approaches. The results revealed that although reducing Kir7.1 expression had some impact on maintaining K+ and Na+ balance, it did not lead to a significant change in the development or magnitude of salt-induced hypertension. Hence, it is probable that Kir7.1 works in conjunction with other basolateral K+ channels to fine-tune membrane potential.
Collapse
Affiliation(s)
- Adrian Zietara
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, United States
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Vladislav Levchenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, United States
| | - Lashodya V Dissanayake
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, United States
| | - Christine A Klemens
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, United States
| | - Aron Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Jerod S Denton
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, United States
- Hypertension and Kidney Research Center, University of South Florida, Tampa, Florida, United States
- James A. Haley Veterans Hospital, Tampa, Florida, United States
| |
Collapse
|
3
|
Schroeder M, Peter VG, Gränse L, Andréasson S, Rivolta C, Kjellström U. A novel phenotype associated with the R162W variant in the KCNJ13 gene. Ophthalmic Genet 2022; 43:500-507. [PMID: 35477418 DOI: 10.1080/13816810.2022.2068041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Pathogenic variants in KCNJ13 have been associated with both autosomal dominant Snowflake vitreoretinal degeneration (SVD) and autosomal recessive Leber congenital amaurosis. SVD is characterized by aberrant vitreoretinal interface leading to increased risk of retinal detachment, crystalline retinal snowflake deposits, optic disc abnormalities, early-onset cataract, and cornea guttae. Reduced dark adaptation and reduced scotopic rod b-waves have also been described. We report a novel phenotype associated with the R162W variant in KCNJ13. METHODS Four affected members of a Swedish family were included. Three of them were examined with best corrected visual acuity, Goldmann perimetry, full-field-and multifocal electroretinography, optical coherence tomography, fundus color photographs, fundus autofluorescence images, slit lamp inspection, and genetic testing. The fourth subject only managed genetic testing. RESULTS All subjects carry the pathogenic missense variant; c.484C>T (NM_002242.4), R162W, in KCNJ13. ERG measurements revealed reduced macular-as well as general retinal function. Two of the subjects had a history of retinal detachment and the two younger subjects demonstrated early onset cataract. They all had structural macular changes and slightly gliotic optic discs. CONCLUSION In this family, the R162W variant in KCNJ13, previously described in association with SVD, causes a somewhat novel phenotype including macular dystrophy and moderate reduction of general retinal function as the main features combined with disc abnormalities, retinal detachment, and presenile cataract that has been described before. In times of up-coming gene-based therapies, it is important to report new genotype-phenotype associations to improve the possibilities to identify future treatment candidates.
Collapse
Affiliation(s)
- Marion Schroeder
- Department of Ophthalmology and Clinical Sciences Lund, Lund University, Skane University Hospital, Lund, Sweden
| | - Virginie G Peter
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland.,Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Lotta Gränse
- Department of Ophthalmology and Clinical Sciences Lund, Lund University, Skane University Hospital, Lund, Sweden
| | - Sten Andréasson
- Department of Ophthalmology and Clinical Sciences Lund, Lund University, Skane University Hospital, Lund, Sweden
| | - Carlo Rivolta
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland.,Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Ulrika Kjellström
- Department of Ophthalmology and Clinical Sciences Lund, Lund University, Skane University Hospital, Lund, Sweden
| |
Collapse
|
4
|
Jiao X, Ma Z, Lei J, Liu P, Cai X, Shahi PK, Chan CC, Fariss R, Pattnaik BR, Dong L, Hejtmancik JF. Retinal Development and Pathophysiology in Kcnj13 Knockout Mice. Front Cell Dev Biol 2022; 9:810020. [PMID: 35096838 PMCID: PMC8790323 DOI: 10.3389/fcell.2021.810020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/10/2021] [Indexed: 11/22/2022] Open
Abstract
Purpose: We constructed and characterized knockout and conditional knockout mice for KCNJ13, encoding the inwardly rectifying K+ channel of the Kir superfamily Kir7.1, mutations in which cause both Snowflake Vitreoretinal Degeneration (SVD) and Retinitis pigmentosa (RP) to further elucidate the pathology of this disease and to develop a potential model system for gene therapy trials. Methods: A Kcnj13 knockout mouse line was constructed by inserting a gene trap cassette expressing beta-galactosidase flanked by FRT sites in intron 1 with LoxP sites flanking exon two and converted to a conditional knockout by FLP recombination followed by crossing with C57BL/6J mice having Cre driven by the VMD2 promoter. Lentiviral replacement of Kcnj13 was driven by the EF1a or VMD2 promoters. Results: Blue-Gal expression is evident in E12.5 brain ventricular choroid plexus, lens, neural retina layer, and anterior RPE. In the adult eye expression is seen in the ciliary body, RPE and choroid. Adult conditional Kcnj13 ko mice show loss of photoreceptors in the outer nuclear layer, inner nuclear layer thinning with loss of bipolar cells, and thinning and disruption of the outer plexiform layer, correlating with Cre expression in the overlying RPE which, although preserved, shows morphological disruption. Fundoscopy and OCT show signs of retinal degeneration consistent with the histology, and photopic and scotopic ERGs are decreased in amplitude or extinguished. Lentiviral based replacement of Kcnj13 resulted in increased ERG c- but not a- or b- wave amplitudes. Conclusion: Ocular KCNJ13 expression starts in the choroid, lens, ciliary body, and anterior retina, while later expression centers on the RPE with no/lower expression in the neuroretina. Although KCNJ13 expression is not required for survival of the RPE, it is necessary for RPE maintenance of the photoreceptors, and loss of the photoreceptor, outer plexiform, and outer nuclear layers occur in adult KCNJ13 cKO mice, concomitant with decreased amplitude and eventual extinguishing of the ERG and signs of retinitis pigmentosa on fundoscopy and OCT. Kcnj13 replacement resulting in recovery of the ERG c- but not a- and b-waves is consistent with the degree of photoreceptor degeneration seen on histology.
Collapse
Affiliation(s)
- Xiaodong Jiao
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Zhiwei Ma
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jingqi Lei
- Genetic Engineering Core, National Eye Institute, National Institute of Health, Bethesda, MD, United States
| | - Pinghu Liu
- Genetic Engineering Core, National Eye Institute, National Institute of Health, Bethesda, MD, United States
| | - Xiaoyu Cai
- Genetic Engineering Core, National Eye Institute, National Institute of Health, Bethesda, MD, United States
| | - Pawan K Shahi
- Departments of Pediatrics and Ophthalmology and Visual Sciences and the McPherson Eye Research Institute, University of Wisconsin, Madison, AL, United States
| | - Chi-Chao Chan
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Robert Fariss
- Imaging Core, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Bikash R Pattnaik
- Departments of Pediatrics and Ophthalmology and Visual Sciences and the McPherson Eye Research Institute, University of Wisconsin, Madison, AL, United States
| | - Lijin Dong
- Genetic Engineering Core, National Eye Institute, National Institute of Health, Bethesda, MD, United States
| | - J Fielding Hejtmancik
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
5
|
Kir Channel Molecular Physiology, Pharmacology, and Therapeutic Implications. Handb Exp Pharmacol 2021; 267:277-356. [PMID: 34345939 DOI: 10.1007/164_2021_501] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
For the past two decades several scholarly reviews have appeared on the inwardly rectifying potassium (Kir) channels. We would like to highlight two efforts in particular, which have provided comprehensive reviews of the literature up to 2010 (Hibino et al., Physiol Rev 90(1):291-366, 2010; Stanfield et al., Rev Physiol Biochem Pharmacol 145:47-179, 2002). In the past decade, great insights into the 3-D atomic resolution structures of Kir channels have begun to provide the molecular basis for their functional properties. More recently, computational studies are beginning to close the time domain gap between in silico dynamic and patch-clamp functional studies. The pharmacology of these channels has also been expanding and the dynamic structural studies provide hope that we are heading toward successful structure-based drug design for this family of K+ channels. In the present review we focus on placing the physiology and pharmacology of this K+ channel family in the context of atomic resolution structures and in providing a glimpse of the promising future of therapeutic opportunities.
Collapse
|
6
|
Weaver CD, Denton JS. Next-generation inward rectifier potassium channel modulators: discovery and molecular pharmacology. Am J Physiol Cell Physiol 2021; 320:C1125-C1140. [PMID: 33826405 DOI: 10.1152/ajpcell.00548.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Inward rectifying potassium (Kir) channels play important roles in both excitable and nonexcitable cells of various organ systems and could represent valuable new drug targets for cardiovascular, metabolic, immune, and neurological diseases. In nonexcitable epithelial cells of the kidney tubule, for example, Kir1.1 (KCNJ1) and Kir4.1 (KCNJ10) are linked to sodium reabsorption in the thick ascending limb of Henle's loop and distal convoluted tubule, respectively, and have been explored as novel-mechanism diuretic targets for managing hypertension and edema. G protein-coupled Kir channels (Kir3) channels expressed in the central nervous system are critical effectors of numerous signal transduction pathways underlying analgesia, addiction, and respiratory-depressive effects of opioids. The historical dearth of pharmacological tool compounds for exploring the therapeutic potential of Kir channels has led to a molecular target-based approach using high-throughput screen (HTS) of small-molecule libraries and medicinal chemistry to develop "next-generation" Kir channel modulators that are both potent and specific for their targets. In this article, we review recent efforts focused specifically on discovery and improvement of target-selective molecular probes. The reader is introduced to fluorescence-based thallium flux assays that have enabled much of this work and then provided with an overview of progress made toward developing modulators of Kir1.1 (VU590, VU591), Kir2.x (ML133), Kir3.X (ML297, GAT1508, GiGA1, VU059331), Kir4.1 (VU0134992), and Kir7.1 (ML418). We discuss what is known about the small molecules' molecular mechanisms of action, in vitro and in vivo pharmacology, and then close with our view of what critical work remains to be done.
Collapse
Affiliation(s)
- C David Weaver
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee.,Department of Chemistry, Vanderbilt University, Nashville, Tennessee.,Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee
| | - Jerod S Denton
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee.,Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee.,Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
7
|
Vera E, Cornejo I, Niemeyer MI, Sepúlveda FV, Cid LP. Altered phosphatidylinositol regulation of mutant inwardly rectifying K + Kir7.1 channels associated with inherited retinal degeneration disease. J Physiol 2020; 599:593-608. [PMID: 33219695 DOI: 10.1113/jp280681] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/12/2020] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Kir7.1 K+ channel expressed in retinal pigment epithelium is mutated in inherited retinal degeneration diseases. We study Kir7.1 in heterologous expression to test the hypothesis that pathological R162 mutation to neutral amino acids results in loss of a crucial site that binds PI(4,5)P2 . Although R162W mutation inactivates Kir7.1, changes to smaller volume (e.g. Gln) amino acids are tolerated or even enhance function (Ala or Cys). Chemical modification of Kir7.1-R162C confirms that large residues of the size of Trp are incompatible with normal channel function even if positively charged. In addition to R162, K164 (and possibly K159) forms a binding site for the phosphoinositide and is essential for channel activity. R162 substitution with a large, neutral side chain like Trp exerts a dominant negative effect on Kir7.1 activity such that less than one fifth of the full activity is expected in a cell expressing the same amount of mutant and wild-type channels. ABSTRACT Mutations in the Kir7.1 K+ channel, highly expressed in retinal pigment epithelium, have been linked to inherited retinal degeneration diseases. Examples are mutations changing Arg 162 to Trp in snowflake vitreoretinal degeneration (SVD) and Gln in retinitis pigmentosa. R162 is believed to be part of a site that binds PI(4,5)P2 and stabilises the open state. We have tested the hypothesis that R162 mutation to neutral amino acids will result in the loss of this crucial interaction to the detriment of channel function. Our findings indicate that although R612W mutation inactivates Kir7.1, changes to smaller volume (e.g. Gln) amino acids are tolerated or even enhance function (Ala or Cys). Cys chemical modification of Kir7.1-R162C confirms that large residues of the size of Trp are incompatible with normal channel function even if positively charged. Experiments titrating the levels of plasma membrane PI(4,5)P2 with voltage-dependent phosphatase DrVSP reveal that, in addition to R162, K164 (and possibly K159) forms a binding site for the phosphoinositide and ensures channel activity. Finally, the use of a concatemeric approach shows that substitution of R162 with a large, neutral side chain mimicking a Trp residue exerts a dominant negative effect on Kir7.1 activity such that less than one fifth of the full activity is expected in heterozygous cells carrying the SVD mutation. Our results suggest that if mutations in the human KCNJ13 gene resulting in the neutralisation of R162 and Kir7.1 malfunction led to retinal degeneration diseases, their severity might depend on the nature of the side chain of the replacing amino acid.
Collapse
Affiliation(s)
- Erwin Vera
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | | | | | | | - L Pablo Cid
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| |
Collapse
|
8
|
Sensing through Non-Sensing Ocular Ion Channels. Int J Mol Sci 2020; 21:ijms21186925. [PMID: 32967234 PMCID: PMC7554890 DOI: 10.3390/ijms21186925] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/12/2022] Open
Abstract
Ion channels are membrane-spanning integral proteins expressed in multiple organs, including the eye. In the eye, ion channels are involved in various physiological processes, like signal transmission and visual processing. A wide range of mutations have been reported in the corresponding genes and their interacting subunit coding genes, which contribute significantly to an array of blindness, termed ocular channelopathies. These mutations result in either a loss- or gain-of channel functions affecting the structure, assembly, trafficking, and localization of channel proteins. A dominant-negative effect is caused in a few channels formed by the assembly of several subunits that exist as homo- or heteromeric proteins. Here, we review the role of different mutations in switching a “sensing” ion channel to “non-sensing,” leading to ocular channelopathies like Leber’s congenital amaurosis 16 (LCA16), cone dystrophy, congenital stationary night blindness (CSNB), achromatopsia, bestrophinopathies, retinitis pigmentosa, etc. We also discuss the various in vitro and in vivo disease models available to investigate the impact of mutations on channel properties, to dissect the disease mechanism, and understand the pathophysiology. Innovating the potential pharmacological and therapeutic approaches and their efficient delivery to the eye for reversing a “non-sensing” channel to “sensing” would be life-changing.
Collapse
|
9
|
Papanikolaou M, Butt AM, Lewis A. A critical role for the inward rectifying potassium channel Kir7.1 in oligodendrocytes of the mouse optic nerve. Brain Struct Funct 2020; 225:925-934. [PMID: 32086565 PMCID: PMC7166203 DOI: 10.1007/s00429-020-02043-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 02/06/2020] [Indexed: 01/27/2023]
Abstract
Inward rectifying potassium channels (Kir) are a large family of ion channels that play key roles in ion homeostasis in oligodendrocytes, the myelinating cells of the central nervous system (CNS). Prominent expression of Kir4.1 has been indicated in oligodendrocytes, but the extent of expression of other Kir subtypes is unclear. Here, we used qRT-PCR to determine expression of Kir channel transcripts in the mouse optic nerve, a white matter tract comprising myelinated axons and the glia that support them. A novel finding was the high relative expression of Kir7.1, comparable to that of Kir4.1, the main glial Kir channel. Significantly, Kir7.1 immunofluorescence labelling in optic nerve sections and in isolated cells was localised to oligodendrocyte somata. Kir7.1 are known as a K+ transporting channels and, using patch clamp electrophysiology and the Kir7.1 blocker VU590, we demonstrated Kir7.1 channels carry a significant proportion of the whole cell potassium conductance in oligodendrocytes isolated from mouse optic nerves. Notably, oligodendrocytes are highly susceptible to ischemia/hypoxia and this is due at least in part to disruption of ion homeostasis. A key finding of this study is that blockade of Kir7.1 with VU590 compromised oligodendrocyte cell integrity and compounds oligodendroglial loss in ischemia/hypoxia in the oxygen-glucose deprivation (OGD) model in isolated intact optic nerves. These data reveal Kir7.1 channels are molecularly and functionally expressed in oligodendrocytes and play an important role in determining oligodendrocyte survival and myelin integrity.
Collapse
Affiliation(s)
- Maria Papanikolaou
- Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Science, University of Portsmouth, Portsmouth, PO1 2DT, UK
| | - Arthur M Butt
- Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Science, University of Portsmouth, Portsmouth, PO1 2DT, UK.
| | - Anthony Lewis
- Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Science, University of Portsmouth, Portsmouth, PO1 2DT, UK
| |
Collapse
|
10
|
Manis AD, Hodges MR, Staruschenko A, Palygin O. Expression, localization, and functional properties of inwardly rectifying K + channels in the kidney. Am J Physiol Renal Physiol 2020; 318:F332-F337. [PMID: 31841387 PMCID: PMC7052651 DOI: 10.1152/ajprenal.00523.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/10/2019] [Accepted: 12/10/2019] [Indexed: 12/13/2022] Open
Abstract
Inwardly rectifying K+ (Kir) channels are expressed in multiple organs and cell types and play critical roles in cellular function. Most notably, Kir channels are major determinants of the resting membrane potential and K+ homeostasis. The renal outer medullary K+ channel (Kir1.1) was the first renal Kir channel identified and cloned in the kidney over two decades ago. Since then, several additional members, including classical and ATP-regulated Kir family classes, have been identified to be expressed in the kidney and to contribute to renal ion transport. Although the ATP-regulated Kir channel class remains the most well known due to severe pathological phenotypes associated with their mutations, progress is being made in defining the properties, localization, and physiological functions of other renal Kir channels, including those localized to the basolateral epithelium. This review is primarily focused on the current knowledge of the expression and localization of renal Kir channels but will also briefly describe their proposed functions in the kidney.
Collapse
Affiliation(s)
- Anna D Manis
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Matthew R Hodges
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Alexander Staruschenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin
| | - Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
11
|
Burgos J, Villanueva S, Ojeda M, Cornejo I, Cid LP, Sepúlveda FV. Kir7.1 inwardly rectifying K + channel is expressed in ciliary body non pigment epithelial cells and might contribute to intraocular pressure regulation. Exp Eye Res 2019; 186:107723. [PMID: 31319081 DOI: 10.1016/j.exer.2019.107723] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/26/2019] [Accepted: 07/10/2019] [Indexed: 02/08/2023]
Abstract
Inwardly rectifying K+ channel Kir7.1 is expressed in epithelia where it shares membrane localisation with the Na+/K+-pump. The ciliary body epithelium (CBE) of the eye is a determinant of intraocular pressure (IOP) through NaCl-driven fluid secretion of aqueous humour. In the present study we explored the presence Kir7.1 in this epithelium in the mouse and its possible functional role in the generation of IOP. Use heterozygous animals for total Kir7.1 knockout expressing β-galactosidase under the control of Kir7.1 promoter, identified the expression of Kir7.1 in non-pigmented epithelial cells of CBE. Using conditional, floxed knockout Kir7.1 mice as negative controls, we found Kir7.1 at the basolateral membrane of the same CBE cell layer. This was confirmed using a knockin mouse expressing the Kir7.1 protein tagged with a haemagglutinin epitope. Measurements using the conditional knockout mouse show only a minor effect of Kir7.1 inactivation on steady-state IOP. Transient increases in IOP in response to general anaesthetics, or to water injection, are absent or markedly curtailed in Kir7.1-deficient mice. These results suggest a role for Kir7.1 in IOP regulation through a possible modulation of aqueous humour production by the CBE non-pigmented epithelial cells. The location of Kir7.1 in the CBE, together with the effect of its removal on dynamic changes in IOP, point to a possible role of the channel as a leak pathway preventing cellular overload of K+ during the secretion process. Kir7.1 could be used as a potential therapeutic target in pathological conditions leading to elevated intraocular pressure.
Collapse
Affiliation(s)
- Johanna Burgos
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia, Chile
| | - Sandra Villanueva
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | - Margarita Ojeda
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia, Chile
| | - Isabel Cornejo
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia, Chile
| | - L Pablo Cid
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia, Chile
| | | |
Collapse
|
12
|
Papanikolaou M, Lewis A, Butt AM. Glial and neuronal expression of the Inward Rectifying Potassium Channel Kir7.1 in the adult mouse brain. J Anat 2019; 235:984-996. [PMID: 31309576 PMCID: PMC6794205 DOI: 10.1111/joa.13048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2019] [Indexed: 01/01/2023] Open
Abstract
Inward Rectifying Potassium channels (Kir) are a large family of ion channels that play key roles in ion homeostasis and neuronal excitability. The most recently described Kir subtype is Kir7.1, which is known as a K+ transporting subtype. Earlier studies localised Kir7.1 to subpopulations of neurones in the brain. However, the pattern of Kir7.1 expression across the brain has not previously been examined. Here, we have determined neuronal and glial expression of Kir7.1 in the adult mouse brain, using immunohistochemistry and transgenic mouse lines expressing reporters specific for astrocytes [glial fibrillary acidic protein‐enhanced green fluorescent protein (GFAP‐EGFP], myelinating oligodendrocytes (PLP‐DsRed), oligodendrocyte progenitor cells (OPC, Pdgfra‐creERT2/Rosa26‐YFP double‐transgenic mice) and all oligodendrocyte lineage cells (SOX10‐EGFP). The results demonstrate significant neuronal Kir7.1 immunostaining in the cortex, hippocampus, cerebellum and pons, as well as the striatum and hypothalamus. In addition, astrocytes are shown to be immunopositive for Kir7.1 throughout grey and white matter, with dense immunostaining on cell somata, primary processes and perivascular end‐feet. Immunostaining for Kir7.1 was observed in oligodendrocytes, myelin and OPCs throughout the brain, although immunostaining was heterogeneous. Neuronal and glial expression of Kir7.1 is confirmed using neurone‐glial cortical cultures and optic nerve glial cultures. Notably, Kir7.1 have been shown to regulate the excitability of thalamic neurones and our results indicate this may be a widespread function of Kir7.1 in neurones throughout the brain. Moreover, based on the function of Kir7.1 in multiple transporting epithelia, Kir7.1 are likely to play an equivalent role in the primary glial function of K+ homeostasis. Our results indicate Kir7.1 are far more pervasive in the brain than previously recognised and have potential importance in regulating neuronal and glial function.
Collapse
Affiliation(s)
- Maria Papanikolaou
- Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Science, University of Portsmouth, Portsmouth, UK
| | - Anthony Lewis
- Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Science, University of Portsmouth, Portsmouth, UK
| | - Arthur M Butt
- Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Science, University of Portsmouth, Portsmouth, UK
| |
Collapse
|
13
|
Vera E, Cornejo I, Burgos J, Niemeyer MI, Sepúlveda FV, Cid LP. A novel Kir7.1 splice variant expressed in various mouse tissues shares organisational and functional properties with human Leber amaurosis-causing mutations of this K + channel. Biochem Biophys Res Commun 2019; 514:574-579. [PMID: 31056263 DOI: 10.1016/j.bbrc.2019.04.169] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 04/24/2019] [Indexed: 01/27/2023]
Abstract
Kir7.1 is an inwardly rectifying K+ channel present in epithelia where it shares membrane localization with the Na+/K+-pump. In the present communication we report the presence of a novel splice variant of Kir7.1 in mouse tissues including kidney, lung, choroid plexus and retinal pigment epithelium (RPE). The variant named mKir7.1-SV2 lacks most of the C-terminus domain but is predicted to have the two transmembrane domains and permeation pathway unaffected. Similarly truncated predicted proteins, Kir7.1-R166X and Kir7.1-Q219X, would arise from mutations associated with Leber Congenital Amaurosis, a rare recessive hereditary retinal disease that results in vision loss at early age. We found that mKir7.1-SV2 and the pathological variants do not produce any channel activity when expressed alone in HEK-293 cells due to their scarce presence in the plasma membrane. Simultaneous expression with the full length Kir7.1 however leads to a reduction in activity of the wild-type channel that might be due to partial proteasome degradation of WT-mutant channel heteromers.
Collapse
Affiliation(s)
- Erwin Vera
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia, Chile
| | - Isabel Cornejo
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia, Chile
| | - Johanna Burgos
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia, Chile
| | | | | | - L Pablo Cid
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia, Chile.
| |
Collapse
|
14
|
Cornejo I, Villanueva S, Burgos J, López-Cayuqueo KI, Chambrey R, Julio-Kalajzić F, Buelvas N, Niemeyer MI, Figueiras-Fierro D, Brown PD, Sepúlveda FV, Cid LP. Tissue Distribution of Kir7.1 Inwardly Rectifying K + Channel Probed in a Knock-in Mouse Expressing a Haemagglutinin-Tagged Protein. Front Physiol 2018; 9:428. [PMID: 29740340 PMCID: PMC5925607 DOI: 10.3389/fphys.2018.00428] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 04/05/2018] [Indexed: 11/13/2022] Open
Abstract
Kir7.1 encoded by the Kcnj13 gene in the mouse is an inwardly rectifying K+ channel present in epithelia where it shares membrane localization with the Na+/K+-pump. Further investigations of the localisation and function of Kir7.1 would benefit from the availability of a knockout mouse, but perinatal mortality attributed to cleft palate in the neonate has thwarted this research. To facilitate localisation studies we now use CRISPR/Cas9 technology to generate a knock-in mouse, the Kir7.1-HA that expresses the channel tagged with a haemagglutinin (HA) epitope. The availability of antibodies for the HA epitope allows for application of western blot and immunolocalisation methods using widely available anti-HA antibodies with WT tissues providing unambiguous negative control. We demonstrate that Kir7.1-HA cloned from the choroid plexus of the knock-in mouse has the electrophysiological properties of the native channel, including characteristically large Rb+ currents. These large Kir7.1-mediated currents are accompanied by abundant apical membrane Kir7.1-HA immunoreactivity. WT-controlled western blots demonstrate the presence of Kir7.1-HA in the eye and the choroid plexus, trachea and lung, and intestinal epithelium but exclusively in the ileum. In the kidney, and at variance with previous reports in the rat and guinea-pig, Kir7.1-HA is expressed in the inner medulla but not in the cortex or outer medulla. In isolated tubules immunoreactivity was associated with inner medulla collecting ducts but not thin limbs of the loop of Henle. Kir7.1-HA shows basolateral expression in the respiratory tract epithelium from trachea to bronchioli. The channel also appears basolateral in the epithelium of the nasal cavity and nasopharynx in newborn animals. We show that HA-tagged Kir7.1 channel introduced in the mouse by a knock-in procedure has functional properties similar to the native protein and the animal thus generated has clear advantages in localisation studies. It might therefore become a useful tool to unravel Kir7.1 function in the different organs where it is expressed.
Collapse
Affiliation(s)
| | - Sandra Villanueva
- Centro de Estudios Científicos, Valdivia, Chile.,Universidad Austral de Chile, Valdivia, Chile
| | - Johanna Burgos
- Centro de Estudios Científicos, Valdivia, Chile.,Universidad Austral de Chile, Valdivia, Chile
| | - Karen I López-Cayuqueo
- Centro de Estudios Científicos, Valdivia, Chile.,Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche S970, PARCC, Hôpital Européen Georges Pompidou, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Régine Chambrey
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche S970, PARCC, Hôpital Européen Georges Pompidou, Assistance Publique Hôpitaux de Paris, Paris, France
| | | | | | | | | | - Peter D Brown
- School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | | | - L P Cid
- Centro de Estudios Científicos, Valdivia, Chile
| |
Collapse
|
15
|
Palygin O, Pochynyuk O, Staruschenko A. Role and mechanisms of regulation of the basolateral K ir 4.1/K ir 5.1K + channels in the distal tubules. Acta Physiol (Oxf) 2017; 219:260-273. [PMID: 27129733 PMCID: PMC5086442 DOI: 10.1111/apha.12703] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 01/28/2016] [Accepted: 04/28/2016] [Indexed: 12/11/2022]
Abstract
Epithelial K+ channels are essential for maintaining electrolyte and fluid homeostasis in the kidney. It is recognized that basolateral inward-rectifying K+ (Kir ) channels play an important role in the control of resting membrane potential and transepithelial voltage, thereby modulating water and electrolyte transport in the distal part of nephron and collecting duct. Monomeric Kir 4.1 (encoded by Kcnj10 gene) and heteromeric Kir 4.1/Kir 5.1 (Kir 4.1 together with Kir 5.1 (Kcnj16)) channels are abundantly expressed at the basolateral membranes of the distal convoluted tubule and the cortical collecting duct cells. Loss-of-function mutations in KCNJ10 cause EAST/SeSAME tubulopathy in humans associated with salt wasting, hypomagnesaemia, metabolic alkalosis and hypokalaemia. In contrast, mice lacking Kir 5.1 have severe renal phenotype that, apart from hypokalaemia, is the opposite of the phenotype seen in EAST/SeSAME syndrome. Experimental advances using genetic animal models provided critical insights into the physiological role of these channels in electrolyte homeostasis and the control of kidney function. Here, we discuss current knowledge about K+ channels at the basolateral membrane of the distal tubules with specific focus on the homomeric Kir 4.1 and heteromeric Kir 4.1/Kir 5.1 channels. Recently identified molecular mechanisms regulating expression and activity of these channels, such as cell acidification, dopamine, insulin and insulin-like growth factor-1, Src family protein tyrosine kinases, as well as the role of these channels in NCC-mediated transport in the distal convoluted tubules, are also described.
Collapse
Affiliation(s)
- Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | | |
Collapse
|
16
|
Swale DR, Kurata H, Kharade SV, Sheehan J, Raphemot R, Voigtritter KR, Figueroa EE, Meiler J, Blobaum AL, Lindsley CW, Hopkins CR, Denton JS. ML418: The First Selective, Sub-Micromolar Pore Blocker of Kir7.1 Potassium Channels. ACS Chem Neurosci 2016; 7:1013-23. [PMID: 27184474 DOI: 10.1021/acschemneuro.6b00111] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The inward rectifier potassium (Kir) channel Kir7.1 (KCNJ13) has recently emerged as a key regulator of melanocortin signaling in the brain, electrolyte homeostasis in the eye, and uterine muscle contractility during pregnancy. The pharmacological tools available for exploring the physiology and therapeutic potential of Kir7.1 have been limited to relatively weak and nonselective small-molecule inhibitors. Here, we report the discovery in a fluorescence-based high-throughput screen of a novel Kir7.1 channel inhibitor, VU714. Site-directed mutagenesis of pore-lining amino acid residues identified glutamate 149 and alanine 150 as essential determinants of VU714 activity. Lead optimization with medicinal chemistry generated ML418, which exhibits sub-micromolar activity (IC50 = 310 nM) and superior selectivity over other Kir channels (at least 17-fold selective over Kir1.1, Kir2.1, Kir2.2, Kir2.3, Kir3.1/3.2, and Kir4.1) except for Kir6.2/SUR1 (equally potent). Evaluation in the EuroFins Lead Profiling panel of 64 GPCRs, ion-channels, and transporters for off-target activity of ML418 revealed a relatively clean ancillary pharmacology. While ML418 exhibited low CLHEP in human microsomes which could be modulated with lipophilicity adjustments, it showed high CLHEP in rat microsomes regardless of lipophilicity. A subsequent in vivo PK study of ML418 by intraperitoneal (IP) administration (30 mg/kg dosage) revealed a suitable PK profile (Cmax = 0.20 μM and Tmax = 3 h) and favorable CNS distribution (mouse brain/plasma Kp of 10.9 to support in vivo studies. ML418, which represents the current state-of-the-art in Kir7.1 inhibitors, should be useful for exploring the physiology of Kir7.1 in vitro and in vivo.
Collapse
Affiliation(s)
| | | | | | - Jonathan Sheehan
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | | | | | | | - Jens Meiler
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | | | | | | | | |
Collapse
|
17
|
Su XT, Zhang C, Wang L, Gu R, Lin DH, Wang WH. Disruption of KCNJ10 (Kir4.1) stimulates the expression of ENaC in the collecting duct. Am J Physiol Renal Physiol 2016; 310:F985-93. [PMID: 26887833 PMCID: PMC5002054 DOI: 10.1152/ajprenal.00584.2015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 02/12/2016] [Indexed: 11/22/2022] Open
Abstract
Kcnj10 encodes the inwardly rectifying K(+) channel 4.1 (Kir4.1) and is expressed in the basolateral membrane of late thick ascending limb, distal convoluted tubule (DCT), connecting tubule (CNT), and cortical collecting duct (CCD). In the present study, we perform experiments in postneonatal day 9 Kcnj10(-/-) or wild-type mice to examine the role of Kir.4.1 in contributing to the basolateral K(+) conductance in the CNT and CCD, and to investigate whether the disruption of Kir4.1 upregulates the expression of the epithelial Na(+) channel (ENaC). Immunostaining shows that Kir4.1 is expressed in the basolateral membrane of CNT and CCD. Patch-clamp studies detect three types of K(+) channels (23, 40, and 60 pS) in the basolateral membrane of late CNT and initial CCD in wild-type (WT) mice. However, only 23- and 60-pS K(+) channels but not the 40-pS K(+) channel were detected in Kcnj10(-/-) mice, suggesting that Kir.4.1 is a key component of the 40-pS K(+) channel in the CNT/CCD. Moreover, the depletion of Kir.4.1 did not increase the probability of finding the 23- and 60-pS K(+) channel in the CNT/CCD. We next used the perforated whole cell recording to measure the K(+) reversal voltage in the CNT/CCD as an index of cell membrane potential. Under control conditions, the K(+) reversal potential was -69 mV in WT mice and -61 mV in Kcnj10(-/-) mice, suggesting that Kir4.1 partially participates in generating membrane potential in the CNT/CCD. Western blotting and immunostaining showed that the expression of ENaCβ and ENaCγ subunits from a renal medulla section of Kcnj10(-/-) mice was significantly increased compared with that of WT mice. Also, the disruption of Kir4.1 increased aquaporin 2 expression. We conclude that Kir4.1 is expressed in the CNT and CCD and partially participates in generating the cell membrane potential. Also, increased ENaC expression in medullary CD of Kcnj10(-/-) mice is a compensatory action in response to the impaired Na(+) transport in the DCT.
Collapse
Affiliation(s)
- Xiao-Tong Su
- Department of Pharmacology, New York Medical College, Valhalla, New York; and
| | - Chengbiao Zhang
- Department of Pharmacology, New York Medical College, Valhalla, New York; and
| | - Lijun Wang
- Department of Pharmacology, New York Medical College, Valhalla, New York; and Department of Physiology, Harbin Medical University, Harbin, China
| | - Ruimin Gu
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Dao-Hong Lin
- Department of Pharmacology, New York Medical College, Valhalla, New York; and
| | - Wen-Hui Wang
- Department of Pharmacology, New York Medical College, Valhalla, New York; and
| |
Collapse
|
18
|
Tojo A, Kinugasa S, Fujita T, Wilcox CS. A local renal renin-angiotensin system activation via renal uptake of prorenin and angiotensinogen in diabetic rats. Diabetes Metab Syndr Obes 2016; 9:1-10. [PMID: 26848273 PMCID: PMC4723098 DOI: 10.2147/dmso.s91245] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The mechanism of activation of local renal renin-angiotensin system (RAS) has not been clarified in diabetes mellitus (DM). We hypothesized that the local renal RAS will be activated via increased glomerular filtration and tubular uptake of prorenin and angiotensinogen in diabetic kidney with microalbuminuria. Streptozotocin (STZ)-induced DM and control rats were injected with human prorenin and subsequently with human angiotensinogen. Human prorenin uptake was increased in podocytes, proximal tubules, macula densa, and cortical collecting ducts of DM rats where prorenin receptor (PRR) was expressed. Co-immunoprecipitation of kidney homogenates in DM rats revealed binding of human prorenin to the PRR and to megalin. The renal uptake of human angiotensinogen was increased in DM rats at the same nephron sites as prorenin. Angiotensin-converting enzyme was increased in podocytes, but decreased in the proximal tubules in DM rats, which may have contributed to unchanged renal levels of angiotensin despite increased angiotensinogen. The systolic blood pressure increased more after the injection of 20 μg of angiotensinogen in DM rats than in controls, accompanied by an increased uptake of human angiotensinogen in the vascular endothelium. In conclusion, endocytic uptake of prorenin and angiotensinogen in the kidney and vasculature in DM rats was contributed to increased tissue RAS and their pressor response to angiotensinogen.
Collapse
Affiliation(s)
- Akihiro Tojo
- Division of Nephrology and Endocrinology, The University of Tokyo, Tokyo, Japan
- Correspondence: Akihiro Tojo, Division of Nephrology and Endocrinology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan, Tel +81 3 3815 5411 ext 37219, Fax +81 3 3814 0021, Email
| | - Satoshi Kinugasa
- Division of Nephrology and Endocrinology, The University of Tokyo, Tokyo, Japan
| | - Toshiro Fujita
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Christopher S Wilcox
- Division of Nephrology and Hypertension, Center for Hypertension, Kidney and Vascular Research, Georgetown University, Washington, DC, USA
| |
Collapse
|
19
|
Abstract
More than two dozen types of potassium channels, with different biophysical and regulatory properties, are expressed in the kidney, influencing renal function in many important ways. Recently, a confluence of discoveries in areas from human genetics to physiology, cell biology, and biophysics has cast light on the special function of five different potassium channels in the distal nephron, encoded by the genes KCNJ1, KCNJ10, KCNJ16, KCNMA1, and KCNN3. Research aimed at understanding how these channels work in health and go awry in disease has transformed our understanding of potassium balance and provided new insights into mechanisms of renal sodium handling and the maintenance of blood pressure. This review focuses on recent advances in this rapidly evolving field.
Collapse
Affiliation(s)
- Paul A Welling
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201;
| |
Collapse
|
20
|
Villanueva S, Burgos J, López-Cayuqueo KI, Lai KMV, Valenzuela DM, Cid LP, Sepúlveda FV. Cleft Palate, Moderate Lung Developmental Retardation and Early Postnatal Lethality in Mice Deficient in the Kir7.1 Inwardly Rectifying K+ Channel. PLoS One 2015; 10:e0139284. [PMID: 26402555 PMCID: PMC4581704 DOI: 10.1371/journal.pone.0139284] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 09/09/2015] [Indexed: 12/15/2022] Open
Abstract
Kir7.1 is an inwardly rectifying K+ channel of the Kir superfamily encoded by the kcnj13 gene. Kir7.1 is present in epithelial tissues where it colocalizes with the Na+/K+-pump probably serving to recycle K+ taken up by the pump. Human mutations affecting Kir7.1 are associated with retinal degeneration diseases. We generated a mouse lacking Kir7.1 by ablation of the Kcnj13 gene. Homozygous mutant null mice die hours after birth and show cleft palate and moderate retardation in lung development. Kir7.1 is expressed in the epithelium covering the palatal processes at the time at which palate sealing takes place and our results suggest it might play an essential role in late palatogenesis. Our work also reveals a second unexpected role in the development and the physiology of the respiratory system, where Kir7.1 is expressed in epithelial cells all along the respiratory tree.
Collapse
Affiliation(s)
| | - Johanna Burgos
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Doctorado en Ciencias Veterinarias de la Universidad Austral de Chile, Valdivia, Chile
| | | | - Ka-Man Venus Lai
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, United States of America
| | - David M. Valenzuela
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, United States of America
| | - L. Pablo Cid
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | | |
Collapse
|
21
|
McCloskey C, Rada C, Bailey E, McCavera S, van den Berg HA, Atia J, Rand DA, Shmygol A, Chan YW, Quenby S, Brosens JJ, Vatish M, Zhang J, Denton JS, Taggart MJ, Kettleborough C, Tickle D, Jerman J, Wright P, Dale T, Kanumilli S, Trezise DJ, Thornton S, Brown P, Catalano R, Lin N, England SK, Blanks AM. The inwardly rectifying K+ channel KIR7.1 controls uterine excitability throughout pregnancy. EMBO Mol Med 2015; 6:1161-74. [PMID: 25056913 PMCID: PMC4197863 DOI: 10.15252/emmm.201403944] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Abnormal uterine activity in pregnancy causes a range of important clinical disorders, including preterm birth, dysfunctional labour and post-partum haemorrhage. Uterine contractile patterns are controlled by the generation of complex electrical signals at the myometrial smooth muscle plasma membrane. To identify novel targets to treat conditions associated with uterine dysfunction, we undertook a genome-wide screen of potassium channels that are enriched in myometrial smooth muscle. Computational modelling identified Kir7.1 as potentially important in regulating uterine excitability during pregnancy. We demonstrate Kir7.1 current hyper-polarizes uterine myocytes and promotes quiescence during gestation. Labour is associated with a decline, but not loss, of Kir7.1 expression. Knockdown of Kir7.1 by lentiviral expression of miRNA was sufficient to increase uterine contractile force and duration significantly. Conversely, overexpression of Kir7.1 inhibited uterine contractility. Finally, we demonstrate that the Kir7.1 inhibitor VU590 as well as novel derivative compounds induces profound, long-lasting contractions in mouse and human myometrium; the activity of these inhibitors exceeds that of other uterotonic drugs. We conclude Kir7.1 regulates the transition from quiescence to contractions in the pregnant uterus and may be a target for therapies to control uterine contractility.
Collapse
Affiliation(s)
- Conor McCloskey
- Division of Reproductive Health, Clinical Sciences Research Laboratories, Warwick Medical School University of Warwick, Coventry, UK
| | - Cara Rada
- Division of Basic Science Research, Department of Obstetrics and Gynecology, School of Medicine Washington University in St. Louis,, St. Louis, MO, USA
| | - Elizabeth Bailey
- Division of Reproductive Health, Clinical Sciences Research Laboratories, Warwick Medical School University of Warwick, Coventry, UK
| | - Samantha McCavera
- Division of Reproductive Health, Clinical Sciences Research Laboratories, Warwick Medical School University of Warwick, Coventry, UK
| | - Hugo A van den Berg
- Warwick Systems Biology & Mathematics Institute University of Warwick, Coventry, UK
| | - Jolene Atia
- Division of Reproductive Health, Clinical Sciences Research Laboratories, Warwick Medical School University of Warwick, Coventry, UK
| | - David A Rand
- Warwick Systems Biology & Mathematics Institute University of Warwick, Coventry, UK
| | - Anatoly Shmygol
- Division of Reproductive Health, Clinical Sciences Research Laboratories, Warwick Medical School University of Warwick, Coventry, UK
| | - Yi-Wah Chan
- Division of Reproductive Health, Clinical Sciences Research Laboratories, Warwick Medical School University of Warwick, Coventry, UK
| | - Siobhan Quenby
- Division of Reproductive Health, Clinical Sciences Research Laboratories, Warwick Medical School University of Warwick, Coventry, UK
| | - Jan J Brosens
- Division of Reproductive Health, Clinical Sciences Research Laboratories, Warwick Medical School University of Warwick, Coventry, UK
| | - Manu Vatish
- Division of Reproductive Health, Clinical Sciences Research Laboratories, Warwick Medical School University of Warwick, Coventry, UK
| | - Jie Zhang
- Division of Reproductive Health, Clinical Sciences Research Laboratories, Warwick Medical School University of Warwick, Coventry, UK
| | - Jerod S Denton
- Vanderbilt Institute of Chemical Biology, Vanderbilt Institute for Global Health Vanderbilt University School of Medicine Medical Center North, Nashville, TN, USA
| | - Michael J Taggart
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | | | - David Tickle
- Centre for Therapeutics and Discovery, Medical Research Council Technologies, London, UK
| | - Jeff Jerman
- Centre for Therapeutics and Discovery, Medical Research Council Technologies, London, UK
| | - Paul Wright
- Centre for Therapeutics and Discovery, Medical Research Council Technologies, London, UK
| | - Timothy Dale
- BioPark, Essen BioScience Ltd, Welwyn Garden City, Hertfordshire, UK
| | | | - Derek J Trezise
- BioPark, Essen BioScience Ltd, Welwyn Garden City, Hertfordshire, UK
| | | | - Pamela Brown
- MRC Centre for Reproductive Health (CRH), Queen's Medical Research Institute University of Edinburgh, Edinburgh, UK
| | - Roberto Catalano
- MRC Centre for Reproductive Health (CRH), Queen's Medical Research Institute University of Edinburgh, Edinburgh, UK
| | - Nan Lin
- Department of Mathematics, Washington University, St. Louis, MO, USA
| | - Sarah K England
- Division of Basic Science Research, Department of Obstetrics and Gynecology, School of Medicine Washington University in St. Louis,, St. Louis, MO, USA
| | - Andrew M Blanks
- Division of Reproductive Health, Clinical Sciences Research Laboratories, Warwick Medical School University of Warwick, Coventry, UK
| |
Collapse
|
22
|
Pattnaik BR, Shahi PK, Marino MJ, Liu X, York N, Brar S, Chiang J, Pillers DAM, Traboulsi EI. A Novel KCNJ13 Nonsense Mutation and Loss of Kir7.1 Channel Function Causes Leber Congenital Amaurosis (LCA16). Hum Mutat 2015; 36:720-7. [PMID: 25921210 DOI: 10.1002/humu.22807] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 04/20/2015] [Indexed: 11/08/2022]
Abstract
Mutations in the KCNJ13 gene that encodes the inwardly rectifying potassium channel Kir7.1 cause snowflake vitreoretinal degeneration (SVD) and leber congenital amaurosis (LCA). Kir7.1 controls the microenvironment between the photoreceptors and the retinal pigment epithelium (RPE) and also contributes to the function of other organs such as uterus and brain. Heterologous expressions of the mutant channel have suggested a dominant-negative loss of Kir7.1 function in SVD, but parallel studies in LCA16 have been lacking. Herein, we report the identification of a novel nonsense mutation in the second exon of the KCNJ13 gene that leads to a premature stop codon in association with LCA16. We have determined that the mutation results in a severe truncation of the Kir7.1 C-terminus, alters protein localization, and disrupts potassium currents. Coexpression of the mutant and wild-type channel has no negative influence on the wild-type channel function, consistent with the normal clinical phenotype of carrier individuals. By suppressing Kir7.1 function in mice, we were able to reproduce the severe LCA electroretinogram phenotype. Thus, we have extended the observation that Kir7.1 mutations are associated with vision disorders to include novel insights into the molecular mechanism of disease pathobiology in LCA16.
Collapse
Affiliation(s)
- Bikash R Pattnaik
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin.,Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, Wisconsin.,McPherson Eye Research Institute, University of Wisconsin, Madison, Wisconsin
| | - Pawan K Shahi
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin
| | - Meghan J Marino
- Center for Genetic Eye Diseases and Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Xinying Liu
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin
| | - Nathaniel York
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin
| | - Simran Brar
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin
| | - John Chiang
- Casey Molecular Diagnostic Laboratory, Oregon Health & Science University, Portland, Oregon
| | - De-Ann M Pillers
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin.,McPherson Eye Research Institute, University of Wisconsin, Madison, Wisconsin
| | - Elias I Traboulsi
- Center for Genetic Eye Diseases and Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| |
Collapse
|
23
|
Sepúlveda FV, Pablo Cid L, Teulon J, Niemeyer MI. Molecular aspects of structure, gating, and physiology of pH-sensitive background K2P and Kir K+-transport channels. Physiol Rev 2015; 95:179-217. [PMID: 25540142 DOI: 10.1152/physrev.00016.2014] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
K(+) channels fulfill roles spanning from the control of excitability to the regulation of transepithelial transport. Here we review two groups of K(+) channels, pH-regulated K2P channels and the transport group of Kir channels. After considering advances in the molecular aspects of their gating based on structural and functional studies, we examine their participation in certain chosen physiological and pathophysiological scenarios. Crystal structures of K2P and Kir channels reveal rather unique features with important consequences for the gating mechanisms. Important tasks of these channels are discussed in kidney physiology and disease, K(+) homeostasis in the brain by Kir channel-equipped glia, and central functions in the hearing mechanism in the inner ear and in acid secretion by parietal cells in the stomach. K2P channels fulfill a crucial part in central chemoreception probably by virtue of their pH sensitivity and are central to adrenal secretion of aldosterone. Finally, some unorthodox behaviors of the selectivity filters of K2P channels might explain their normal and pathological functions. Although a great deal has been learned about structure, molecular details of gating, and physiological functions of K2P and Kir K(+)-transport channels, this has been only scratching at the surface. More molecular and animal studies are clearly needed to deepen our knowledge.
Collapse
Affiliation(s)
- Francisco V Sepúlveda
- Centro de Estudios Científicos, Valdivia, Chile; UPMC Université Paris 06, Team 3, Paris, France; and Institut National de la Santé et de la Recherche Médicale, UMR_S 1138, Paris, France
| | - L Pablo Cid
- Centro de Estudios Científicos, Valdivia, Chile; UPMC Université Paris 06, Team 3, Paris, France; and Institut National de la Santé et de la Recherche Médicale, UMR_S 1138, Paris, France
| | - Jacques Teulon
- Centro de Estudios Científicos, Valdivia, Chile; UPMC Université Paris 06, Team 3, Paris, France; and Institut National de la Santé et de la Recherche Médicale, UMR_S 1138, Paris, France
| | - María Isabel Niemeyer
- Centro de Estudios Científicos, Valdivia, Chile; UPMC Université Paris 06, Team 3, Paris, France; and Institut National de la Santé et de la Recherche Médicale, UMR_S 1138, Paris, France
| |
Collapse
|
24
|
Zhang C, Wang L, Zhang J, Su XT, Lin DH, Scholl UI, Giebisch G, Lifton RP, Wang WH. KCNJ10 determines the expression of the apical Na-Cl cotransporter (NCC) in the early distal convoluted tubule (DCT1). Proc Natl Acad Sci U S A 2014; 111:11864-9. [PMID: 25071208 PMCID: PMC4136599 DOI: 10.1073/pnas.1411705111] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The renal phenotype induced by loss-of-function mutations of inwardly rectifying potassium channel (Kir), Kcnj10 (Kir4.1), includes salt wasting, hypomagnesemia, metabolic alkalosis and hypokalemia. However, the mechanism by which Kir.4.1 mutations cause the tubulopathy is not completely understood. Here we demonstrate that Kcnj10 is a main contributor to the basolateral K conductance in the early distal convoluted tubule (DCT1) and determines the expression of the apical Na-Cl cotransporter (NCC) in the DCT. Immunostaining demonstrated Kcnj10 and Kcnj16 were expressed in the basolateral membrane of DCT, and patch-clamp studies detected a 40-pS K channel in the basolateral membrane of the DCT1 of p8/p10 wild-type Kcnj10(+/+) mice (WT). This 40-pS K channel is absent in homozygous Kcnj10(-/-) (knockout) mice. The disruption of Kcnj10 almost completely eliminated the basolateral K conductance and decreased the negativity of the cell membrane potential in DCT1. Moreover, the lack of Kcnj10 decreased the basolateral Cl conductance, inhibited the expression of Ste20-related proline-alanine-rich kinase and diminished the apical NCC expression in DCT. We conclude that Kcnj10 plays a dominant role in determining the basolateral K conductance and membrane potential of DCT1 and that the basolateral K channel activity in the DCT determines the apical NCC expression possibly through a Ste20-related proline-alanine-rich kinase-dependent mechanism.
Collapse
Affiliation(s)
- Chengbiao Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou 221002, China;Department of Pharmacology, New York Medical College, Valhalla, NY 10595; and
| | - Lijun Wang
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595; and
| | - Junhui Zhang
- Department of Genetics, Howard Hughes Medical Institute, and
| | - Xiao-Tong Su
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595; and
| | - Dao-Hong Lin
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595; and
| | - Ute I Scholl
- Department of Genetics, Howard Hughes Medical Institute, and
| | - Gerhard Giebisch
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510
| | | | - Wen-Hui Wang
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595; and
| |
Collapse
|
25
|
Swale DR, Kharade SV, Denton JS. Cardiac and renal inward rectifier potassium channel pharmacology: emerging tools for integrative physiology and therapeutics. Curr Opin Pharmacol 2013; 15:7-15. [PMID: 24721648 DOI: 10.1016/j.coph.2013.11.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 11/05/2013] [Indexed: 01/01/2023]
Abstract
Inward rectifier potassium (Kir) channels play fundamental roles in cardiac and renal function and may represent unexploited drug targets for cardiovascular diseases. However, the limited pharmacology of Kir channels has slowed progress toward exploring their integrative physiology and therapeutic potential. Here, we review recent progress toward developing the small-molecule pharmacology for Kir2.x, Kir4.1, and Kir7.1 and discuss common mechanistic themes that may help guide future Kir channel-directed drug discovery efforts.
Collapse
Affiliation(s)
- Daniel R Swale
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Sujay V Kharade
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Jerod S Denton
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, United States; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, United States; Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, TN 37232, United States; Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN 37232, United States.
| |
Collapse
|
26
|
Denton JS, Pao AC, Maduke M. Novel diuretic targets. Am J Physiol Renal Physiol 2013; 305:F931-42. [PMID: 23863472 PMCID: PMC3798746 DOI: 10.1152/ajprenal.00230.2013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 07/12/2013] [Indexed: 01/11/2023] Open
Abstract
As the molecular revolution continues to inform a deeper understanding of disease mechanisms and pathways, there exist unprecedented opportunities for translating discoveries at the bench into novel therapies for improving human health. Despite the availability of several different classes of antihypertensive medications, only about half of the 67 million Americans with hypertension manage their blood pressure appropriately. A broader selection of structurally diverse antihypertensive drugs acting through different mechanisms would provide clinicians with greater flexibility in developing effective treatment regimens for an increasingly diverse and aging patient population. An emerging body of physiological, genetic, and pharmacological evidence has implicated several renal ion-transport proteins, or regulators thereof, as novel, yet clinically unexploited, diuretic targets. These include the renal outer medullary potassium channel, ROMK (Kir1.1), Kir4.1/5.1 potassium channels, ClC-Ka/b chloride channels, UTA/B urea transporters, the chloride/bicarbonate exchanger pendrin, and the STE20/SPS1-related proline/alanine-rich kinase (SPAK). The molecular pharmacology of these putative targets is poorly developed or lacking altogether; however, recent efforts by a few academic and pharmaceutical laboratories have begun to lessen this critical barrier. Here, we review the evidence in support of the aforementioned proteins as novel diuretic targets and highlight examples where progress toward developing small-molecule pharmacology has been made.
Collapse
Affiliation(s)
- Jerod S Denton
- T4208 Medical Center North, 1161 21st Ave. South, Nashville, TN 37232.
| | | | | |
Collapse
|
27
|
Pattnaik BR, Tokarz S, Asuma MP, Schroeder T, Sharma A, Mitchell JC, Edwards AO, Pillers DAM. Snowflake vitreoretinal degeneration (SVD) mutation R162W provides new insights into Kir7.1 ion channel structure and function. PLoS One 2013; 8:e71744. [PMID: 23977131 PMCID: PMC3747230 DOI: 10.1371/journal.pone.0071744] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 07/02/2013] [Indexed: 01/08/2023] Open
Abstract
Snowflake Vitreoretinal Degeneration (SVD) is associated with the R162W mutation of the Kir7.1 inwardly-rectifying potassium channel. Kir7.1 is found at the apical membrane of Retinal Pigment Epithelial (RPE) cells, adjacent to the photoreceptor neurons. The SVD phenotype ranges from RPE degeneration to an abnormal b-wave to a liquid vitreous. We sought to determine how this mutation alters the structure and function of the human Kir7.1 channel. In this study, we expressed a Kir7.1 construct with the R162W mutation in CHO cells to evaluate function of the ion channel. Compared to the wild-type protein, the mutant protein exhibited a non-functional Kir channel that resulted in depolarization of the resting membrane potential. Upon co-expression with wild-type Kir7.1, R162W mutant showed a reduction of IKir7.1 and positive shift in ‘0’ current potential. Homology modeling based on the structure of a bacterial Kir channel protein suggested that the effect of R162W mutation is a result of loss of hydrogen bonding by the regulatory lipid binding domain of the cytoplasmic structure.
Collapse
Affiliation(s)
- Bikash R. Pattnaik
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, Wisconsin, United States of America
- McPherson Eye Research Institute, University of Wisconsin, Madison, Wisconsin, United States of America
- * E-mail:
| | - Sara Tokarz
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Matti P. Asuma
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Tyler Schroeder
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Anil Sharma
- Department of Experimental Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Julie C. Mitchell
- Department of Biochemistry, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Albert O. Edwards
- Institute for Molecular Biology, University of Oregon, and Oregon Retina, Eugene, Oregon, United States of America
| | - De-Ann M. Pillers
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, United States of America
- McPherson Eye Research Institute, University of Wisconsin, Madison, Wisconsin, United States of America
| |
Collapse
|
28
|
Zhou X, Zhang Z, Shin MK, Horwitz SB, Levorse JM, Zhu L, Sharif-Rodriguez W, Streltsov DY, Dajee M, Hernandez M, Pan Y, Urosevic-Price O, Wang L, Forrest G, Szeto D, Zhu Y, Cui Y, Michael B, Balogh LA, Welling PA, Wade JB, Roy S, Sullivan KA. Heterozygous disruption of renal outer medullary potassium channel in rats is associated with reduced blood pressure. Hypertension 2013; 62:288-94. [PMID: 23753405 DOI: 10.1161/hypertensionaha.111.01051] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The renal outer medullary potassium channel (ROMK, KCNJ1) mediates potassium recycling and facilitates sodium reabsorption through the Na(+)/K(+)/2Cl(-) cotransporter in the loop of Henle and potassium secretion at the cortical collecting duct. Human genetic studies indicate that ROMK homozygous loss-of-function mutations cause type II Bartter syndrome, featuring polyuria, renal salt wasting, and hypotension; humans heterozygous for ROMK mutations identified in the Framingham Heart Study have reduced blood pressure. ROMK null mice recapitulate many of the features of type II Bartter syndrome. We have generated an ROMK knockout rat model in Dahl salt-sensitive background by using zinc finger nuclease technology and investigated the effects of knocking out ROMK on systemic and renal hemodynamics and kidney histology in the Dahl salt-sensitive rats. The ROMK(-/-) pups recapitulated features identified in the ROMK null mice. The ROMK(+/-) rats, when challenged with a 4% salt diet, exhibited a reduced blood pressure compared with their ROMK(+/+) littermates. More importantly, when challenged with an 8% salt diet, the Dahl salt-sensitive rats with 50% less ROMK expression showed increased protection from salt-induced blood pressure elevation and signs of protection from renal injury. Our findings in ROMK knockout Dahl salt-sensitive rats, together with the previous reports in humans and mice, underscore a critical role of ROMK in blood pressure regulation.
Collapse
Affiliation(s)
- Xiaoyan Zhou
- Department of Cardiovascular Diseases, Merck Research Laboratories, 126 E Lincoln Ave, Rahway, NJ 07065, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Zhang W, Zhang X, Wang H, Sharma AK, Edwards AO, Hughes BA. Characterization of the R162W Kir7.1 mutation associated with snowflake vitreoretinopathy. Am J Physiol Cell Physiol 2012; 304:C440-9. [PMID: 23255580 DOI: 10.1152/ajpcell.00363.2012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
KCNJ13 encodes Kir7.1, an inwardly rectifying K(+) channel that is expressed in multiple ion-transporting epithelia. A mutation in KCNJ13 resulting in an arginine-to-tryptophan change at residue 162 (R162W) of Kir7.1 was associated with snowflake vitreoretinal degeneration, an inherited autosomal-dominant disease characterized by vitreous degeneration and mild retinal degeneration. We used the Xenopus laevis oocyte expression system to assess the functional properties of the R162W (mutant) Kir7.1 channel and determine how wild-type (WT) Kir7.1 is affected by the presence of the mutant subunit. Recordings obtained via the two-electrode voltage-clamp technique revealed that injection of oocytes with mutant Kir7.1 cRNA resulted in currents and cation selectivity that were indistinguishable from those in water-injected oocytes, suggesting that the mutant protein does not form functional channels in the plasma membrane. Coinjection of oocytes with equal amounts of mutant and WT Kir7.1 cRNAs resulted in inward K(+) and Rb(+) currents with amplitudes that were ∼17% of those in oocytes injected with WT Kir7.1 cRNA alone, demonstrating a dominant-negative effect of the mutant subunit. Similar to oocytes injected with WT Kir7.1 cRNA alone, coinjected oocytes exhibited inwardly rectifying Rb(+) currents that were more than seven times larger than K(+) currents, indicating that mutant subunits did not alter Kir7.1 channel selectivity. Immunostaining of Xenopus oocytes or Madin-Darby canine kidney cells expressing mutant or WT Kir7.1 demonstrated distribution of both proteins primarily in the plasma membrane. Our data suggest that the R162W mutation suppresses Kir7.1 channel activity, possibly by negatively impacting gating by membrane phosphadidylinositol 4,5-bisphosphate.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | | | | | | | | | | |
Collapse
|
30
|
Felix JP, Priest BT, Solly K, Bailey T, Brochu RM, Liu CJ, Kohler MG, Kiss L, Alonso-Galicia M, Tang H, Pasternak A, Kaczorowski GJ, Garcia ML. The Inwardly Rectifying Potassium Channel Kir1.1: Development of Functional Assays to Identify and Characterize Channel Inhibitors. Assay Drug Dev Technol 2012; 10:417-31. [DOI: 10.1089/adt.2012.462] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- John P. Felix
- Department of Ion Channels, Merck Research Laboratories, North Wales, Pennsylvania
| | - Birgit T. Priest
- Department of Ion Channels, Merck Research Laboratories, North Wales, Pennsylvania
| | - Kelli Solly
- Department of Automated Biotechnology, Merck Research Laboratories, North Wales, Pennsylvania
| | - Timothy Bailey
- Department of Ion Channels, Merck Research Laboratories, North Wales, Pennsylvania
| | - Richard M. Brochu
- Department of Ion Channels, Merck Research Laboratories, North Wales, Pennsylvania
| | - Chou J. Liu
- Department of Ion Channels, Merck Research Laboratories, North Wales, Pennsylvania
| | - Martin G. Kohler
- Department of Ion Channels, Merck Research Laboratories, North Wales, Pennsylvania
| | - Laszlo Kiss
- Department of Automated Biotechnology, Merck Research Laboratories, North Wales, Pennsylvania
| | | | - Haifeng Tang
- Department of Medicinal Chemistry, Merck Research Laboratories, Rahway, New Jersey
| | - Alexander Pasternak
- Department of Medicinal Chemistry, Merck Research Laboratories, Rahway, New Jersey
| | | | - Maria L. Garcia
- Department of Ion Channels, Merck Research Laboratories, North Wales, Pennsylvania
| |
Collapse
|
31
|
Hamilton KL, Devor DC. Basolateral membrane K+ channels in renal epithelial cells. Am J Physiol Renal Physiol 2012; 302:F1069-81. [PMID: 22338089 DOI: 10.1152/ajprenal.00646.2011] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The major function of epithelial tissues is to maintain proper ion, solute, and water homeostasis. The tubule of the renal nephron has an amazingly simple structure, lined by epithelial cells, yet the segments (i.e., proximal tubule vs. collecting duct) of the nephron have unique transport functions. The functional differences are because epithelial cells are polarized and thus possess different patterns (distributions) of membrane transport proteins in the apical and basolateral membranes of the cell. K(+) channels play critical roles in normal physiology. Over 90 different genes for K(+) channels have been identified in the human genome. Epithelial K(+) channels can be located within either or both the apical and basolateral membranes of the cell. One of the primary functions of basolateral K(+) channels is to recycle K(+) across the basolateral membrane for proper function of the Na(+)-K(+)-ATPase, among other functions. Mutations of these channels can cause significant disease. The focus of this review is to provide an overview of the basolateral K(+) channels of the nephron, providing potential physiological functions and pathophysiology of these channels, where appropriate. We have taken a "K(+) channel gene family" approach in presenting the representative basolateral K(+) channels of the nephron. The basolateral K(+) channels of the renal epithelia are represented by members of the KCNK, KCNJ, KCNQ, KCNE, and SLO gene families.
Collapse
Affiliation(s)
- Kirk L Hamilton
- Department of Physiology, Otago School of Medical Sciences, University of Otago, PO Box 913, Dunedin, New Zealand.
| | | |
Collapse
|
32
|
Raphemot R, Lonergan DF, Nguyen TT, Utley T, Lewis LM, Kadakia R, Weaver CD, Gogliotti R, Hopkins C, Lindsley CW, Denton JS. Discovery, characterization, and structure-activity relationships of an inhibitor of inward rectifier potassium (Kir) channels with preference for Kir2.3, Kir3.x, and Kir7.1. Front Pharmacol 2011; 2:75. [PMID: 22275899 PMCID: PMC3254186 DOI: 10.3389/fphar.2011.00075] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 11/07/2011] [Indexed: 12/03/2022] Open
Abstract
The inward rectifier family of potassium (Kir) channels is comprised of at least 16 family members exhibiting broad and often overlapping cellular, tissue, or organ distributions. The discovery of disease-causing mutations in humans and experiments on knockout mice has underscored the importance of Kir channels in physiology and in some cases raised questions about their potential as drug targets. However, the paucity of potent and selective small-molecule modulators targeting specific family members has with few exceptions mired efforts to understand their physiology and assess their therapeutic potential. A growing body of evidence suggests that G protein-coupled inward rectifier K (GIRK) channels of the Kir3.X subfamily may represent novel targets for the treatment of atrial fibrillation. In an effort to expand the molecular pharmacology of GIRK, we performed a thallium (Tl(+)) flux-based high-throughput screen of a Kir1.1 inhibitor library for modulators of GIRK. One compound, termed VU573, exhibited 10-fold selectivity for GIRK over Kir1.1 (IC(50) = 1.9 and 19 μM, respectively) and was therefore selected for further study. In electrophysiological experiments performed on Xenopus laevis oocytes and mammalian cells, VU573 inhibited Kir3.1/3.2 (neuronal GIRK) and Kir3.1/3.4 (cardiac GIRK) channels with equal potency and preferentially inhibited GIRK, Kir2.3, and Kir7.1 over Kir1.1 and Kir2.1.Tl(+) flux assays were established for Kir2.3 and the M125R pore mutant of Kir7.1 to support medicinal chemistry efforts to develop more potent and selective analogs for these channels. The structure-activity relationships of VU573 revealed few analogs with improved potency, however two compounds retained most of their activity toward GIRK and Kir2.3 and lost activity toward Kir7.1. We anticipate that the VU573 series will be useful for exploring the physiology and structure-function relationships of these Kir channels.
Collapse
Affiliation(s)
- Rene Raphemot
- Department of Anesthesiology, Vanderbilt University School of MedicineNashville, TN, USA
- Department of Pharmacology, Vanderbilt University School of MedicineNashville, TN, USA
| | - Daniel F. Lonergan
- Department of Anesthesiology, Vanderbilt University School of MedicineNashville, TN, USA
| | - Thuy T. Nguyen
- Department of Anesthesiology, Vanderbilt University School of MedicineNashville, TN, USA
- Department of Pharmacology, Vanderbilt University School of MedicineNashville, TN, USA
| | - Thomas Utley
- Department of Pharmacology, Vanderbilt University School of MedicineNashville, TN, USA
| | - L. Michelle Lewis
- Vanderbilt Institute of Chemical Biology, Vanderbilt UniversityNashville, TN, USA
| | - Rishin Kadakia
- Department of Anesthesiology, Vanderbilt University School of MedicineNashville, TN, USA
| | - C. David Weaver
- Department of Pharmacology, Vanderbilt University School of MedicineNashville, TN, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt UniversityNashville, TN, USA
| | - Rocco Gogliotti
- Department of Pharmacology, Vanderbilt University School of MedicineNashville, TN, USA
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of MedicineNashville, TN, USA
| | - Corey Hopkins
- Department of Pharmacology, Vanderbilt University School of MedicineNashville, TN, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt UniversityNashville, TN, USA
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of MedicineNashville, TN, USA
- Department of Chemistry, Vanderbilt UniversityNashville, TN, USA
- Vanderbilt Specialized Chemistry Center for Accelerated Probe Development, Molecular Libraries Probe Production Centers NetworkNashville, TN, USA
| | - Craig W. Lindsley
- Department of Pharmacology, Vanderbilt University School of MedicineNashville, TN, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt UniversityNashville, TN, USA
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of MedicineNashville, TN, USA
- Department of Chemistry, Vanderbilt UniversityNashville, TN, USA
- Vanderbilt Specialized Chemistry Center for Accelerated Probe Development, Molecular Libraries Probe Production Centers NetworkNashville, TN, USA
| | - Jerod S. Denton
- Department of Anesthesiology, Vanderbilt University School of MedicineNashville, TN, USA
- Department of Pharmacology, Vanderbilt University School of MedicineNashville, TN, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt UniversityNashville, TN, USA
| |
Collapse
|
33
|
Selective albuminuria via podocyte albumin transport in puromycin nephrotic rats is attenuated by an inhibitor of NADPH oxidase. Kidney Int 2011; 80:1328-38. [PMID: 21849973 DOI: 10.1038/ki.2011.282] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The mechanism of selective albuminuria in minimal change nephrotic syndrome, in which glomerular capillaries are diffusely covered by effaced podocyte foot processes with reduced slit diaphragms, is unknown. Podocyte injury is due, in part, to NADPH-induced oxidative stress. Here we studied mechanism of selective albuminuria in puromycin aminonucleoside (PAN) nephrotic rats, a model of minimal change nephrotic syndrome. In these rats, Evans Blue-labeled human albumin was taken up by podocytes and its urinary excretion markedly increased, with retained selectivity for albumin. Immunogold scanning electron micrographic images found increased human albumin in podocyte vesicles and on the apical membrane in nephrotic compared with control rats. Apocynin, an inhibitor of NADPH oxidase, decreased superoxide production in podocytes, and inhibited endocytosis and urinary albumin excretion. Real-time confocal microscopy found an initial delay in the appearance of Evans Blue-labeled human albumin in the tubular lumen, reflecting the time needed for transcellular transport. Immunoprecipitation analysis indicated that FcRn, a receptor for albumin transport, mediated podocyte albumin transport, and treatment with anti-FcRn antibody reduced proteinuria in these nephrotic rats. Thus, podocyte albumin transport was enhanced in PAN nephrotic rats by means of FcRn, which may explain the mechanism of selective proteinuria. This was blocked by apocynin, suggesting a new therapeutic approach.
Collapse
|
34
|
Small-molecule modulators of inward rectifier K+ channels: recent advances and future possibilities. Future Med Chem 2011; 2:757-74. [PMID: 20543968 DOI: 10.4155/fmc.10.179] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Inward rectifier potassium (Kir) channels have been postulated as therapeutic targets for several common disorders including hypertension, cardiac arrhythmias and pain. With few exceptions, however, the small-molecule pharmacology of this family is limited to nonselective cardiovascular and neurologic drugs with off-target activity toward inward rectifiers. Consequently, the actual therapeutic potential and 'drugability' of most Kir channels has not yet been determined experimentally. The purpose of this review is to provide a comprehensive summary of publicly disclosed Kir channel small-molecule modulators and highlight recent targeted drug-discovery efforts toward Kir1.1 and Kir2.1. The review concludes with a brief speculation on how the field of Kir channel pharmacology will develop over the coming years and a discussion of the increasingly important role academic laboratories will play in this progress.
Collapse
|
35
|
Cha SK, Huang C, Ding Y, Qi X, Huang CL, Miller RT. Calcium-sensing receptor decreases cell surface expression of the inwardly rectifying K+ channel Kir4.1. J Biol Chem 2011; 286:1828-35. [PMID: 21084311 PMCID: PMC3023478 DOI: 10.1074/jbc.m110.160390] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 11/07/2010] [Indexed: 11/06/2022] Open
Abstract
The Ca(2+)-sensing receptor (CaR) regulates salt and water transport in the kidney as demonstrated by the association of gain of function CaR mutations with a Bartter syndrome-like, salt-wasting phenotype, but the precise mechanism for this effect is not fully established. We found previously that the CaR interacts with and inactivates an inwardly rectifying K(+) channel, Kir4.1, which is expressed in the distal nephron that contributes to the basolateral K(+) conductance, and in which loss of function mutations are associated with a complex phenotype that includes renal salt wasting. We now find that CaR inactivates Kir4.1 by reducing its cell surface expression. Mutant CaRs reduced Kir4.1 cell surface expression and current density in HEK-293 cells in proportion to their signaling activity. Mutant, activated Gα(q) reduced cell surface expression and current density of Kir4.1, and these effects were blocked by RGS4, a protein that blocks signaling via Gα(i) and Gα(q). Other α subunits had insignificant effects. Knockdown of caveolin-1 blocked the effect of Gα(q) on Kir4.1, whereas knockdown of the clathrin heavy chain had no effect. CaR had no comparable effect on the renal outer medullary K(+) channel, an apical membrane distal nephron K(+) channel that is internalized by clathrin-coated vesicles. Co-immunoprecipitation studies showed that the CaR and Kir4.1 physically associate with caveolin-1 in HEK cells and in kidney extracts. Thus, the CaR decreases cell surface expression of Kir4.1 channels via a mechanism that involves Gα(q) and caveolin. These results provide a novel molecular basis for the inhibition of renal NaCl transport by the CaR.
Collapse
Affiliation(s)
- Seung-Kuy Cha
- From the Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Chunfa Huang
- the Departments of Medicine and
- the Louis Stokes Veteran Affairs Medical Center, Cleveland, Ohio 44106, and
| | | | | | - Chou-Long Huang
- From the Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - R. Tyler Miller
- the Departments of Medicine and
- Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio 44106
- the Louis Stokes Veteran Affairs Medical Center, Cleveland, Ohio 44106, and
- the Rammelkamp Center for Research and Education, MetroHealth System Campus, Cleveland, Ohio 44109
| |
Collapse
|
36
|
Bhave G, Chauder BA, Liu W, Dawson ES, Kadakia R, Nguyen TT, Lewis LM, Meiler J, Weaver CD, Satlin LM, Lindsley CW, Denton JS. Development of a selective small-molecule inhibitor of Kir1.1, the renal outer medullary potassium channel. Mol Pharmacol 2011; 79:42-50. [PMID: 20926757 PMCID: PMC3014278 DOI: 10.1124/mol.110.066928] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Accepted: 10/06/2010] [Indexed: 11/22/2022] Open
Abstract
The renal outer medullary potassium (K+) channel, ROMK (Kir1.1), is a putative drug target for a novel class of loop diuretic that would lower blood volume and pressure without causing hypokalemia. However, the lack of selective ROMK inhibitors has hindered efforts to assess its therapeutic potential. In a high-throughput screen for small-molecule modulators of ROMK, we previously identified a potent and moderately selective ROMK antagonist, 7,13-bis(4-nitrobenzyl)-1,4,10-trioxa-7,13-diazacyclopentadecane (VU590), that also inhibits Kir7.1. Because ROMK and Kir7.1 are coexpressed in the nephron, VU590 is not a good probe of ROMK function in the kidney. Here we describe the development of the structurally related inhibitor 2,2'-oxybis(methylene)bis(5-nitro-1H-benzo[d]imidazole) (VU591), which is as potent as VU590 but is selective for ROMK over Kir7.1 and more than 65 other potential off-targets. VU591 seems to block the intracellular pore of the channel. The development of VU591 may enable studies to explore the viability of ROMK as a diuretic target.
Collapse
Affiliation(s)
- Gautam Bhave
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Hibino H, Inanobe A, Furutani K, Murakami S, Findlay I, Kurachi Y. Inwardly rectifying potassium channels: their structure, function, and physiological roles. Physiol Rev 2010; 90:291-366. [PMID: 20086079 DOI: 10.1152/physrev.00021.2009] [Citation(s) in RCA: 1135] [Impact Index Per Article: 75.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Inwardly rectifying K(+) (Kir) channels allow K(+) to move more easily into rather than out of the cell. They have diverse physiological functions depending on their type and their location. There are seven Kir channel subfamilies that can be classified into four functional groups: classical Kir channels (Kir2.x) are constitutively active, G protein-gated Kir channels (Kir3.x) are regulated by G protein-coupled receptors, ATP-sensitive K(+) channels (Kir6.x) are tightly linked to cellular metabolism, and K(+) transport channels (Kir1.x, Kir4.x, Kir5.x, and Kir7.x). Inward rectification results from pore block by intracellular substances such as Mg(2+) and polyamines. Kir channel activity can be modulated by ions, phospholipids, and binding proteins. The basic building block of a Kir channel is made up of two transmembrane helices with cytoplasmic NH(2) and COOH termini and an extracellular loop which folds back to form the pore-lining ion selectivity filter. In vivo, functional Kir channels are composed of four such subunits which are either homo- or heterotetramers. Gene targeting and genetic analysis have linked Kir channel dysfunction to diverse pathologies. The crystal structure of different Kir channels is opening the way to understanding the structure-function relationships of this simple but diverse ion channel family.
Collapse
Affiliation(s)
- Hiroshi Hibino
- Department of Pharmacology, Graduate School of Medicine and The Center for Advanced Medical Engineering and Informatics, Osaka University, Osaka 565-0871, Japan
| | | | | | | | | | | |
Collapse
|
38
|
Lewis LM, Bhave G, Chauder BA, Banerjee S, Lornsen KA, Redha R, Fallen K, Lindsley CW, Weaver CD, Denton JS. High-throughput screening reveals a small-molecule inhibitor of the renal outer medullary potassium channel and Kir7.1. Mol Pharmacol 2009; 76:1094-103. [PMID: 19706730 PMCID: PMC2774996 DOI: 10.1124/mol.109.059840] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Accepted: 08/25/2009] [Indexed: 11/22/2022] Open
Abstract
The renal outer medullary potassium channel (ROMK) is expressed in the kidney tubule and critically regulates sodium and potassium balance. The physiological functions of other inward rectifying K(+) (Kir) channels expressed in the nephron, such as Kir7.1, are less well understood in part due to the lack of selective pharmacological probes targeting inward rectifiers. In an effort to identify Kir channel probes, we performed a fluorescence-based, high-throughput screen (HTS) of 126,009 small molecules for modulators of ROMK function. Several antagonists were identified in the screen. One compound, termed VU590, inhibits ROMK with submicromolar affinity, but has no effect on Kir2.1 or Kir4.1. Low micromolar concentrations inhibit Kir7.1, making VU590 the first small-molecule inhibitor of Kir7.1. Structure-activity relationships of VU590 were defined using small-scale parallel synthesis. Electrophysiological analysis indicates that VU590 is an intracellular pore blocker. VU590 and other compounds identified by HTS will be instrumental in defining Kir channel structure, physiology, and therapeutic potential.
Collapse
Affiliation(s)
- L Michelle Lewis
- Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sindic A, Huang C, Chen AP, Ding Y, Miller-Little WA, Che D, Romero MF, Miller RT. MUPP1 complexes renal K+ channels to alter cell surface expression and whole cell currents. Am J Physiol Renal Physiol 2009; 297:F36-45. [PMID: 19420109 DOI: 10.1152/ajprenal.90559.2008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We previously found that the Ca(2+)-sensing receptor (CaR) interacts with and inactivates the inwardly rectifying K(+) channel Kir4.2 that is expressed in the kidney cortex and that has a COOH-terminal PDZ domain. To identify potential scaffolding proteins that could organize a macromolecular signaling complex involving the CaR and Kir4.2, we used yeast two-hybrid cloning with the COOH-terminal 125 amino acids (AA) of Kir4.2 as bait to screen a human kidney cDNA library. We identified two independent partial cDNAs corresponding to the COOH-terminal 900 AA of MUPP1, a protein containing 13 PDZ binding domains that is expressed in the kidney in tight junctions and lateral borders of epithelial cells. When expressed in human embryonic kidney (HEK)-293 cells, Kir4.2 coimmunoprecipitates reciprocally with MUPP1 but not with a Kir4.2 construct lacking the four COOH-terminal amino acids, Kir5.1, or the CaR. MUPP1 and Kir4.2 coimmunoprecipitate reciprocally from rat kidney cortex extracts. Coexpression of MUPP1 with Kir4.2 in HEK-293 cells leads to reduced cell surface expression of Kir4.2 as assessed by cell surface biotinylation. Coexpression of MUPP1 and Kir4.2 in Xenopus oocytes results in reduced whole cell currents compared with expression of Kir4.2 alone, whereas expression of Kir4.2DeltaPDZ results in minimal currents and is not affected by coexpression with MUPP1. Immunofluorescence studies of oocytes demonstrate that MUPP1 reduces Kir4.2 membrane localization. These results indicate that Kir4.2 interacts selectively with MUPP1 to affect its cell surface expression. Thus MUPP1 and Kir4.2 may participate in a protein complex in the nephron that could regulate transport of K(+) as well as other ions.
Collapse
Affiliation(s)
- Aleksandra Sindic
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Zhang W, Zitron E, Bloehs R, Müller-Krebs S, Scholz E, Zeier M, Katus H, Karle C, Schwenger V. Dual regulation of renal Kir7.1 potassium channels by protein Kinase A and protein Kinase C. Biochem Biophys Res Commun 2008; 377:981-6. [DOI: 10.1016/j.bbrc.2008.10.110] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Accepted: 10/19/2008] [Indexed: 10/24/2022]
|
41
|
Frindt G, Shah A, Edvinsson J, Palmer LG. Dietary K regulates ROMK channels in connecting tubule and cortical collecting duct of rat kidney. Am J Physiol Renal Physiol 2008; 296:F347-54. [PMID: 19036846 DOI: 10.1152/ajprenal.90527.2008] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The activity of ROMK channels in rat kidney tubule cells was assessed as tertiapin-Q (TPNQ)-sensitive current under whole cell clamp conditions. With an external K(+) concentration of 5 mM and an internal K(+) concentration of 140 mM and the membrane potential clamped to 0 mV, TPNQ blocked outward currents in principal cells of the cortical collecting duct (CCD) outer medullary collecting duct and connecting tubule (CNT). The apparent K(i) was 5.0 nM, consistent with its interaction with ROMK. The TPNQ-sensitive current reversed at voltages close to the equilibrium potential for K(+). The currents were reduced when the pipette solution contained ATP. In the CCD, the average TPNQ-sensitive outward current (I(SK)) was 476 +/- 48 pA/cell in control animals on a 1% KCl diet. I(SK) increased to 1,255 +/- 140 pA when animals were maintained on a high-K (10% KCl) diet for 7 days and decreased to 314 +/- 46 pA after 7 days on a low-K (0.1% KCl) diet. In the CNT, I(SK) was 360 +/- 30 pA on control, 1,160 +/- 110 on high-K, and 166 +/- 16 pA on low-K diets. The results indicate that ROMK channel activity is highly regulated by dietary K in both the CCD and the CNT.
Collapse
Affiliation(s)
- Gustavo Frindt
- Dept. Physiology and Biophysics, Weill Medical College of Cornell U., 1300 York Ave., New York, NY 10065, USA
| | | | | | | |
Collapse
|
42
|
Lachheb S, Cluzeaud F, Bens M, Genete M, Hibino H, Lourdel S, Kurachi Y, Vandewalle A, Teulon J, Paulais M. Kir4.1/Kir5.1 channel forms the major K+ channel in the basolateral membrane of mouse renal collecting duct principal cells. Am J Physiol Renal Physiol 2008; 294:F1398-407. [PMID: 18367659 DOI: 10.1152/ajprenal.00288.2007] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
K(+) channels in the basolateral membrane of mouse cortical collecting duct (CCD) principal cells were identified with patch-clamp technique, real-time PCR, and immunohistochemistry. In cell-attached membrane patches, three K(+) channels with conductances of approximately 75, 40, and 20 pS were observed, but the K(+) channel with the intermediate conductance (40 pS) predominated. In inside-out membrane patches exposed to an Mg(2+)-free medium, the current-voltage relationship of the intermediate-conductance channel was linear with a conductance of 38 pS. Addition of 1.3 mM internal Mg(2+) had no influence on the inward conductance (G(in) = 35 pS) but reduced outward conductance (G(out)) to 13 pS, yielding a G(in)/G(out) of 3.2. The polycation spermine (6 x 10(-7) M) reduced its activity on inside-out membrane patches by 50% at a clamp potential of 60 mV. Channel activity was also dependent on intracellular pH (pH(i)): a sigmoid relationship between pH(i) and channel normalized current (NP(o)) was observed with a pK of 7.24 and a Hill coefficient of 1.7. By real-time PCR on CCD extracts, inwardly rectifying K(+) (Kir)4.1 and Kir5.1, but not Kir4.2, mRNAs were detected. Kir4.1 and Kir5.1 proteins cellularly colocalized with aquaporin 2 (AQP2), a specific marker of CCD principal cells, while AQP2-negative cells (i.e., intercalated cells) showed no staining. Dietary K(+) had no influence on the properties of the intermediate-conductance channel, but a Na(+)-depleted diet increased its open probability by approximately 25%. We conclude that the Kir4.1/Kir5.1 channel is a major component of the K(+) conductance in the basolateral membrane of mouse CCD principal cells.
Collapse
MESH Headings
- Animals
- Cell Polarity/physiology
- Cloning, Molecular
- Immunohistochemistry
- In Vitro Techniques
- Kidney Cortex/physiology
- Kidney Tubules, Collecting/cytology
- Kidney Tubules, Collecting/physiology
- Male
- Mice
- Mice, Inbred Strains
- Models, Biological
- Patch-Clamp Techniques
- Potassium Channels, Inwardly Rectifying/genetics
- Potassium Channels, Inwardly Rectifying/physiology
- Potassium, Dietary/pharmacokinetics
- RNA, Messenger/metabolism
- Sodium, Dietary/pharmacokinetics
- Kir5.1 Channel
Collapse
Affiliation(s)
- Sahran Lachheb
- Université Pierre et Marie Curie, 75720 Paris Cedex 06, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Hughes BA, Swaminathan A. Modulation of the Kir7.1 potassium channel by extracellular and intracellular pH. Am J Physiol Cell Physiol 2007; 294:C423-31. [PMID: 18094146 DOI: 10.1152/ajpcell.00393.2007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Inwardly rectifying K(+) (K(ir)) channels in the apical membrane of the retinal pigment epithelium (RPE) contribute to extracellular K(+) homeostasis in the distal retina by mediating K(+) secretion. Multiple lines of evidence suggest that these channels are composed of Kir7.1. Previously, we showed that native K(ir) channels in bovine RPE are modulated by changes in intracellular pH in the physiological range. In the present study, we used the Xenopus laevis oocyte expression system to investigate the pH dependence of cloned human Kir7.1 channels and several point mutants involving histidine residues in the NH(2) and COOH termini. Kir7.1 channels were inhibited by strong extracellular acidification and modulated by intracellular pH in a biphasic manner, with maximal activity at about intracellular pH (pH(i)) 7.0 and inhibition by acidification or alkalinization. Replacement of histidine 26 (H26) in the NH(2) terminus with alanine eliminated the requirement of protons for channel activity and increased sensitivity to proton-induced inhibition, resulting in maximal channel activity at alkaline pH(i) and smaller whole cell currents at resting pH(i) compared with wild-type Kir7.1. When H26 was replaced with arginine, the pH(i) sensitivity profile was similar to that of the H26A mutant but with the pK(a) shifted to a more acidic value, giving rise to whole cell current amplitude at resting pH(i) that was comparable to that of wild-type Kir7.1. These results indicate that Kir7.1 channels are modulated by intracellular protons by diverse mechanisms and suggest that H26 is important for channel activation at physiological pH(i) and that it influences an unidentified proton-induced inhibitory mechanism.
Collapse
Affiliation(s)
- Bret A Hughes
- Department of Ophthalmology, University of Michigan, Ann Arbor, MI 48105, USA.
| | | |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW The connecting tubule is emerging as a nephron segment critical to the regulation of Na+ and K+ excretion and the maintenance of homeostasis for these ions. The segment is difficult to study, however, and much of the available information we have concerning its functions is indirect. Here, we review the major transport mechanisms and transporters found in this segment and outline several unsolved problems in the field. RECENT FINDINGS Recent electrophysiological and immunohistochemical measurements together with theoretical studies provide a more comprehensive view of ion transport in the connecting tubule. New signaling pathways governing Na+ and K+ transport have also been described. SUMMARY Key questions about how Na+ and K+ transport are regulated remain unanswered. Is the connecting tubule the site of final regulation of both Na+ and K+ excretion? If so, how are the transport rates of these two ions independently controlled?
Collapse
Affiliation(s)
- Lawrence G Palmer
- Department of Physiology and Biophysics, Weill Medical College of Cornell University, New York, New York 10021, USA.
| | | |
Collapse
|
45
|
Expression of Kir7.1 and a novel Kir7.1 splice variant in native human retinal pigment epithelium. Exp Eye Res 2007; 86:81-91. [PMID: 18035352 DOI: 10.1016/j.exer.2007.09.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2007] [Revised: 09/24/2007] [Accepted: 09/25/2007] [Indexed: 11/20/2022]
Abstract
Previous studies on bovine retinal pigment epithelium (RPE) established that Kir7.1 channels compose this epithelium's large apical membrane K+ conductance. The purpose of this study was to determine whether Kir7.1 and potential Kir7.1 splice variants are expressed in native adult human RPE and, if so, to determine their function and how they are generated. RT-PCR analysis indicated that human RPE expresses full-length Kir7.1 and a novel Kir7.1 splice variant, designated Kir7.1S. Analysis of the human Kir7.1 gene (KCNJ13) organization revealed that it contains three exons, two introns, and a novel alternative 5' splice site in exon 2. In human RPE, the alternative usage of two competing 5' splice sites in exon 2 gives rise to transcripts encoding full-length Kir7.1 and Kir7.1S, which is predicted to encode a truncated protein. Real-time PCR indicated that Kir7.1 transcript is nearly as abundant as GAPDH mRNA in human RPE whereas Kir7.1S transcript expression is 4-fold lower. Western blot analysis showed that the splice variant is translated in Xenopus oocytes injected with Kir7.1S cRNA and revealed the expression of full-length Kir7.1 but not Kir7.1S in adult human RPE. Co-expression of Kir7.1 with Kir7.1S in Xenopus oocytes had no effect on either the kinetics or amplitude of Kir7.1 currents. This study confirms the expression of Kir7.1 in human RPE, identifies a Kir7.1 splice variant resulting in predicted changes in protein sequence, and indicates that there is no functional interaction between this splice variant and full-length Kir7.1.
Collapse
|
46
|
Tricarico D, Mele A, Liss B, Ashcroft FM, Lundquist AL, Desai RR, George AL, Conte Camerino D. Reduced expression of Kir6.2/SUR2A subunits explains KATP deficiency in K+-depleted rats. Neuromuscul Disord 2007; 18:74-80. [PMID: 17825556 DOI: 10.1016/j.nmd.2007.07.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2007] [Revised: 05/17/2007] [Accepted: 07/25/2007] [Indexed: 11/24/2022]
Abstract
We investigated on the mechanism responsible for the reduced ATP-sensitive K(+)(K(ATP)) channel activity recorded from skeletal muscle of K(+)-depleted rats. Patch-clamp and gene expression measurements of K(ATP) channel subunits were performed. A down-regulation of the K(ATP) channel subunits Kir6.2(-70%) and SUR2A(-46%) in skeletal muscles of K(+)-depleted rats but no changes in the expression of Kir6.1, SUR1 and SUR2B subunits were observed. A reduced K(ATP) channel currents of -69.5% in K(+)-depleted rats was observed. The Kir6.2/SUR2A-B agonist cromakalim showed similar potency in activating the K(ATP) channels of normokalaemic and K(+)-depleted rats but reduced efficacy in K(+)-depleted rats. The Kir6.2/SUR1-2B agonist diazoxide activated K(ATP) channels in normokalaemic and K(+)-depleted rats with equal potency and efficacy. The down-regulation of the Kir6.2 explains the reduced K(ATP) channel activity in K(+)-depleted rats. The lower expression of SUR2A explains the reduced efficacy of cromakalim; preserved SUR1 expression accounts for the efficacy of diazoxide. Kir6.2/SUR2A deficiency is associated with impaired muscle function in K(+)-depleted rats and in hypoPP.
Collapse
Affiliation(s)
- Domenico Tricarico
- Department of Pharmacobiology, Faculty of Pharmacy, University of Bari, via Orabona no 4, 70120 Bari, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Huang C, Miller RT. The calcium-sensing receptor and its interacting proteins. J Cell Mol Med 2007; 11:923-34. [PMID: 17979874 PMCID: PMC4401264 DOI: 10.1111/j.1582-4934.2007.00114.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2007] [Accepted: 08/21/2007] [Indexed: 01/27/2023] Open
Abstract
Seven membrane-spanning, or G protein-coupled receptors were originally thought to act through het-erotrimeric G proteins that in turn activate intracellular enzymes or ion channels, creating relatively simple, linear signalling pathways. Although this basic model remains true in that this family does act via a relatively small number of G proteins, these signalling systems are considerably more complex because the receptors interact with or are located near additional proteins that are often unique to a receptor or subset of receptors. These additional proteins give receptors their unique signalling personalities. The extracellular Ca-sensing receptor (CaR) signals via Galpha(i), Galpha(q) and Galpha(12/13), but its effects in vivo demonstrate that the signalling pathways controlled by these subunits are not sufficient to explain all its biologic effects. Additional structural or signalling proteins that interact with the CaR may explain its behaviour more fully. Although the CaR is less well studied in this respect than other receptors, several CaR-interacting proteins such as filamin, a potential scaffolding protein, receptor activity modifying proteins (RAMPs) and potassium channels may contribute to the unique characteristics of the CaR. The CaR also appears to interact with additional proteins common to other G protein-coupled receptors such as arrestins, G protein receptor kinases, protein kinase C, caveolin and proteins in the ubiquitination pathway. These proteins probably represent a few initial members of CaR-based signalling complex. These and other proteins may not all be associated with the CaR in all tissues, but they form the basis for understanding the complete nature of CaR signalling.
Collapse
Affiliation(s)
- Chunfa Huang
- Departments of Medicine and Physiology, Case-Western Reserve University, Louis Stokes VAMC Rammelkamp Center for Research, Metro Health Medical Center, Cleveland, Ohio, USA
| | - R Tyler Miller
- Departments of Medicine and Physiology, Case-Western Reserve University, Louis Stokes VAMC Rammelkamp Center for Research, Metro Health Medical Center, Cleveland, Ohio, USA
| |
Collapse
|
48
|
Huang C, Sindic A, Hill CE, Hujer KM, Chan KW, Sassen M, Wu Z, Kurachi Y, Nielsen S, Romero MF, Miller RT. Interaction of the Ca2+-sensing receptor with the inwardly rectifying potassium channels Kir4.1 and Kir4.2 results in inhibition of channel function. Am J Physiol Renal Physiol 2007; 292:F1073-81. [PMID: 17122384 DOI: 10.1152/ajprenal.00269.2006] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The Ca(2+)-sensing receptor (CaR), a G protein-coupled receptor, is expressed in many epithelial tissues including the parathyroid glands, kidney, and GI tract. Although its role in regulating PTH levels and Ca(2+) metabolism are best characterized, it may also regulate salt and water transport in the kidney as demonstrated by recent reports showing association of potent gain-of-function mutations in the CaR with a Bartter-like, salt-wasting phenotype. To determine whether this receptor interacts with novel proteins that control ion transport, we screened a human adult kidney cDNA library with the COOH-terminal 219 amino acid cytoplasmic tail of the CaR as bait using the yeast two-hybrid system. We identified two independent clones coding for approximately 125 aa from the COOH terminus of the inwardly rectifying K(+) channel, Kir4.2. The CaR and Kir4.2 as well as Kir4.1 (another member of Kir4 subfamily) were reciprocally coimmunoprecipitated from HEK-293 cells in which they were expressed, but the receptor did not coimmunoprecipitate with Kir5.1 or Kir1.1. Both Kir4.1 and Kir4.2 were immunoprecipitated from rat kidney extracts with the CaR. In Xenopus laevis oocytes, expression of the CaR with either Kir4.1 or Kir4.2 channels resulted in inactivation of whole cell current as measured by two-electrode voltage clamp, but the nonfunctional CaR mutant CaR(R796W), and that does not coimmunoprecipitate with the channels, had no effect. Kir4.1 and the CaR were colocalized in the basolateral membrane of the distal nephron. The CaR interacts directly with Kir4.1 and Kir4.2 and can decrease their currents, which in turn could reduce recycling of K(+) for the basolateral Na(+)-K(+)-ATPase and thereby contribute to inhibition of Na(+) reabsorption.
Collapse
Affiliation(s)
- Chunfa Huang
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Millar ID, Taylor HC, Cooper GJ, Kibble JD, Robson L. A Kir2.3-like K+ Conductance in Mouse Cortical Collecting Duct Principal Cells. J Membr Biol 2006; 211:173-84. [PMID: 17091215 DOI: 10.1007/s00232-006-0036-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2006] [Revised: 07/11/2006] [Indexed: 10/23/2022]
Abstract
K(+) channels play an important role in renal collecting duct cell function. The current study examined barium (Ba(2+))-sensitive whole-cell K(+) currents (IKBa) in mouse isolated collecting duct principal cells. IKBa demonstrated strong inward rectification and was inhibited by Ba(2+) in a dose- and voltage-dependent fashion, with the K (d) decreasing with hyperpolarization. The electrical distance of block by Ba(2+) was around 8.5%. As expected for voltage-dependent inhibition, the association constant increased with hyperpolarization, suggesting that the rate of Ba(2+) entry was increased at negative potentials. The dissociation constant also increased with hyperpolarization, consistent with the movement of Ba(2+) ions into the intracellular compartment at negative potentials. These properties are not consistent with ROMK but are consistent with the properties of Kir2.3. Kir2.3 is thought to be the dominant basolateral K(+) channel in principal cells. This study provides functional evidence for the expression of Kir2.3 in mouse cortical collecting ducts and confirms the expression of Kir2.3 in this segment of the renal tubule using reverse-transcriptase polymerase chain reaction. The conductance described here is the first report of a macroscopic K(+) conductance in mouse principal cells that shares the biophysical profile of Kir2.3. The properties and dominant nature of the conductance suggest that it plays an important role in K(+) handling in the principal cells of the cortical collecting duct.
Collapse
Affiliation(s)
- I D Millar
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | | | | | | | | |
Collapse
|
50
|
Yabuki A, Taharaguchi S, Ichii O, Kojima M, Nishi Y, Mifune H, Kamimura R, Matsumoto M, Suzuki S. Immunohistochemical localization of ghrelin in rodent kidneys. Histochem Cell Biol 2006; 126:231-8. [PMID: 16514547 DOI: 10.1007/s00418-006-0165-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2006] [Indexed: 12/27/2022]
Abstract
Ghrelin is a novel peptide hormone, originally identified in the rat and human stomach that plays various important roles. In the present study, we report the intra-renal localization of ghrelin in laboratory rodents. Kidneys from 3 month-old mice, rats and hamsters of both sexes were analyzed by immunohistochemistry. Positive signals were clearly observed in the epithelium of the distal tubules, whereas other segments of the nephron or interstitial cells, including juxtaglomerular cells, showed negative reactions. Pre-embedding immunoelectron microscopy revealed positive signals exclusively on the basolateral membrane in the distal tubular cells and in the collecting ducts. In addition, prepro-ghrelin gene expression was assessed by RT-PCR, and the expected 329-bp prepro-ghrelin mRNA was clearly detected in the kidney. On Western blot analysis, although a specific band for ghrelin (3 kDa) was not detected in the kidney, the expected band for prepro-ghrelin (13 kDa) was clearly detected in both the stomach and the kidney. This paper clarified the intra-renal localization of ghrelin.
Collapse
Affiliation(s)
- Akira Yabuki
- Laboratory of Veterinary Anatomy, Department of Veterinary Medicine, Faculty of Agriculture, Kagoshima University, 1-21-24 Korimoto, 890-0065 Kagoshima, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|