54 results on '"Kashlan OB"'
Search Results
2. Loss of the alpha subunit distal furin cleavage site blunts ENaC activation following Na + restriction.
- Author
-
Nickerson AJ, Sheng S, Cox NA, Szekely KG, Marciszyn AL, Lam T, Chen J, Gingras S, Kashlan OB, Kirabo A, Hughey RP, Ray EC, and Kleyman TR
- Subjects
- Animals, Mice, Male, Sodium metabolism, Colon metabolism, Mice, Inbred C57BL, Aldosterone metabolism, Diet, Sodium-Restricted, Epithelial Sodium Channels metabolism, Epithelial Sodium Channels genetics, Furin metabolism, Furin genetics
- Abstract
Epithelial Na
+ channels (ENaCs) are activated by proteolysis of the α and γ subunits at specific sites flanking embedded inhibitory tracts. To examine the role of α subunit proteolysis in channel activation in vivo, we generated mice lacking the distal furin cleavage site in the α subunit (αF2M mice). On a normal Na+ control diet, no differences in ENaC protein abundance in kidney or distal colon were noted between wild-type (WT) and αF2M mice. Patch-clamp analyses revealed similar levels of ENaC activity in kidney tubules, while no physiologically relevant differences in blood chemistry or aldosterone levels were detected. Male αF2M mice did exhibit diminished ENaC activity in the distal colon, as measured by amiloride-sensitive short-circuit current (ISC ). Following dietary Na+ restriction, WT and αF2M mice had similar natriuretic and colonic ISC responses to amiloride. However, single-channel activity was significantly lower in kidney tubules from Na+ -restricted αF2M mice compared with WT littermates. ENaC α and γ subunit expression in kidney and distal colon were also enhanced in Na+ -restricted αF2M vs. WT mice, in association with higher aldosterone levels. These data provide evidence that disrupting α subunit proteolysis impairs ENaC activity in vivo, requiring compensation in response to Na+ restriction. KEY POINTS: The epithelial Na+ channel (ENaC) is activated by proteolytic cleavage in vitro, but key questions regarding the role of ENaC proteolysis in terms of whole-animal physiology remain to be addressed. We studied the in vivo importance of this mechanism by generating a mouse model with a genetic disruption to a key cleavage site in the ENaC's α subunit (αF2M mice). We found that αF2M mice did not exhibit a physiologically relevant phenotype under normal dietary conditions, but have impaired ENaC activation (channel open probability) in the kidney during salt restriction. ENaC function at the organ level was preserved in salt-restricted αF2M mice, but this was associated with higher aldosterone levels and increased expression of ENaC subunits, suggesting compensation was required to maintain homeostasis. These results provide the first evidence that ENaC α subunit proteolysis is a key regulator of channel activity in vivo., (© 2024 The Author(s). The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society.)- Published
- 2024
- Full Text
- View/download PDF
3. Varying Selection Pressure for a Na+ Sensing Site in Epithelial Na+ Channel Subunits Reflect Divergent Roles in Na+ Homeostasis.
- Author
-
Wang XP, Srinivasan P, El Hamdaoui M, Blobner BM, Grytz R, and Kashlan OB
- Subjects
- Animals, Humans, Binding Sites, Vertebrates genetics, Protein Subunits metabolism, Protein Subunits genetics, Phylogeny, Epithelial Sodium Channels genetics, Epithelial Sodium Channels metabolism, Homeostasis, Sodium metabolism, Selection, Genetic, Evolution, Molecular
- Abstract
The epithelial Na+ channel (ENaC) emerged early in vertebrates and has played a role in Na+ and fluid homeostasis throughout vertebrate evolution. We previously showed that proteolytic activation of the channel evolved at the water-to-land transition of vertebrates. Sensitivity to extracellular Na+, known as Na+ self-inhibition, reduces ENaC function when Na+ concentrations are high and is a distinctive feature of the channel. A fourth ENaC subunit, δ, emerged in jawed fishes from an α subunit gene duplication. Here, we analyzed 849 α and δ subunit sequences and found that a key Asp in a postulated Na+ binding site was nearly always present in the α subunit, but frequently lost in the δ subunit (e.g. human). Analysis of site evolution and codon substitution rates provide evidence that the ancestral α subunit had the site and that purifying selection for the site relaxed in the δ subunit after its divergence from the α subunit, coinciding with a loss of δ subunit expression in renal tissues. We also show that the proposed Na+ binding site in the α subunit is a bona fide site by conferring novel function to channels comprising human δ subunits. Together, our findings provide evidence that ENaC Na+ self-inhibition improves fitness through its role in Na+ homeostasis in vertebrates., (© The Author(s) 2024. Published by Oxford University Press on behalf of Society for Molecular Biology and Evolution.)
- Published
- 2024
- Full Text
- View/download PDF
4. SGLT2-independent effects of canagliflozin on NHE3 and mitochondrial complex I activity inhibit proximal tubule fluid transport and albumin uptake.
- Author
-
Albalawy WN, Youm EB, Shipman KE, Trull KJ, Baty CJ, Long KR, Rbaibi Y, Wang XP, Fagunloye OG, White KA, Jurczak MJ, Kashlan OB, and Weisz OA
- Subjects
- Animals, Mice, Male, Sodium-Glucose Transporter 2 metabolism, Endocytosis drug effects, Mice, Inbred C57BL, Albumins metabolism, Mitochondria metabolism, Mitochondria drug effects, Benzhydryl Compounds, Glucosides, Kidney Tubules, Proximal drug effects, Kidney Tubules, Proximal metabolism, Kidney Tubules, Proximal enzymology, Sodium-Hydrogen Exchanger 3 metabolism, Canagliflozin pharmacology, Sodium-Glucose Transporter 2 Inhibitors pharmacology
- Abstract
Beyond glycemic control, SGLT2 inhibitors (SGLT2is) have protective effects on cardiorenal function. Renoprotection has been suggested to involve inhibition of NHE3 leading to reduced ATP-dependent tubular workload and mitochondrial oxygen consumption. NHE3 activity is also important for regulation of endosomal pH, but the effects of SGLT2i on endocytosis are unknown. We used a highly differentiated cell culture model of proximal tubule (PT) cells to determine the direct effects of SGLT2i on Na
+ -dependent fluid transport and endocytic uptake in this nephron segment. Strikingly, canagliflozin but not empagliflozin reduced fluid transport across cell monolayers and dramatically inhibited endocytic uptake of albumin. These effects were independent of glucose and occurred at clinically relevant concentrations of drug. Canagliflozin acutely inhibited surface NHE3 activity, consistent with a direct effect, but did not affect endosomal pH or NHE3 phosphorylation. In addition, canagliflozin rapidly and selectively inhibited mitochondrial complex I activity. Inhibition of mitochondrial complex I by metformin recapitulated the effects of canagliflozin on endocytosis and fluid transport, whereas modulation of downstream effectors AMPK and mTOR did not. Mice given a single dose of canagliflozin excreted twice as much urine over 24 h compared with empagliflozin-treated mice despite similar water intake. We conclude that canagliflozin selectively suppresses Na+ -dependent fluid transport and albumin uptake in PT cells via direct inhibition of NHE3 and of mitochondrial function upstream of the AMPK/mTOR axis. These additional targets of canagliflozin contribute significantly to reduced PT Na+ -dependent fluid transport in vivo. NEW & NOTEWORTHY Reduced NHE3-mediated Na+ transport has been suggested to underlie the cardiorenal protection provided by SGLT2 inhibitors. We found that canagliflozin, but not empagliflozin, reduced NHE3-dependent fluid transport and endocytic uptake in cultured proximal tubule cells. These effects were independent of SGLT2 activity and resulted from inhibition of mitochondrial complex I and NHE3. Studies in mice are consistent with greater effects of canagliflozin versus empagliflozin on fluid transport. Our data suggest that these selective effects of canagliflozin contribute to reduced Na+ -dependent transport in proximal tubule cells.- Published
- 2024
- Full Text
- View/download PDF
5. Epithelial Na + Channels Function as Extracellular Sensors.
- Author
-
Kashlan OB, Wang XP, Sheng S, and Kleyman TR
- Subjects
- Humans, Animals, Epithelial Sodium Channels metabolism, Epithelial Sodium Channels physiology
- Abstract
The epithelial Na
+ channel (ENaC) resides on the apical surfaces of specific epithelia in vertebrates and plays a critical role in extracellular fluid homeostasis. Evidence that ENaC senses the external environment emerged well before the molecular identity of the channel was reported three decades ago. This article discusses progress toward elucidating the mechanisms through which specific external factors regulate ENaC function, highlighting insights gained from structural studies of ENaC and related family members. It also reviews our understanding of the role of ENaC regulation by the extracellular environment in physiology and disease. After familiarizing the reader with the channel's physiological roles and structure, we describe the central role protein allostery plays in ENaC's sensitivity to the external environment. We then discuss each of the extracellular factors that directly regulate the channel: proteases, cations and anions, shear stress, and other regulators specific to particular extracellular compartments. For each regulator, we discuss the initial observations that led to discovery, studies investigating molecular mechanism, and the physiological and pathophysiological implications of regulation. © 2024 American Physiological Society. Compr Physiol 14:5407-5447, 2024., (Copyright © 2024 American Physiological Society. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
6. MicroRNA-19 is regulated by aldosterone in a sex-specific manner to alter kidney sodium transport.
- Author
-
Farrell CE, Liu X, Yagan NO, Suda AC, Cerqueira DM, Bodnar AJ, Kashlan OB, Subramanya AR, Ho J, and Butterworth MB
- Subjects
- Female, Mice, Animals, Aldosterone metabolism, Protein Serine-Threonine Kinases metabolism, Glucocorticoids, Phylogeny, Kidney metabolism, Sodium metabolism, Epithelial Sodium Channels metabolism, MicroRNAs genetics, MicroRNAs metabolism
- Abstract
A key regulator of blood pressure homeostasis is the steroid hormone aldosterone, which is released as the final signaling hormone of the renin-angiotensin-aldosterone-signaling (RAAS) system. Aldosterone increases sodium (Na
+ ) reabsorption in the kidney distal nephron to regulate blood volume. Unregulated RAAS signaling can lead to hypertension and cardiovascular disease. The serum and glucocorticoid kinase (SGK1) coordinates much of the Na+ reabsorption in the cortical collecting duct (CCD) tubular epithelial cells. We previously demonstrated that aldosterone alters the expression of microRNAs (miRs) in CCD principal cells. The aldosterone-regulated miRs can modulate Na+ transport and the cellular response to aldosterone signaling. However, the sex-specific regulation of miRs by aldosterone in the kidney distal nephron has not been explored. In this study, we report that miR-19, part of the miR-17-92 cluster, is upregulated in female mouse CCD cells in response to aldosterone activation. Mir-19 binding to the 3'-untranslated region of SGK1 was confirmed using a dual-luciferase reporter assay. Increasing miR-19 expression in CCD cells decreased SGK1 message and protein expression. Removal of this cluster using a nephron-specific, inducible knockout mouse model increased SGK1 expression in female mouse CCD cells. The miR-19-induced decrease in SGK1 protein expression reduced the response to aldosterone stimulation and may account for sex-specific differences in aldosterone signaling. By examining evolution of the miR-17-92 cluster, phylogenetic sequence analysis indicated that this cluster arose at the same time that other Na+ -sparing and salt regulatory proteins, specifically SGK1, first emerged, indicating a conserved role for these miRs in kidney function of salt and water homeostasis. NEW & NOTEWORTHY Expression of the microRNA-17-92 cluster is upregulated by aldosterone in mouse cortical collecting duct principal cells, exclusively in female mice. MiR-19 in this cluster targets the serum and glucocorticoid kinase (SGK1) to downregulate both mRNA and protein expression, resulting in a decrease in sodium transport across epithelial cells of the collecting duct. The miR-17-92 cluster is evolutionarily conserved and may act as a novel feedback regulator for aldosterone signaling in females.- Published
- 2024
- Full Text
- View/download PDF
7. Mice lacking γENaC palmitoylation sites maintain benzamil-sensitive Na+ transport despite reduced channel activity.
- Author
-
Nickerson AJ, Mutchler SM, Sheng S, Cox NA, Ray EC, Kashlan OB, Carattino MD, Marciszyn AL, Winfrey A, Gingras S, Kirabo A, Hughey RP, and Kleyman TR
- Subjects
- Mice, Male, Animals, Epithelial Sodium Channels genetics, Epithelial Sodium Channels metabolism, Sodium metabolism, Amiloride pharmacology, Lipoylation
- Abstract
Epithelial Na+ channels (ENaCs) control extracellular fluid volume by facilitating Na+ absorption across transporting epithelia. In vitro studies showed that Cys-palmitoylation of the γENaC subunit is a major regulator of channel activity. We tested whether γ subunit palmitoylation sites are necessary for channel function in vivo by generating mice lacking the palmitoylated cysteines (γC33A,C41A) using CRISPR/Cas9 technology. ENaCs in dissected kidney tubules from γC33A,C41A mice had reduced open probability compared with wild-type (WT) littermates maintained on either standard or Na+-deficient diets. Male mutant mice also had higher aldosterone levels than WT littermates following Na+ restriction. However, γC33A,C41A mice did not have reduced amiloride-sensitive Na+ currents in the distal colon or benzamil-induced natriuresis compared to WT mice. We identified a second, larger conductance cation channel in the distal nephron with biophysical properties distinct from ENaC. The activity of this channel was higher in Na+-restricted γC33A,C41A versus WT mice and was blocked by benzamil, providing a possible compensatory mechanism for reduced prototypic ENaC function. We conclude that γ subunit palmitoylation sites are required for prototypic ENaC activity in vivo but are not necessary for amiloride/benzamil-sensitive Na+ transport in the distal nephron or colon.
- Published
- 2023
- Full Text
- View/download PDF
8. Receptor-associated protein impairs ligand binding to megalin and megalin-dependent endocytic flux in proximal tubule cells.
- Author
-
Long KR, Rbaibi Y, Kashlan OB, and Weisz OA
- Subjects
- Ligands, Cell Membrane metabolism, Endocytosis physiology, Kidney Tubules, Proximal metabolism, Low Density Lipoprotein Receptor-Related Protein-2 genetics, Low Density Lipoprotein Receptor-Related Protein-2 metabolism, Albumins metabolism
- Abstract
Proximal tubule (PT) cells retrieve albumin and a broad array of other ligands from the glomerular ultrafiltrate. Efficient uptake of albumin requires PT expression of both megalin and cubilin receptors. Although most proteins engage cubilin selectively, megalin is required to maintain robust flux through the apical endocytic pathway. Receptor-associated protein (RAP) is a chaperone that directs megalin to the cell surface, and recombinant RAP dramatically inhibits the uptake of numerous megalin and cubilin ligands. The mechanism by which this occurs has been suggested to involve competitive inhibition of ligand binding and/or conformational changes in megalin that prevent interaction with ligands and/or with cubilin. To discriminate between these possibilities, we determined the effect of RAP on endocytosis of albumin, which binds to cubilin and megalin receptors with high and low affinity, respectively. Uptake was quantified in opossum kidney (OK) cells and in megalin or cubilin ( Cubn ) knockout (KO) clones. Surprisingly, RAP inhibited fluid-phase uptake in addition to receptor-mediated uptake in OK cells and Cubn KO cells but had no effect on endocytosis when megalin was absent. The apparent K
i for RAP inhibition of albumin uptake was 10-fold higher in Cubn KO cells compared with parental OK cells. We conclude that in addition to its predicted high-affinity competition for ligand binding to megalin, the primary effect of RAP on PT cell endocytosis is to globally dampen megalin-dependent endocytic flux. Our data explain the complex effects of RAP on binding and uptake of filtered proteins and reveal a novel role in modulating endocytosis in PT cells. NEW & NOTEWORTHY Receptor-associated protein inhibits binding and uptake of all known endogenous ligands by megalin and cubilin receptors via unknown mechanism(s). Here, we took advantage of recently generated knockout cell lines to dissect the effect of this protein on megalin- and cubilin-mediated endocytosis. Our study reveals a novel role for receptor-associated protein in blocking megalin-stimulated endocytic uptake of fluid-phase markers and receptor-bound ligands in proximal tubule cells in addition to its direct effect on ligand binding to megalin receptors.- Published
- 2023
- Full Text
- View/download PDF
9. Lhs1 dependent ERAD is determined by transmembrane domain context.
- Author
-
Sukhoplyasova M, Keith AM, Perrault EM, Vorndran HE, Jordahl AS, Yates ME, Pastor A, Li Z, Freaney ML, Deshpande RA, Adams DB, Guerriero CJ, Shi S, Kleyman TR, Kashlan OB, Brodsky JL, and Buck TM
- Subjects
- Animals, Cytosol, Lipid Bilayers, Membrane Proteins genetics, Mammals, Endoplasmic Reticulum-Associated Degradation, Endoplasmic Reticulum
- Abstract
Transmembrane proteins have unique requirements to fold and integrate into the endoplasmic reticulum (ER) membrane. Most notably, transmembrane proteins must fold in three separate environments: extracellular domains fold in the oxidizing environment of the ER lumen, transmembrane domains (TMDs) fold within the lipid bilayer, and cytosolic domains fold in the reducing environment of the cytosol. Moreover, each region is acted upon by a unique set of chaperones and monitored by components of the ER associated quality control machinery that identify misfolded domains in each compartment. One factor is the ER lumenal Hsp70-like chaperone, Lhs1. Our previous work established that Lhs1 is required for the degradation of the unassembled α-subunit of the epithelial sodium channel (αENaC), but not the homologous β- and γENaC subunits. However, assembly of the ENaC heterotrimer blocked the Lhs1-dependent ER associated degradation (ERAD) of the α-subunit, yet the characteristics that dictate the specificity of Lhs1-dependent ERAD substrates remained unclear. We now report that Lhs1-dependent substrates share a unique set of features. First, all Lhs1 substrates appear to be unglycosylated, and second they contain two TMDs. Each substrate also contains orphaned or unassembled TMDs. Additionally, interfering with inter-subunit assembly of the ENaC trimer results in Lhs1-dependent degradation of the entire complex. Finally, our work suggests that Lhs1 is required for a subset of ERAD substrates that also require the Hrd1 ubiquitin ligase. Together, these data provide hints as to the identities of as-yet unconfirmed substrates of Lhs1 and potentially of the Lhs1 homolog in mammals, GRP170., (© 2023 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society.)
- Published
- 2023
- Full Text
- View/download PDF
10. Mineralocorticoid receptor-independent activation of ENaC in bile duct ligated mice.
- Author
-
Wang XP, Mutchler SM, Carrisoza-Gáytan R, Al-Bataineh M, Baty CJ, Vandevender A, Srinivasan P, Tan RJ, Jurczak MJ, Satlin LM, and Kashlan OB
- Abstract
Sodium and fluid retention in liver disease is classically thought to result from reduced effective circulating volume and stimulation of the renin-angiotensin-aldosterone system (RAAS). Aldosterone dives Na
+ retention by activating the mineralocorticoid receptor and promoting the maturation and apical surface expression of the epithelial Na+ channel (ENaC), found in the aldosterone-sensitive distal nephron. However, evidence of fluid retention without RAAS activation suggests the involvement of additional mechanisms. Liver disease can greatly increase plasma and urinary bile acid concentrations and have been shown to activate ENaC in vitro . We hypothesize that elevated bile acids in liver disease activate ENaC and drive fluid retention independent of RAAS. We therefore increased circulating bile acids in mice through bile duct ligation (BDL) and measured effects on urine and body composition, while using spironolactone to antagonize the mineralocorticoid receptor. We found BDL lowered blood [K+ ] and hematocrit, and increased benzamil-sensitive natriuresis compared to sham, consistent with ENaC activation. BDL mice also gained significantly more body water. Blocking ENaC reversed fluid gains in BDL mice but had no effect in shams. In isolated collecting ducts from rabbits, taurocholic acid stimulated net Na+ absorption but had no effect on K+ secretion or flow-dependent ion fluxes. Our results provide experimental evidence for a novel aldosterone-independent mechanism for sodium and fluid retention in liver disease which may provide additional therapeutic options for liver disease patients., Competing Interests: Conflict of Interest: Authors declare no conflict of interest.- Published
- 2023
- Full Text
- View/download PDF
11. Megalin, cubilin, and Dab2 drive endocytic flux in kidney proximal tubule cells.
- Author
-
Rbaibi Y, Long KR, Shipman KE, Ren Q, Baty CJ, Kashlan OB, and Weisz OA
- Subjects
- Animals, Mice, Adaptor Proteins, Signal Transducing metabolism, Albumins metabolism, Apoptosis Regulatory Proteins metabolism, Endocytosis physiology, Kidney Tubules, Proximal metabolism, Mice, Knockout, Low Density Lipoprotein Receptor-Related Protein-2, Receptors, Cell Surface metabolism
- Abstract
The kidney proximal tubule (PT) elaborates a uniquely high-capacity apical endocytic pathway to retrieve albumin and other proteins that escape the glomerular filtration barrier. Megalin and cubilin/amnionless (CUBAM) receptors engage Dab2 in these cells to mediate clathrin-dependent uptake of filtered ligands. Knockout of megalin or Dab2 profoundly inhibits apical endocytosis and is believed to atrophy the endocytic pathway. We generated CRISPR/Cas9 knockout (KO) clones lacking cubilin, megalin, or Dab2 expression in highly differentiated PT cells and determined the impact on albumin internalization and endocytic pathway function. KO of each component had different effects on the concentration dependence of albumin uptake as well its distribution within PT cells. Reduced uptake of a fluid phase marker was also observed, with megalin KO cells having the most dramatic decline. Surprisingly, protein levels and distribution of key endocytic proteins were preserved in KO PT cell lines and in megalin KO mice, despite the reduced endocytic activity. Our data highlight specific functions of megalin, cubilin, and Dab2 in apical endocytosis and demonstrate that these proteins drive endocytic flux without compromising the physical integrity of the apical endocytic pathway. Our studies suggest a novel model to explain how these components coordinate endocytic uptake in PT cells.
- Published
- 2023
- Full Text
- View/download PDF
12. Impaired Endosome Maturation Mediates Tubular Proteinuria in Dent Disease Cell Culture and Mouse Models.
- Author
-
Shipman KE, Baty CJ, Long KR, Rbaibi Y, Cowan IA, Gerges M, Marciszyn AL, Kashlan OB, Tan RJ, Edwards A, and Weisz OA
- Subjects
- Mice, Animals, Endocytosis, Proteinuria pathology, Endosomes metabolism, Kidney Tubules, Proximal metabolism, Disease Models, Animal, Mice, Knockout, Cell Culture Techniques, Antiporters, Low Density Lipoprotein Receptor-Related Protein-2 genetics, Low Density Lipoprotein Receptor-Related Protein-2 metabolism, Dent Disease genetics, Dent Disease metabolism
- Abstract
Significance Statement: Loss of function of the 2Cl - /H + antiporter ClC-5 in Dent disease causes an unknown impairment in endocytic traffic, leading to tubular proteinuria. The authors integrated data from biochemical and quantitative imaging studies in proximal tubule cells into a mathematical model to determine that loss of ClC-5 impairs endosome acidification and delays early endosome maturation in proximal tubule cells, resulting in reduced megalin recycling, surface expression, and half-life. Studies in a Dent mouse model also revealed subsegment-specific differences in the effects of ClC-5 knockout on proximal tubule subsegments. The approach provides a template to dissect the effects of mutations or perturbations that alter tubular recovery of filtered proteins from the level of individual cells to the entire proximal tubule axis., Background: Loss of function of the 2Cl - /H + antiporter ClC-5 in Dent disease impairs the uptake of filtered proteins by the kidney proximal tubule, resulting in tubular proteinuria. Reduced posttranslational stability of megalin and cubilin, the receptors that bind to and recover filtered proteins, is believed to underlie the tubular defect. How loss of ClC-5 leads to reduced receptor expression remains unknown., Methods: We used biochemical and quantitative imaging data to adapt a mathematical model of megalin traffic in ClC-5 knockout and control cells. Studies in ClC-5 knockout mice were performed to describe the effect of ClC-5 knockout on megalin traffic in the S1 segment and along the proximal tubule axis., Results: The model predicts that ClC-5 knockout cells have reduced rates of exit from early endosomes, resulting in decreased megalin recycling, surface expression, and half-life. Early endosomes had lower [Cl - ] and higher pH. We observed more profound effects in ClC-5 knockout cells expressing the pathogenic ClC-5 E211G mutant. Alterations in the cellular distribution of megalin in ClC-5 knockout mice were consistent with delayed endosome maturation and reduced recycling. Greater reductions in megalin expression were observed in the proximal tubule S2 cells compared with S1, with consequences to the profile of protein retrieval along the proximal tubule axis., Conclusions: Delayed early endosome maturation due to impaired acidification and reduced [Cl - ] accumulation is the primary mediator of reduced proximal tubule receptor expression and tubular proteinuria in Dent disease. Rapid endosome maturation in proximal tubule cells is critical for the efficient recovery of filtered proteins., (Copyright © 2023 by the American Society of Nephrology.)
- Published
- 2023
- Full Text
- View/download PDF
13. Rare Variants in Genes Encoding Subunits of the Epithelial Na + Channel Are Associated With Blood Pressure and Kidney Function.
- Author
-
Blobner BM, Kirabo A, Kashlan OB, Sheng S, Arnett DK, Becker LC, Boerwinkle E, Carlson JC, Gao Y, Gibbs RA, He J, Irvin MR, Kardia SLR, Kelly TN, Kooperberg C, McGarvey ST, Menon VK, Montasser ME, Naseri T, Redline S, Reiner AP, Reupena MS, Smith JA, Sun X, Vaidya D, Viaud-Martinez KA, Weeks DE, Yanek LR, Zhu X, Minster RL, and Kleyman TR
- Subjects
- Humans, Blood Pressure genetics, Phenotype, Kidney, Epithelial Sodium Channels genetics, Sodium
- Abstract
Background: The epithelial Na
+ channel (ENaC) is intrinsically linked to fluid volume homeostasis and blood pressure. Specific rare mutations in SCNN1A , SCNN1B , and SCNN1G , genes encoding the α, β, and γ subunits of ENaC, respectively, are associated with extreme blood pressure phenotypes. No associations between blood pressure and SCNN1D , which encodes the δ subunit of ENaC, have been reported. A small number of sequence variants in ENaC subunits have been reported to affect functional transport in vitro or blood pressure. The effects of the vast majority of rare and low-frequency ENaC variants on blood pressure are not known., Methods: We explored the association of low frequency and rare variants in the genes encoding ENaC subunits, with systolic blood pressure, diastolic blood pressure, mean arterial pressure, and pulse pressure. Using whole-genome sequencing data from 14 studies participating in the Trans-Omics in Precision Medicine Whole-Genome Sequencing Program, and sequence kernel association tests., Results: We found that variants in SCNN1A and SCNN1B were associated with diastolic blood pressure and mean arterial pressure ( P <0.00625). Although SCNN1D is poorly expressed in human kidney tissue, SCNN1D variants were associated with systolic blood pressure, diastolic blood pressure, mean arterial pressure, and pulse pressure ( P <0.00625). ENaC variants in 2 of the 4 subunits ( SCNN1B and SCNN1D ) were also associated with estimated glomerular filtration rate ( P <0.00625), but not with stroke., Conclusions: Our results suggest that variants in extrarenal ENaCs, in addition to ENaCs expressed in kidneys, influence blood pressure and kidney function.- Published
- 2022
- Full Text
- View/download PDF
14. Bile acids regulate the epithelial Na + channel in native tissues through direct binding at multiple sites.
- Author
-
Wang XP, Tomilin V, Nickerson AJ, Tian R, Ertem M, McKernan A, Lei X, Pochynyuk O, and Kashlan OB
- Subjects
- Animals, Mice, Amiloride, Ions metabolism, Oocytes physiology, Sodium metabolism, Taurocholic Acid metabolism, Xenopus laevis metabolism, Sodium Channels metabolism, Bile Acids and Salts pharmacology, Bile Acids and Salts metabolism, Epithelial Sodium Channels metabolism
- Abstract
Bile acids, originally known to emulsify dietary lipids, are now established signalling molecules that regulate physiological processes. Signalling targets several proteins that include the ion channels involved in regulating intestinal motility and bile viscosity. Studies show that bile acids regulate the epithelial sodium channel (ENaC) in cultured cell models and heterologous expression systems. ENaC plays both local and systemic roles in regulating extracellular fluids. Here we investigated whether bile acids regulate ENaC expressed in native tissues. We found that taurocholic acid and taurohyodeoxycholic acid regulated ENaC in both the distal nephron and distal colon. We also tested the hypothesis that regulation occurs through direct binding. Using photoaffinity labelling, we found evidence for specific binding to both the β and γ subunits of the channel. In functional experiments, we found that the α subunit was sufficient for regulation. We also found that regulation by at least one bile acid was voltage-sensitive, suggesting that one binding site may be closely associated with the pore-forming helices of the channel. Our data provide evidence that bile acids regulate ENaC by binding to multiple sites to influence the open probability of the channel. KEY POINTS: Recent studies have shown that bile acids regulate the epithelial sodium channel (ENaC) in vitro. Here we investigated whether bile acids regulate ENaC in native tissues and whether bile acids directly bind the channel. We found that bile acids regulate ENaC expressed in the mouse cortical collecting duct and mouse colon by modulating open probability. Photoaffinity labelling experiments showed specific binding to the β and γ subunits of the channel, while channels comprising only α subunits were sensitive to taurocholic acid in functional experiments using Xenopus oocytes. Taurocholic acid regulation of ENaC was voltage-dependent, providing evidence for binding to pore-forming helices. Our data indicate that bile acids are ENaC regulatory effectors that may have a role in the physiology and pathophysiology of several systems., (© 2022 The Authors. The Journal of Physiology © 2022 The Physiological Society.)
- Published
- 2022
- Full Text
- View/download PDF
15. Activation by cleavage of the epithelial Na + channel α and γ subunits independently coevolved with the vertebrate terrestrial migration.
- Author
-
Wang XP, Balchak DM, Gentilcore C, Clark NL, and Kashlan OB
- Subjects
- Amphibian Proteins genetics, Amphibian Proteins metabolism, Animals, Epithelial Sodium Channels metabolism, Fish Proteins genetics, Fish Proteins metabolism, Fishes metabolism, Xenopus laevis metabolism, Epithelial Sodium Channels genetics, Evolution, Molecular, Fishes genetics, Xenopus laevis genetics
- Abstract
Vertebrates evolved mechanisms for sodium conservation and gas exchange in conjunction with migration from aquatic to terrestrial habitats. Epithelial Na
+ channel (ENaC) function is critical to systems responsible for extracellular fluid homeostasis and gas exchange. ENaC is activated by cleavage at multiple specific extracellular polybasic sites, releasing inhibitory tracts from the channel's α and γ subunits. We found that proximal and distal polybasic tracts in ENaC subunits coevolved, consistent with the dual cleavage requirement for activation observed in mammals. Polybasic tract pairs evolved with the terrestrial migration and the appearance of lungs, coincident with the ENaC activator aldosterone, and appeared independently in the α and γ subunits. In summary, sites within ENaC for protease activation developed in vertebrates when renal Na+ conservation and alveolar gas exchange were required for terrestrial survival., Competing Interests: XW, DB, CG, NC, OK No competing interests declared, (© 2022, Wang et al.)- Published
- 2022
- Full Text
- View/download PDF
16. Distinct functions of megalin and cubilin receptors in recovery of normal and nephrotic levels of filtered albumin.
- Author
-
Ren Q, Weyer K, Rbaibi Y, Long KR, Tan RJ, Nielsen R, Christensen EI, Baty CJ, Kashlan OB, and Weisz OA
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Albuminuria genetics, Albuminuria physiopathology, Animals, Apoptosis Regulatory Proteins genetics, Apoptosis Regulatory Proteins metabolism, Cell Line, Disease Models, Animal, Endocytosis, Female, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Kidney Tubules, Proximal physiopathology, Kinetics, Low Density Lipoprotein Receptor-Related Protein-2 deficiency, Low Density Lipoprotein Receptor-Related Protein-2 genetics, Membrane Proteins genetics, Membrane Proteins metabolism, Mice, Knockout, Models, Biological, Nephrosis genetics, Nephrosis physiopathology, Opossums, Receptors, Cell Surface deficiency, Receptors, Cell Surface genetics, Albuminuria metabolism, Kidney Tubules, Proximal metabolism, Low Density Lipoprotein Receptor-Related Protein-2 metabolism, Nephrosis metabolism, Receptors, Cell Surface metabolism, Serum Albumin metabolism
- Abstract
Proximal tubule (PT) cells express a single saturable albumin-binding site whose affinity matches the estimated tubular concentration of albumin; however, albumin uptake capacity is greatly increased under nephrotic conditions. Deciphering the individual contributions of megalin and cubilin to the uptake of normal and nephrotic levels of albumin is impossible in vivo, as knockout of megalin in mice globally disrupts PT endocytic uptake. We quantified concentration-dependent albumin uptake in an optimized opossum kidney cell culture model and fit the kinetic profiles to identify albumin-binding affinities and uptake capacities. Mathematical deconvolution fit best to a three-component model that included saturable high- and low-affinity uptake sites for albumin and underlying nonsaturable uptake consistent with passive uptake of albumin in the fluid phase. Knockdown of cubilin or its chaperone amnionless selectively reduced the binding capacity of the high-affinity site, whereas knockdown of megalin impacted the low-affinity site. Knockdown of disabled-2 decreased the capacities of both binding sites. Additionally, knockdown of megalin or disabled-2 profoundly inhibited the uptake of a fluid phase marker, with cubilin knockdown having a more modest effect. We propose a novel model for albumin retrieval along the PT in which cubilin and megalin receptors have different functions in recovering filtered albumin in proximal tubule cells. Cubilin binding to albumin is tuned to capture normally filtered levels of the protein. In contrast, megalin binding to albumin is of lower affinity, and its expression is also essential for enabling the recovery of high concentrations of albumin in the fluid phase.
- Published
- 2020
- Full Text
- View/download PDF
17. Murine epithelial sodium (Na + ) channel regulation by biliary factors.
- Author
-
Wang XP, Im SJ, Balchak DM, Montalbetti N, Carattino MD, Ray EC, and Kashlan OB
- Subjects
- Animals, Antioxidants pharmacology, Cholagogues and Choleretics pharmacology, Epithelial Sodium Channels genetics, Gastrointestinal Agents pharmacology, Humans, Ion Transport, Lipoylation, Mice, Oocytes cytology, Oocytes drug effects, Oocytes metabolism, Proteolysis, Xenopus laevis, Bile Acids and Salts pharmacology, Bilirubin pharmacology, Deoxycholic Acid pharmacology, Epithelial Sodium Channels metabolism, Gene Expression Regulation drug effects, Sodium metabolism
- Abstract
The epithelial sodium channel (ENaC) mediates Na
+ transport in several epithelia, including the aldosterone-sensitive distal nephron, distal colon, and biliary epithelium. Numerous factors regulate ENaC activity, including extracellular ligands, post-translational modifications, and membrane-resident lipids. However, ENaC regulation by bile acids and conjugated bilirubin, metabolites that are abundant in the biliary tree and intestinal tract and are sometimes elevated in the urine of individuals with advanced liver disease, remains poorly understood. Here, using a Xenopus oocyte-based system to express and functionally study ENaC, we found that, depending on the bile acid used, bile acids both activate and inhibit mouse ENaC. Whether bile acids were activating or inhibiting was contingent on the position and orientation of specific bile acid moieties. For example, a hydroxyl group at the 12-position and facing the hydrophilic side (12α-OH) was activating. Taurine-conjugated bile acids, which have reduced membrane permeability, affected ENaC activity more strongly than did their more membrane-permeant unconjugated counterparts, suggesting that bile acids regulate ENaC extracellularly. Bile acid-dependent activation was enhanced by amino acid substitutions in ENaC that depress open probability and was precluded by proteolytic cleavage that increases open probability, consistent with an effect of bile acids on ENaC open probability. Bile acids also regulated ENaC in a cortical collecting duct cell line, mirroring the results in Xenopus oocytes. We also show that bilirubin conjugates activate ENaC. These results indicate that ENaC responds to compounds abundant in bile and that their ability to regulate this channel depends on the presence of specific functional groups., (© 2019 Wang et al.)- Published
- 2019
- Full Text
- View/download PDF
18. The epithelial Na + channel γ subunit autoinhibitory tract suppresses channel activity by binding the γ subunit's finger-thumb domain interface.
- Author
-
Balchak DM, Thompson RN, and Kashlan OB
- Subjects
- Animals, Binding Sites, Epithelial Sodium Channels metabolism, Humans, Oocytes, Proteolysis, Xenopus laevis, Allosteric Regulation, Epithelial Sodium Channels physiology, Protein Interaction Domains and Motifs, Protein Subunits metabolism
- Abstract
Epithelial Na
+ channel (ENaC) maturation and activation require proteolysis of both the α and γ subunits. Cleavage at multiple sites in the finger domain of each subunit liberates their autoinhibitory tracts. Synthetic peptides derived from the proteolytically released fragments inhibit the channel, likely by reconstituting key interactions removed by the proteolysis. We previously showed that a peptide derived from the α subunit's autoinhibitory sequence (α-8) binds at the α subunit's finger-thumb domain interface. Despite low sequence similarity between the α and γ subunit finger domains, we hypothesized that a peptide derived from the γ subunit's autoinhibitory sequence (γ-11) inhibits the channel through an analogous mechanism. Using Xenopus oocytes, we found here that channels lacking a γ subunit thumb domain were no longer sensitive to γ-11, but remained sensitive to α-8. We identified finger domain sites in the γ subunit that dramatically reduced γ-11 inhibition. Using cysteines and sulfhydryl reactive cross-linkers introduced into both the peptide and the subunit, we also could cross-link γ-11 to both the finger domain and the thumb domain of the γ subunit. Our results suggest that α-8 and γ-11 occupy similar binding pockets within their respective subunits, and that proteolysis of the α and γ subunits activate the channel through analogous mechanisms., (© 2018 Balchak et al.)- Published
- 2018
- Full Text
- View/download PDF
19. Pore-lining residues of MEC-4 and MEC-10 channel subunits tune the Caenorhabditis elegans degenerin channel's response to shear stress.
- Author
-
Shi S, Mutchler SM, Blobner BM, Kashlan OB, and Kleyman TR
- Subjects
- Amino Acid Sequence, Amino Acids chemistry, Amino Acids genetics, Animals, Caenorhabditis elegans growth & development, Caenorhabditis elegans Proteins chemistry, Caenorhabditis elegans Proteins genetics, Cells, Cultured, Membrane Proteins chemistry, Membrane Proteins genetics, Mutagenesis, Site-Directed, Mutation, Oocytes cytology, Oocytes metabolism, Protein Conformation, Sequence Homology, Xenopus laevis growth & development, Amino Acids metabolism, Caenorhabditis elegans metabolism, Caenorhabditis elegans Proteins metabolism, Membrane Proteins metabolism, Stress, Mechanical, Xenopus laevis metabolism
- Abstract
The Caenorhabditis elegans MEC-4/MEC-10 channel mediates the worm's response to gentle body touch and is activated by laminar shear stress (LSS) when expressed in Xenopus oocytes. Substitutions at multiple sites within the second transmembrane domain (TM2) of MEC-4 or MEC-10 abolish the gentle touch response in worms, but the roles of these residues in mechanosensing are unclear. The present study therefore examined the role of specific MEC-4 and MEC-10 TM2 residues in the channel's response to LSS. We found that introducing mutations within the TM2 of MEC-4 or MEC-10 not only altered channel activity, but also affected the channel's response to LSS. This response was enhanced by Cys substitutions at selected MEC-4 sites (Phe
715 , Gly716 , Gln718 , and Leu719 ) between the degenerin and the putative amiloride-binding sites in this subunit. In contrast, the LSS response was largely blunted in MEC-10 variants bearing single Cys substitutions in the regions preceding and following the amiloride-binding site (Gly677 -Leu681 ), as well as with four MEC-10 touch-deficient mutations that introduced charged residues into the TM2 domain. An enhanced response to LSS was observed with a MEC-10 mutation in the putative selectivity filter. Overall, MEC-4 or MEC-10 mutants that altered the channel's LSS response are primarily clustered between the degenerin site and the selectivity filter, a region that probably forms the narrowest portion of the channel pore. Our results suggest that pore-lining residues of MEC-4 and MEC-10 have important yet different roles in tuning the channel's response to mechanical forces., (© 2018 Shi et al.)- Published
- 2018
- Full Text
- View/download PDF
20. Conserved cysteines in the finger domain of the epithelial Na + channel α and γ subunits are proximal to the dynamic finger-thumb domain interface.
- Author
-
Blobner BM, Wang XP, and Kashlan OB
- Subjects
- Animals, Cysteine chemistry, Cysteine genetics, Cysteine metabolism, Epithelial Sodium Channels genetics, Epithelial Sodium Channels metabolism, Humans, Protein Domains, Xenopus laevis, Epithelial Sodium Channels chemistry
- Abstract
The epithelial Na
+ channel (ENaC) is a member of the ENaC/degenerin family of ion channels. In the structure of a related family member, the "thumb" domain's base interacts with the pore, and its tip interacts with the divergent "finger" domain. Between the base and tip, the thumb domain is characterized by a conserved five-rung disulfide ladder holding together two anti-parallel α helices. The ENaC α and γ subunits' finger domains harbor autoinhibitory tracts that can be proteolytically liberated to activate the channel and also host an ENaC-specific pair of cysteines. Using a crosslinking approach, we show that one of the finger domain cysteines in the α subunit (αCys-263) and both of the finger domain cysteines in the γ subunit (γCys-213 and γCys-220) lie near the dynamic finger-thumb domain interface. Our data suggest that the αCys-256/αCys-263 pair is not disulfide-bonded. In contrast, we found that the γCys-213/γCys-220 pair is disulfide-bonded. Our data also suggest that the γ subunit lacks the terminal rung in the thumb domain disulfide ladder, suggesting asymmetry between the subunits. We also observed functional asymmetry between the α and γ subunit finger-thumb domain interfaces; crosslinks bridging the α subunit finger-thumb interface only inhibited ENaC currents, whereas crosslinks bridging the γ subunit finger-thumb interface activated or inhibited currents dependent on the length of the crosslinker. Our data suggest that reactive cysteines lie at the dynamic finger-thumb interfaces of the α and γ subunits and may play a yet undefined role in channel regulation., (© 2018 by The American Society for Biochemistry and Molecular Biology, Inc.)- Published
- 2018
- Full Text
- View/download PDF
21. N-linked glycans are required on epithelial Na + channel subunits for maturation and surface expression.
- Author
-
Kashlan OB, Kinlough CL, Myerburg MM, Shi S, Chen J, Blobner BM, Buck TM, Brodsky JL, Hughey RP, and Kleyman TR
- Subjects
- Animals, Epithelial Sodium Channels chemistry, Epithelial Sodium Channels genetics, Glycosylation, Mechanotransduction, Cellular, Membrane Potentials, Mutation, Protein Conformation, Protein Folding, Protein Transport, Rats, Inbred F344, Structure-Activity Relationship, Trypsin metabolism, Xenopus laevis, Epithelial Sodium Channels metabolism, Protein Processing, Post-Translational, Sodium metabolism
- Abstract
Epithelial Na
+ channel (ENaC) subunits undergo N-linked glycosylation in the endoplasmic reticulum where they assemble into an αβγ complex. Six, 13, and 5 consensus sites (Asn-X-Ser/Thr) for N-glycosylation reside in the extracellular domains of the mouse α-, β-, and γ-subunits, respectively. Because the importance of ENaC N-linked glycans has not been fully addressed, we examined the effect of preventing N-glycosylation of specific subunits on channel function, expression, maturation, and folding. Heterologous expression in Xenopus oocytes or Fischer rat thyroid cells with αβγ-ENaC lacking N-linked glycans on a single subunit reduced ENaC activity as well as the inhibitory response to extracellular Na+ . The lack of N-linked glycans on the β-subunit also precluded channel activation by trypsin. However, channel activation by shear stress was N-linked glycan independent, regardless of which subunit was modified. We also discovered that the lack of N-linked glycans on any one subunit reduced the total and surface levels of cognate subunits. The lack of N-linked glycans on the β-subunit had the largest effect on total levels, with the lack of N-linked glycans on the γ- and α-subunits having intermediate and modest effects, respectively. Finally, channels with wild-type β-subunits were more sensitive to limited trypsin proteolysis than channels lacking N-linked glycans on the β-subunit. Our results indicate that N-linked glycans on each subunit are required for proper folding, maturation, surface expression, and function of the channel.- Published
- 2018
- Full Text
- View/download PDF
22. Epithelial Na + Channel Regulation by Extracellular and Intracellular Factors.
- Author
-
Kleyman TR, Kashlan OB, and Hughey RP
- Subjects
- Animals, Biological Transport, Humans, Lipoylation, Cell Membrane metabolism, Epithelial Sodium Channels metabolism, Stress, Physiological physiology
- Abstract
Epithelial Na
+ channels (ENaCs) are members of the ENaC/degenerin family of ion channels that evolved to respond to extracellular factors. In addition to being expressed in the distal aspects of the nephron, where ENaCs couple the absorption of filtered Na+ to K+ secretion, these channels are found in other epithelia as well as nonepithelial tissues. This review addresses mechanisms by which ENaC activity is regulated by extracellular factors, including proteases, Na+ , and shear stress. It also addresses other factors, including acidic phospholipids and modification of ENaC cytoplasmic cysteine residues by palmitoylation, which enhance channel activity by altering interactions of the channel with the plasma membrane.- Published
- 2018
- Full Text
- View/download PDF
23. Collecting duct principal cell transport processes and their regulation.
- Author
-
Pearce D, Soundararajan R, Trimpert C, Kashlan OB, Deen PM, and Kohan DE
- Subjects
- Animals, Aquaporin 2 metabolism, Body Water metabolism, Epithelial Sodium Channels metabolism, Humans, Ion Transport, Kidney Tubules, Collecting cytology, Potassium metabolism, Potassium Channels, Inwardly Rectifying metabolism, Signal Transduction, Sodium metabolism, Sodium-Potassium-Exchanging ATPase metabolism, Epithelial Cells metabolism, Kidney Tubules, Collecting metabolism, Water-Electrolyte Balance
- Abstract
The principal cell of the kidney collecting duct is one of the most highly regulated epithelial cell types in vertebrates. The effects of hormonal, autocrine, and paracrine factors to regulate principal cell transport processes are central to the maintenance of fluid and electrolyte balance in the face of wide variations in food and water intake. In marked contrast with the epithelial cells lining the proximal tubule, the collecting duct is electrically tight, and ion and osmotic gradients can be very high. The central role of principal cells in salt and water transport is reflected by their defining transporters-the epithelial Na(+) channel (ENaC), the renal outer medullary K(+) channel, and the aquaporin 2 (AQP2) water channel. The coordinated regulation of ENaC by aldosterone, and AQP2 by arginine vasopressin (AVP) in principal cells is essential for the control of plasma Na(+) and K(+) concentrations, extracellular fluid volume, and BP. In addition to these essential hormones, additional neuronal, physical, and chemical factors influence Na(+), K(+), and water homeostasis. Notably, a variety of secreted paracrine and autocrine agents such as bradykinin, ATP, endothelin, nitric oxide, and prostaglandin E2 counterbalance and limit the natriferic effects of aldosterone and the water-retaining effects of AVP. Considerable recent progress has improved our understanding of the transporters, receptors, second messengers, and signaling events that mediate principal cell responses to changing environments in health and disease. This review primarily addresses the structure and function of the key transporters and the complex interplay of regulatory factors that modulate principal cell ion and water transport., (Copyright © 2015 by the American Society of Nephrology.)
- Published
- 2015
- Full Text
- View/download PDF
24. Na+ inhibits the epithelial Na+ channel by binding to a site in an extracellular acidic cleft.
- Author
-
Kashlan OB, Blobner BM, Zuzek Z, Tolino M, and Kleyman TR
- Subjects
- Acid Sensing Ion Channels genetics, Action Potentials, Allosteric Regulation, Amiloride chemistry, Amino Acid Sequence, Animals, Binding Sites, Epithelial Sodium Channel Blockers chemistry, Epithelial Sodium Channels genetics, Gene Expression, Ion Transport, Mice, Models, Molecular, Molecular Sequence Data, Mutation, Oocytes, Patch-Clamp Techniques, Protein Binding, Protein Structure, Secondary, Protein Structure, Tertiary, Protein Subunits genetics, Sequence Alignment, Xenopus laevis, Acid Sensing Ion Channels chemistry, Epithelial Sodium Channels chemistry, Protein Subunits chemistry, Sodium chemistry
- Abstract
The epithelial Na(+) channel (ENaC) has a key role in the regulation of extracellular fluid volume and blood pressure. ENaC belongs to a family of ion channels that sense the external environment. These channels have large extracellular regions that are thought to interact with environmental cues, such as Na(+), Cl(-), protons, proteases, and shear stress, which modulate gating behavior. We sought to determine the molecular mechanism by which ENaC senses high external Na(+) concentrations, resulting in an inhibition of channel activity. Both our structural model of an ENaC α subunit and the resolved structure of an acid-sensing ion channel (ASIC1) have conserved acidic pockets in the periphery of the extracellular region of the channel. We hypothesized that these acidic pockets host inhibitory allosteric Na(+) binding sites. Through site-directed mutagenesis targeting the acidic pocket, we modified the inhibitory response to external Na(+). Mutations at selected sites altered the cation inhibitory preference to favor Li(+) or K(+) rather than Na(+). Channel activity was reduced in response to restraining movement within this region by cross-linking structures across the acidic pocket. Our results suggest that residues within the acidic pocket form an allosteric effector binding site for Na(+). Our study supports the hypothesis that an acidic cleft is a key ligand binding locus for ENaC and perhaps other members of the ENaC/degenerin family., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
25. Cysteine palmitoylation of the γ subunit has a dominant role in modulating activity of the epithelial sodium channel.
- Author
-
Mukherjee A, Mueller GM, Kinlough CL, Sheng N, Wang Z, Mustafa SA, Kashlan OB, Kleyman TR, and Hughey RP
- Subjects
- Animals, Binding Sites, Cytoplasm metabolism, Dogs, Epithelial Sodium Channel Blockers pharmacology, Humans, Ion Channel Gating drug effects, Madin Darby Canine Kidney Cells, Mice, Protein Subunits antagonists & inhibitors, Sodium pharmacology, Cysteine metabolism, Epithelial Sodium Channels chemistry, Epithelial Sodium Channels metabolism, Lipoylation, Protein Subunits chemistry, Protein Subunits metabolism
- Abstract
The epithelial sodium channel (ENaC) is composed of three homologous subunits (α, β, and γ) with cytoplasmic N and C termini. Our previous work revealed that two cytoplasmic Cys residues in the β subunit, βCys-43 and βCys-557, are Cys-palmitoylated. ENaCs with mutant βC43A/C557A exhibit normal surface expression but enhanced Na(+) self-inhibition and reduced channel open probability. Although the α subunit is not palmitoylated, we now show that the two cytoplasmic Cys residues in the γ subunit are palmitoylated. ENaCs with mutant γC33A, γC41A, or γC33A/C41A exhibit reduced activity compared with wild type channels but normal surface expression and normal levels of α and γ subunit-activating cleavage. These mutant channels have significantly enhanced Na(+) self-inhibition and reduced open probability compared with wild type ENaCs. Channel activity was enhanced by co-expression with the palmitoyltransferase DHHC2 that also co-immunoprecipitates with ENaCs. Secondary structure prediction of the N terminus of the γ subunit places γCys-33 within an α-helix and γCys-44 on a coil before the first transmembrane domain within a short tract that includes a well conserved His-Gly motif, where mutations have been associated with altered channel gating. Our current and previous results suggest that palmitoylation of the β and γ subunits of ENaCs enhances interactions of their respective cytoplasmic domains with the plasma membrane and stabilizes the open state of the channel. Comparison of activities of channels lacking palmitoylation sites in individual or multiple subunits revealed that γ subunit palmitoylation has a dominant role over β subunit palmitoylation in modulating ENaC gating., (© 2014 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
26. The Lhs1/GRP170 chaperones facilitate the endoplasmic reticulum-associated degradation of the epithelial sodium channel.
- Author
-
Buck TM, Plavchak L, Roy A, Donnelly BF, Kashlan OB, Kleyman TR, Subramanya AR, and Brodsky JL
- Subjects
- Adenosine Triphosphate metabolism, Amiloride pharmacology, Animals, Endoplasmic Reticulum metabolism, Epithelial Sodium Channels genetics, Female, Glycoproteins genetics, HEK293 Cells, HSP70 Heat-Shock Proteins genetics, Humans, Immunoblotting, Ion Transport drug effects, Membrane Potentials drug effects, Mutation, Oocytes metabolism, Oocytes physiology, Protein Binding, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins genetics, Sodium metabolism, Xenopus, Endoplasmic Reticulum-Associated Degradation, Epithelial Sodium Channels metabolism, Glycoproteins metabolism, HSP70 Heat-Shock Proteins metabolism, Saccharomyces cerevisiae Proteins metabolism
- Abstract
The epithelial sodium channel, ENaC, plays a critical role in maintaining salt and water homeostasis, and not surprisingly defects in ENaC function are associated with disease. Like many other membrane-spanning proteins, this trimeric protein complex folds and assembles inefficiently in the endoplasmic reticulum (ER), which results in a substantial percentage of the channel being targeted for ER-associated degradation (ERAD). Because the spectrum of factors that facilitates the degradation of ENaC is incomplete, we developed yeast expression systems for each ENaC subunit. We discovered that a conserved Hsp70-like chaperone, Lhs1, is required for maximal turnover of the ENaC α subunit. By expressing Lhs1 ATP binding mutants, we also found that the nucleotide exchange properties of this chaperone are dispensable for ENaC degradation. Consistent with the precipitation of an Lhs1-αENaC complex, Lhs1 holdase activity was instead most likely required to support the ERAD of αENaC. Moreover, a complex containing the mammalian Lhs1 homolog GRP170 and αENaC co-precipitated, and GRP170 also facilitated ENaC degradation in human, HEK293 cells, and in a Xenopus oocyte expression system. In both yeast and higher cell types, the effect of Lhs1 on the ERAD of αENaC was selective for the unglycosylated form of the protein. These data establish the first evidence that Lhs1/Grp170 chaperones can act as mediators of ERAD substrate selection.
- Published
- 2013
- Full Text
- View/download PDF
27. Multiple motifs regulate apical sorting of p75 via a mechanism that involves dimerization and higher-order oligomerization.
- Author
-
Youker RT, Bruns JR, Costa SA, Rbaibi Y, Lanni F, Kashlan OB, Teng H, and Weisz OA
- Subjects
- Animals, Binding Sites genetics, Cell Line, Dogs, Galectins genetics, Galectins metabolism, Glycosylation, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Microscopy, Confocal, Models, Biological, Mutation, Protein Transport, RNA Interference, Receptor, Nerve Growth Factor genetics, Protein Multimerization, Receptor, Nerve Growth Factor chemistry, Receptor, Nerve Growth Factor metabolism, trans-Golgi Network metabolism
- Abstract
The sorting signals that direct proteins to the apical surface of polarized epithelial cells are complex and can include posttranslational modifications, such as N- and O-linked glycosylation. Efficient apical sorting of the neurotrophin receptor p75 is dependent on its O-glycosylated membrane proximal stalk, but how this domain mediates targeting is unknown. Protein oligomerization or clustering has been suggested as a common step in the segregation of all apical proteins. Like many apical proteins, p75 forms dimers, and we hypothesized that formation of higher-order clusters mediated by p75 dimerization and interactions of the stalk facilitate its apical sorting. Using fluorescence fluctuation techniques (photon-counting histogram and number and brightness analyses) to study p75 oligomerization status in vivo, we found that wild-type p75-green fluorescent protein forms clusters in the trans-Golgi network (TGN) but not at the plasma membrane. Disruption of either the dimerization motif or the stalk domain impaired both clustering and polarized delivery. Manipulation of O-glycan processing or depletion of multiple galectins expressed in Madin-Darby canine kidney cells had no effect on p75 sorting, suggesting that the stalk domain functions as a structural prop to position other determinants in the lumenal domain of p75 for oligomerization. Additionally, a p75 mutant with intact dimerization and stalk motifs but with a dominant basolateral sorting determinant (Δ250 mutant) did not form oligomers, consistent with a requirement for clustering in apical sorting. Artificially enhancing dimerization restored clustering to the Δ250 mutant but was insufficient to reroute this mutant to the apical surface. Together these studies demonstrate that clustering in the TGN is required for normal biosynthetic apical sorting of p75 but is not by itself sufficient to reroute a protein to the apical surface in the presence of a strong basolateral sorting determinant. Our studies shed new light on the hierarchy of polarized sorting signals and on the mechanisms by which newly synthesized proteins are segregated in the TGN for eventual apical delivery.
- Published
- 2013
- Full Text
- View/download PDF
28. Inhibitory tract traps the epithelial Na+ channel in a low activity conformation.
- Author
-
Kashlan OB, Blobner BM, Zuzek Z, Carattino MD, and Kleyman TR
- Subjects
- Animals, Epithelial Sodium Channels genetics, Epithelial Sodium Channels metabolism, Mice, Protein Structure, Tertiary, Protein Subunits genetics, Protein Subunits metabolism, Epithelial Sodium Channels chemistry, Models, Molecular, Protein Subunits chemistry, Proteolysis
- Abstract
Proteolysis plays an important role in the maturation and activation of epithelial Na(+) channels (ENaCs). Non-cleaved channels are inactive at high extracellular Na(+) concentrations and fully cleaved channels are constitutively active. Cleavage of the α and γ subunits at multiple sites activates the channel through the release of imbedded inhibitory tracts. Peptides derived from these released tracts are also inhibitory, likely through binding at the inhibitory tract sites. We recently reported a model of the α subunit. We have now cross-linked Cys derivatives of the inhibitory peptide to the channel, using our model to predict sites at a domain interface of the α subunit that is in proximity to the N terminus of the peptide. Furthermore, peptide inhibition was mimicked in the absence of peptide by cross-linking the channel across the domain interface. Our results suggest a dynamic domain interface that can be exploited by inhibitory peptides and provides a mechanism for peptide inhibition and proteolytic activation.
- Published
- 2012
- Full Text
- View/download PDF
29. Epithelial Na(+) channel regulation by cytoplasmic and extracellular factors.
- Author
-
Kashlan OB and Kleyman TR
- Subjects
- Animals, Humans, Ion Channel Gating, Sodium metabolism, Ubiquitin metabolism, Cytoplasm metabolism, Epithelial Sodium Channels metabolism
- Abstract
Electrogenic Na(+) transport across high resistance epithelial is mediated by the epithelial Na(+) channel (ENaC). Our understanding of the mechanisms of ENaC regulation has continued to evolve over the two decades following the cloning of ENaC subunits. This review highlights many of the cellular and extracellular factors that regulate channel trafficking or gating., (Copyright © 2012. Published by Elsevier Inc.)
- Published
- 2012
- Full Text
- View/download PDF
30. Extracellular finger domain modulates the response of the epithelial sodium channel to shear stress.
- Author
-
Shi S, Blobner BM, Kashlan OB, and Kleyman TR
- Subjects
- Animals, Binding Sites genetics, Epithelial Sodium Channels genetics, Female, Ion Channel Gating genetics, Membrane Potentials drug effects, Mice, Models, Molecular, Mutation, Oocytes cytology, Oocytes metabolism, Oocytes physiology, Patch-Clamp Techniques, Protein Subunits chemistry, Protein Subunits genetics, Protein Subunits physiology, Sodium metabolism, Sodium pharmacology, Stress, Mechanical, Xenopus, Epithelial Sodium Channels chemistry, Epithelial Sodium Channels physiology, Ion Channel Gating physiology, Protein Structure, Tertiary
- Abstract
The epithelial sodium channel (ENaC) is regulated by multiple extracellular stimuli, including shear stress. Previous studies suggest that the extracellular finger domains of ENaC α and γ subunits contain allosteric regulatory modules. However, the role of the finger domain in the shear stress response is unknown. We examined whether mutations of specific residues in the finger domain of the α subunit altered the response of channels to shear stress. We observed that Trp substitutions at multiple sites within the tract αLys-250-αLeu-290 altered the magnitude or kinetics of channel activation by shear stress. Consistent with these findings, deletion of two predicted peripheral β strands (αIle-251-αTyr-268) led to slower channel activation by shear stress, suggesting that these structures participate in the shear stress response. The effects of mutations on the shear stress response did not correlate with their effects on allosteric Na(+) inhibition (i.e. Na(+) self-inhibition), indicating a divergence within the finger domain regarding mechanisms by which the channel responds to these two external stimuli. This result contrasts with well correlated effects we previously observed at sites near the extracellular mouth of the pore, suggesting mechanistic convergence in proximity to the pore. Our results suggest that the finger domain has an important role in the modulation of channel activity in response to shear stress.
- Published
- 2012
- Full Text
- View/download PDF
31. Structure of the pentameric ligand-gated ion channel ELIC cocrystallized with its competitive antagonist acetylcholine.
- Author
-
Pan J, Chen Q, Willenbring D, Yoshida K, Tillman T, Kashlan OB, Cohen A, Kong XP, Xu Y, and Tang P
- Subjects
- Acetylcholine metabolism, Crystallography, X-Ray, Cysteine Loop Ligand-Gated Ion Channel Receptors metabolism, Dickeya chrysanthemi cytology, Dickeya chrysanthemi metabolism, Ion Channel Gating, Ligand-Gated Ion Channels antagonists & inhibitors, Models, Molecular, Molecular Dynamics Simulation, Protein Structure, Quaternary, Protein Structure, Tertiary, Quaternary Ammonium Compounds chemistry, Quaternary Ammonium Compounds metabolism, Static Electricity, Acetylcholine chemistry, Cysteine Loop Ligand-Gated Ion Channel Receptors chemistry, Dickeya chrysanthemi chemistry, Ligand-Gated Ion Channels chemistry, Ligand-Gated Ion Channels metabolism
- Abstract
ELIC, the pentameric ligand-gated ion channel from Erwinia chrysanthemi, is a prototype for Cys-loop receptors. Here we show that acetylcholine is a competitive antagonist for ELIC. We determine the acetylcholine-ELIC cocrystal structure to a 2.9-Å resolution and find that acetylcholine binding to an aromatic cage at the subunit interface induces a significant contraction of loop C and other structural rearrangements in the extracellular domain. The side chain of the pore-lining residue F247 reorients and the pore size consequently enlarges, but the channel remains closed. We attribute the inability of acetylcholine to activate ELIC primarily to weak cation-π and electrostatic interactions in the pocket, because an acetylcholine derivative with a simple quaternary-to-tertiary ammonium substitution activates the channel. This study presents a compelling case for understanding the structural underpinning of the functional relationship between agonism and competitive antagonism in the Cys-loop receptors, providing a new framework for developing novel therapeutic drugs.
- Published
- 2012
- Full Text
- View/download PDF
32. ENaC structure and function in the wake of a resolved structure of a family member.
- Author
-
Kashlan OB and Kleyman TR
- Subjects
- Acid Sensing Ion Channels, Amiloride pharmacology, Amino Acid Sequence, Animals, Chlorides physiology, Conserved Sequence, Epithelial Sodium Channels drug effects, Humans, Ion Channel Gating drug effects, Ion Channel Gating physiology, Mice, Molecular Sequence Data, Nerve Tissue Proteins drug effects, Nerve Tissue Proteins physiology, Rats, Sequence Homology, Amino Acid, Sodium Channel Blockers pharmacology, Sodium Channels drug effects, Sodium Channels physiology, Structure-Activity Relationship, Epithelial Sodium Channels physiology
- Abstract
Our understanding of epithelial Na(+) channel (ENaC) structure and function has been profoundly impacted by the resolved structure of the homologous acid-sensing ion channel 1 (ASIC1). The structure of the extracellular and pore regions provide insight into channel assembly, processing, and the ability of these channels to sense the external environment. The absence of intracellular structures precludes insight into important interactions with intracellular factors that regulate trafficking and function. The primary sequences of ASIC1 and ENaC subunits are well conserved within the regions that are within or in close proximity to the plasma membrane, but poorly conserved in peripheral domains that may functionally differentiate family members. This review examines functional data, including ion selectivity, gating, and amiloride block, in light of the resolved ASIC1 structure.
- Published
- 2011
- Full Text
- View/download PDF
33. Galectin-7 modulates the length of the primary cilia and wound repair in polarized kidney epithelial cells.
- Author
-
Rondanino C, Poland PA, Kinlough CL, Li H, Rbaibi Y, Myerburg MM, Al-bataineh MM, Kashlan OB, Pastor-Soler NM, Hallows KR, Weisz OA, Apodaca G, and Hughey RP
- Subjects
- Animals, Cell Membrane physiology, Cells, Cultured, Dogs, Epithelial Cells cytology, Epithelial Cells ultrastructure, Galectins genetics, Humans, Integrin beta1 physiology, Kidney cytology, Kidney ultrastructure, Kidney Tubules, Proximal cytology, Kidney Tubules, Proximal physiology, Kidney Tubules, Proximal ultrastructure, Mice, Mice, Knockout, Protein Binding physiology, Rats, Swine, Cilia physiology, Cilia ultrastructure, Epithelial Cells physiology, Galectins physiology, Kidney physiology, Wound Healing physiology
- Abstract
Galectins (Gal) are β-galactoside-binding proteins that function in epithelial development and homeostasis. An overlapping role for Gal-3 and Gal-7 in wound repair was reported in stratified epithelia. Although Gal-7 was thought absent in simple epithelia, it was reported in a proteomic analysis of cilia isolated from cultured human airway, and we recently identified Gal-7 transcripts in Madin-Darby canine kidney (MDCK) cells (Poland PA, Rondanino C, Kinlough CL, Heimburg-Molinaro J, Arthur CM, Stowell SR, Smith DF, Hughey RP. J Biol Chem 286: 6780-6790, 2011). We now report that Gal-7 is localized exclusively on the primary cilium of MDCK, LLC-PK(1) (pig kidney), and mpkCCD(c14) (mouse kidney) cells as well as on cilia in the rat renal proximal tubule. Gal-7 is also present on most cilia of multiciliated cells in human airway epithelia primary cultures. Interestingly, exogenous glutathione S-transferase (GST)-Gal-7 bound the MDCK apical plasma membrane as well as the cilium, while the lectin Ulex europeaus agglutinin, with glycan preferences similar to Gal-7, bound the basolateral plasma membrane as well as the cilium. In pull-down assays, β1-integrin isolated from either the basolateral or apical/cilia membranes of MDCK cells was similarly bound by GST-Gal-7. Selective localization of Gal-7 to cilia despite the presence of binding sites on all cell surfaces suggests that intracellular Gal-7 is specifically delivered to cilia rather than simply binding to surface glycoconjugates after generalized secretion. Moreover, depletion of Gal-7 using tetracycline-induced short-hairpin RNA in mpkCCD(c14) cells significantly reduced cilia length and slowed wound healing in a scratch assay. We conclude that Gal-7 is selectively targeted to cilia and plays a key role in surface stabilization of glycoconjugates responsible for integrating cilia function with epithelial repair.
- Published
- 2011
- Full Text
- View/download PDF
34. Insights into the mechanism of pore opening of acid-sensing ion channel 1a.
- Author
-
Tolino LA, Okumura S, Kashlan OB, and Carattino MD
- Subjects
- Acid Sensing Ion Channels, Amino Acid Substitution, Animals, Disulfides, Indicators and Reagents pharmacology, Ion Transport drug effects, Ion Transport physiology, Mesylates pharmacology, Mice, Mutagenesis, Mutation, Missense, Nerve Tissue Proteins genetics, Protein Structure, Tertiary, Sodium Channels genetics, Xenopus laevis, Nerve Tissue Proteins metabolism, Sodium Channels metabolism
- Abstract
Acid-sensing ion channels (ASICs) are trimeric cation channels that undergo activation and desensitization in response to extracellular acidification. The underlying mechanism coupling proton binding in the extracellular region to pore gating is unknown. Here we probed the reactivity toward methanethiosulfonate (MTS) reagents of channels with cysteine-substituted residues in the outer vestibule of the pore of ASIC1a. We found that positively-charged MTS reagents trigger pore opening of G428C. Scanning mutagenesis of residues in the region preceding the second transmembrane spanning domain indicated that the MTSET-modified side chain of Cys at position 428 interacts with Tyr-424. This interaction was confirmed by double-mutant cycle analysis. Strikingly, Y424C-G428C monomers were associated by intersubunit disulfide bonds and were insensitive to MTSET. Despite the spatial constraints introduced by these intersubunit disulfide bonds in the outer vestibule of the pore, Y424C-G428C transitions between the resting, open, and desensitized states in response to extracellular acidification. This finding suggests that the opening of the ion conductive pathway involves coordinated rotation of the second transmembrane-spanning domains.
- Published
- 2011
- Full Text
- View/download PDF
35. Base of the thumb domain modulates epithelial sodium channel gating.
- Author
-
Shi S, Ghosh DD, Okumura S, Carattino MD, Kashlan OB, Sheng S, and Kleyman TR
- Subjects
- Acid Sensing Ion Channels, Animals, Epithelial Sodium Channels physiology, Mice, Mutation, Oocytes, Patch-Clamp Techniques, Protein Structure, Tertiary, Sodium, Stress, Mechanical, Xenopus laevis, Epithelial Sodium Channels chemistry, Ion Channel Gating physiology, Nerve Tissue Proteins chemistry, Sodium Channels chemistry
- Abstract
The activity of the epithelial sodium channel (ENaC) is modulated by multiple external factors, including proteases, cations, anions and shear stress. The resolved crystal structure of acid-sensing ion channel 1 (ASIC1), a structurally related ion channel, and mutagenesis studies suggest that the large extracellular region is involved in recognizing external signals that regulate channel gating. The thumb domain in the extracellular region of ASIC1 has a cylinder-like structure with a loop at its base that is in proximity to the tract connecting the extracellular region to the transmembrane domains. This loop has been proposed to have a role in transmitting proton-induced conformational changes within the extracellular region to the gate. We examined whether loops at the base of the thumb domains within ENaC subunits have a similar role in transmitting conformational changes induced by external Na(+) and shear stress. Mutations at selected sites within this loop in each of the subunits altered channel responses to both external Na(+) and shear stress. The most robust changes were observed at the site adjacent to a conserved Tyr residue. In the context of channels that have a low open probability due to retention of an inhibitory tract, mutations in the loop activated channels in a subunit-specific manner. Our data suggest that this loop has a role in modulating channel gating in response to external stimuli, and are consistent with the hypothesis that external signals trigger movements within the extracellular regions of ENaC subunits that are transmitted to the channel gate.
- Published
- 2011
- Full Text
- View/download PDF
36. Constraint-based, homology model of the extracellular domain of the epithelial Na+ channel α subunit reveals a mechanism of channel activation by proteases.
- Author
-
Kashlan OB, Adelman JL, Okumura S, Blobner BM, Zuzek Z, Hughey RP, Kleyman TR, and Grabe M
- Subjects
- Amino Acid Sequence, Animals, Binding Sites, Epithelial Sodium Channel Blockers, Epithelial Sodium Channels genetics, Furin metabolism, Ion Channel Gating, Mice, Molecular Sequence Data, Movement, Mutagenesis, Site-Directed, Mutation, Protein Structure, Tertiary, Epithelial Sodium Channels chemistry, Epithelial Sodium Channels metabolism, Extracellular Space metabolism, Models, Molecular, Peptide Hydrolases metabolism, Sequence Homology, Amino Acid
- Abstract
The epithelial Na(+) channel (ENaC) mediates Na(+) transport across high resistance epithelia. This channel is assembled from three homologous subunits with the majority of the protein's mass found in the extracellular domains. Acid-sensing ion channel 1 (ASIC1) is homologous to ENaC, but a key functional domain is highly divergent. Here we present molecular models of the extracellular region of α ENaC based on a large data set of mutations that attenuate inhibitory peptide binding in combination with comparative modeling based on the resolved structure of ASIC1. The models successfully rationalized the data from the peptide binding screen. We engineered new mutants that had not been tested based on the models and successfully predict sites where mutations affected peptide binding. Thus, we were able to confirm the overall general fold of our structural models. Further analysis suggested that the α subunit-derived inhibitory peptide affects channel gating by constraining motions within two major domains in the extracellular region, the thumb and finger domains.
- Published
- 2011
- Full Text
- View/download PDF
37. Allosteric inhibition of the epithelial Na+ channel through peptide binding at peripheral finger and thumb domains.
- Author
-
Kashlan OB, Boyd CR, Argyropoulos C, Okumura S, Hughey RP, Grabe M, and Kleyman TR
- Subjects
- Allosteric Regulation drug effects, Allosteric Regulation genetics, Animals, Binding Sites, Epithelial Sodium Channels genetics, Mice, Mutation, Peptides genetics, Peptides metabolism, Protein Structure, Tertiary, Protein Subunits genetics, Xenopus laevis, Epithelial Sodium Channel Blockers, Epithelial Sodium Channels metabolism, Peptides pharmacology, Protein Subunits antagonists & inhibitors, Protein Subunits metabolism
- Abstract
The epithelial Na(+) channel (ENaC) mediates the rate-limiting step in transepithelial Na(+) transport in the distal segments of the nephron and in the lung. ENaC subunits are cleaved by proteases, resulting in channel activation due to the release of inhibitory tracts. Peptides derived from these tracts inhibit channel activity. The mechanism by which these intrinsic inhibitory tracts reduce channel activity is unknown, as are the sites where these tracts interact with other residues within the channel. We performed site-directed mutagenesis in large portions of the predicted periphery of the extracellular region of the α subunit and measured the effect of mutations on an 8-residue inhibitory tract-derived peptide. Our data show that the inhibitory peptide likely binds to specific residues within the finger and thumb domains of ENaC. Pairwise interactions between the peptide and the channel were identified by double mutant cycle experiments. Our data suggest that the inhibitory peptide has a specific peptide orientation within its binding site. Extended to the intrinsic inhibitory tract, our data suggest that proteases activate ENaC by removing residues that bind at the finger-thumb domain interface.
- Published
- 2010
- Full Text
- View/download PDF
38. Cys palmitoylation of the beta subunit modulates gating of the epithelial sodium channel.
- Author
-
Mueller GM, Maarouf AB, Kinlough CL, Sheng N, Kashlan OB, Okumura S, Luthy S, Kleyman TR, and Hughey RP
- Subjects
- Amiloride pharmacology, Amino Acid Substitution, Animals, Cell Line, Computer Simulation, Dogs, Epithelial Sodium Channels genetics, Mice, Models, Molecular, Mutation, Mutation, Missense, Oocytes, Protein Structure, Secondary, Protein Structure, Tertiary, Protein Transport physiology, Sodium Channel Blockers pharmacology, Xenopus laevis, Cell Membrane metabolism, Epithelial Sodium Channels metabolism, Ion Channel Gating physiology, Lipoylation physiology, Sodium metabolism
- Abstract
The epithelial Na(+) channel (ENaC) is comprised of three homologous subunits (α, β, and γ) that have a similar topology with two transmembrane domains, a large extracellular region, and cytoplasmic N and C termini. Although ENaC activity is regulated by a number of factors, palmitoylation of its cytoplasmic Cys residues has not been previously described. Fatty acid-exchange chemistry was used to determine whether channel subunits were Cys-palmitoylated. We observed that only the β and γ subunits were modified by Cys palmitoylation. Analyses of ENaCs with mutant β subunits revealed that Cys-43 and Cys-557 were palmitoylated. Xenopus oocytes expressing ENaC with a β C43A,C557A mutant had significantly reduced amiloride-sensitive whole cell currents, enhanced Na(+) self-inhibition, and reduced single channel P(o) when compared with wild-type ENaC, while membrane trafficking and levels of surface expression were unchanged. Computer modeling of cytoplasmic domains indicated that β Cys-43 is in proximity to the first transmembrane α helix, whereas β Cys-557 is within an amphipathic α-helix contiguous with the second transmembrane domain. We propose that β subunit palmitoylation modulates channel gating by facilitating interactions between cytoplasmic domains and the plasma membrane.
- Published
- 2010
- Full Text
- View/download PDF
39. Defining an inhibitory domain in the gamma subunit of the epithelial sodium channel.
- Author
-
Passero CJ, Carattino MD, Kashlan OB, Myerburg MM, Hughey RP, and Kleyman TR
- Subjects
- Animals, Cell Line, Cells, Cultured, Epithelial Sodium Channels drug effects, Epithelial Sodium Channels metabolism, Female, Furin pharmacology, Humans, Kidney Tubules, Collecting cytology, Kidney Tubules, Collecting metabolism, Mice, Oocytes cytology, Oocytes metabolism, Respiratory Mucosa cytology, Respiratory Mucosa metabolism, Serine Endopeptidases pharmacology, Xenopus laevis, Epithelial Sodium Channels analysis, Protein Structure, Tertiary
- Abstract
Proteases activate the epithelial sodium channel (ENaC) by cleaving the large extracellular domains of the α- and γ-subunits and releasing peptides with inhibitory properties. Furin and prostasin activate mouse ENaC by cleaving the γ-subunit at sites flanking a 43 residue inhibitory tract (γE144-K186). To determine whether there is a minimal inhibitory region within this 43 residue tract, we generated serial deletions in the inhibitory tract of the γ-subunit in channels resistant to cleavage by furin and prostasin. We found that partial or complete deletion of a short segment in the γ-subunit, R158-N171, enhanced channel activity. Synthetic peptides overlapping this segment in the γ-subunit further identified a key 11-mer tract, R158-F168 (RFLNLIPLLVF), which inhibited wild-type ENaC expressed in Xenopus laevis oocytes, and endogenous channels in mpkCCD cells and human airway epithelia. Further studies with amino acid-substituted peptides defined residues that are required for inhibition in this key 11-mer tract. The presence of the native γ inhibitory tract in ENaC weakened the intrinsic binding constant of the 11-mer peptide inhibitor, suggesting that the γ inhibitory tract and the 11-mer peptide interact at overlapping sites within the channel.
- Published
- 2010
- Full Text
- View/download PDF
40. Epithelial sodium channel exit from the endoplasmic reticulum is regulated by a signal within the carboxyl cytoplasmic domain of the alpha subunit.
- Author
-
Mueller GM, Kashlan OB, Bruns JB, Maarouf AB, Aridor M, Kleyman TR, and Hughey RP
- Subjects
- Animals, Cell Line, Cytoplasm metabolism, Dogs, Glycoside Hydrolases metabolism, Golgi Apparatus metabolism, Mice, Models, Biological, Mutation, Protein Structure, Tertiary, Time Factors, gamma-Glutamyltransferase metabolism, Endoplasmic Reticulum metabolism, Epithelial Sodium Channels metabolism
- Abstract
Epithelial sodium channels (ENaCs) are assembled in the endoplasmic reticulum (ER) from alpha, beta, and gamma subunits, each with two transmembrane domains, a large extracellular loop, and cytoplasmic amino and carboxyl termini. ENaC maturation involves transit through the Golgi complex where Asn-linked glycans are processed to complex type and the channel is activated by furin-dependent cleavage of the alpha and gamma subunits. To identify signals in ENaC for ER retention/retrieval or ER exit/release, chimera were prepared with the interleukin alpha subunit (Tac) and each of the three cytoplasmic carboxyl termini of mouse ENaC (Tac-Ct) or with gamma-glutamyltranspeptidase and each of the three cytoplasmic amino termini (Nt-GGT). By monitoring acquisition of endoglycosidase H resistance after metabolic labeling, we found no evidence of ER retention of any chimera when compared with control Tac or GGT, but we did observe enhanced exit of Tac-alphaCt when compared with Tac. ER exit of ENaC was assayed after metabolic labeling by following the appearance of cleaved alpha as cleaved alpha subunit, but not non-cleaved alpha, is endoglycosidase H-resistant. Interestingly ER exit of epitope-tagged and truncated alpha (alphaDelta624-699-V5) with full-length betagamma was similar to wild type alpha (+betagamma), whereas ER exit of ENaC lacking the entire cytoplasmic carboxyl tail of alpha (alphaDelta613-699-V5 +betagamma) was significantly reduced. Subsequent analysis of ER exit for ENaCs with mutations within the intervening sequence (613)HRFRSRYWSPG(623) within the context of the full-length alpha revealed that mutation alphaRSRYW(620) to AAAAA significantly reduced ER exit. These data indicate that ER exit of ENaC is regulated by a signal within the alpha subunit carboxyl cytoplasmic tail.
- Published
- 2007
- Full Text
- View/download PDF
41. Small heat shock protein alphaA-crystallin regulates epithelial sodium channel expression.
- Author
-
Kashlan OB, Mueller GM, Qamar MZ, Poland PA, Ahner A, Rubenstein RC, Hughey RP, Brodsky JL, and Kleyman TR
- Subjects
- Animals, Dogs, Endoplasmic Reticulum metabolism, Heat-Shock Proteins metabolism, Mice, Models, Biological, Molecular Chaperones metabolism, Oocytes metabolism, Protein Structure, Tertiary, Sodium chemistry, Sodium Channels chemistry, Sodium Channels metabolism, Xenopus, Epithelium metabolism, Gene Expression Regulation, Sodium Channels biosynthesis, alpha-Crystallin A Chain metabolism, alpha-Crystallin A Chain physiology
- Abstract
Integral membrane proteins are synthesized on the cytoplasmic face of the endoplasmic reticulum (ER). After being translocated or inserted into the ER, they fold and undergo post-translational modifications. Within the ER, proteins are also subjected to quality control checkpoints, during which misfolded proteins may be degraded by proteasomes via a process known as ER-associated degradation. Molecular chaperones, including the small heat shock protein alphaA-crystallin, have recently been shown to play a role in this process. We have now found that alphaA-crystallin is expressed in cultured mouse collecting duct cells, where apical Na(+) transport is mediated by epithelial Na(+) channels (ENaC). ENaC-mediated Na(+) currents in Xenopus oocytes were reduced by co-expression of alphaA-crystallin. This reduction in ENaC activity reflected a decrease in the number of channels expressed at the cell surface. Furthermore, we observed that the rate of ENaC delivery to the cell surface of Xenopus oocytes was significantly reduced by co-expression of alphaA-crystallin, whereas the rate of channel retrieval remained unchanged. We also observed that alphaA-crystallin and ENaC co-immunoprecipitate. These data are consistent with the hypothesis that small heat shock proteins recognize ENaC subunits at ER quality control checkpoints and can target ENaC subunits for ER-associated degradation.
- Published
- 2007
- Full Text
- View/download PDF
42. Distinct structural elements in the first membrane-spanning segment of the epithelial sodium channel.
- Author
-
Kashlan OB, Maarouf AB, Kussius C, Denshaw RM, Blumenthal KM, and Kleyman TR
- Subjects
- Amino Acid Sequence, Animals, Cell Membrane metabolism, Epithelial Sodium Channels genetics, Epithelial Sodium Channels metabolism, Humans, Mice, Models, Molecular, Molecular Sequence Data, Mutagenesis, Site-Directed, Patch-Clamp Techniques, Protein Conformation, Protein Structure, Secondary, Protein Structure, Tertiary, Tryptophan chemistry, Epithelial Sodium Channels chemistry
- Abstract
Epithelial Na+ channels (ENaCs) comprise three subunits that have been proposed to be arranged in either an alpha2betagamma or a higher ordered configuration. Each subunit has two putative membrane-spanning segments (M1 and M2), intracellular amino and carboxyl termini, and a large extracellular loop. We have used the TOXCAT assay (a reporter assay for transmembrane segment homodimerization) to identify residues within the transmembrane segments of ENaC that may participate in important structural interactions within ENaC, with which we identified a candidate site within alphaM1. We performed site-directed mutagenesis at this site and found that, although the mutants reduced channel activity, ENaC protein expression at the plasma membrane was unaffected. To deduce the role of alphaM1 in the pore structure of ENaC, we performed tryptophan-scanning mutagenesis throughout alphaM1 (residues 110-130). We found that mutations within the amino-terminal part of alphaM1 had effects on activity and selectivity with a periodicity consistent with a helical structure but no effect on channel surface expression. We also observed that mutations within the carboxyl-terminal part of alphaM1 had effects on activity and selectivity but with no apparent periodicity. Additionally, these mutants reduced channel surface expression. Our data support a model in which the amino-terminal half of alphaM1 is alpha-helical and packs against structural element(s) that contribute to the ENaC pore. Furthermore, these data suggest that the carboxyl-terminal half of alphaM1 may be helical or assume a different conformation and may be involved in tertiary interactions essential to proper channel folding or assembly. Together, our data suggest that alphaM1 is divided into two distinct regions.
- Published
- 2006
- Full Text
- View/download PDF
43. Recycling of MUC1 is dependent on its palmitoylation.
- Author
-
Kinlough CL, McMahan RJ, Poland PA, Bruns JB, Harkleroad KL, Stremple RJ, Kashlan OB, Weixel KM, Weisz OA, and Hughey RP
- Subjects
- Animals, CHO Cells, Cell Membrane metabolism, Cells, Cultured, Cricetinae, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Half-Life, Immunoprecipitation, Mucin-1 genetics, Receptors, Interleukin-2 genetics, Receptors, Interleukin-2 metabolism, Subcellular Fractions, Transcription Factor AP-1 metabolism, rab GTP-Binding Proteins metabolism, Endocytosis, Mucin-1 metabolism, Palmitates metabolism
- Abstract
MUC1 is a mucin-like transmembrane protein expressed on the apical surface of epithelia, where it protects the cell surface. The cytoplasmic domain has numerous sites for phosphorylation and docking of proteins involved in signal transduction. In a previous study, we showed that the cytoplasmic YXXphi motif Y20HPM and the tyrosine-phosphorylated Y60TNP motif are required for MUC1 clathrin-mediated endocytosis through binding AP-2 and Grb2, respectively (Kinlough, C. L., Poland, P. A., Bruns, J. B., Harkleroad, K. L., and Hughey, R. P. (2004) J. Biol. Chem. 279, 53071-53077). Palmitoylation of transmembrane proteins can affect their membrane trafficking, and the MUC1 sequence CQC3RRK at the boundary of the transmembrane and cytoplasmic domains mimics reported site(s) of S-palmitoylation. [3H]Palmitate labeling of Chinese hamster ovary cells expressing MUC1 with mutations in CQC3RRK revealed that MUC1 is dually palmitoylated at the CQC motif independent of RRK. Lack of palmitoylation did not affect the cold detergent solubility profile of a chimera (Tac ectodomain and MUC1 transmembrane and cytoplasmic domains), the rate of chimera delivery to the cell surface, or its half-life. Calculation of rate constants for membrane trafficking of wild-type and mutant Tac-MUC1 indicated that the lack of palmitoylation blocked recycling, but not endocytosis, and caused the chimera to accumulate in a EGFP-Rab11-positive endosomal compartment. Mutations CQC/AQA and Y20N inhibited Tac-MUC1 co-immunoprecipitation with AP-1, although mutant Y20N had reduced rates of both endocytosis and recycling, but a normal subcellular distribution. The double mutant chimera AQA+Y20N had reduced endocytosis and recycling rates and accumulated in EGFP-Rab11-positive endosomes, indicating that palmitoylation is the dominant feature modulating MUC1 recycling from endosomes back to the plasma membrane.
- Published
- 2006
- Full Text
- View/download PDF
44. Differential effects of Hsc70 and Hsp70 on the intracellular trafficking and functional expression of epithelial sodium channels.
- Author
-
Goldfarb SB, Kashlan OB, Watkins JN, Suaud L, Yan W, Kleyman TR, and Rubenstein RC
- Subjects
- Animals, Cell Communication physiology, Cell Membrane physiology, Cloning, Molecular, Electrophysiology, Epithelial Sodium Channels, Female, Humans, Membrane Potentials, Mice, Transfection, Xenopus, Gene Expression Regulation, HSC70 Heat-Shock Proteins physiology, HSP70 Heat-Shock Proteins physiology, Oocytes physiology, Sodium Channels genetics
- Abstract
The members of the cytoplasmic 70-kDa heat shock protein family are involved in appropriate folding and trafficking of newly synthesized proteins in the cell. Hsc70, which is expressed constitutively, and Hsp70, the expression of which is stress- and heat shock-induced, are often considered to have similar cellular functions in this regard, but there are suggestions that the intracellular functions of these homologous but not identical proteins may differ. We tested the hypothesis that Hsc70 and Hsp70 would have differential effects on the expression of the epithelial sodium channel (ENaC). In Xenopus oocytes, overexpression of human Hsc70 decreased the functional (defined as amiloride-sensitive whole-oocyte current) and surface expression of murine ENaC (mENaC) in a concentration-dependent fashion. In contrast, coinjection of a moderate amount of Hsp70 cRNA (10 ng) increased the functional and surface expression of mENaC, whereas a higher amount of coinjected Hsp70 cRNA (30 ng) decreased mENaC functional and surface expression. The increase in mENaC functional expression with coinjection of 10 ng of Hsp70 cRNA was antagonized by the additional coinjection of Hsc70 cRNA in a concentration-dependent fashion. These data are consistent with Hsc70 and Hsp70 having differential and antagonistic effects with regard to the intracellular trafficking of mENaC in oocytes, which may have an impact on our understanding and potential treatment of diseases of aberrant ion channel trafficking.
- Published
- 2006
- Full Text
- View/download PDF
45. On the interaction between amiloride and its putative alpha-subunit epithelial Na+ channel binding site.
- Author
-
Kashlan OB, Sheng S, and Kleyman TR
- Subjects
- Amiloride analogs & derivatives, Animals, Binding Sites, Diuretics pharmacology, Dose-Response Relationship, Drug, Epithelial Sodium Channels, Glycine chemistry, Histidine chemistry, Hydrogen-Ion Concentration, Ions, Kinetics, Models, Chemical, Models, Molecular, Mutagenesis, Site-Directed, Mutation, Oocytes metabolism, Patch-Clamp Techniques, Protein Binding, Protein Conformation, Protein Structure, Quaternary, Protein Structure, Tertiary, RNA, Complementary metabolism, Serine chemistry, Sodium chemistry, Xenopus laevis, Amiloride chemistry, Amiloride pharmacology, Sodium Channels chemistry
- Abstract
The epithelial Na+ channel (ENaC) belongs to the structurally conserved ENaC/Degenerin superfamily. These channels are blocked by amiloride and its analogues. Several amino acid residues have been implicated in amiloride binding. Primary among these are alphaSer-583, betaGly-525, and gammaGly-542, which are present at a homologous site within the three subunits of ENaC. Mutations of the beta and gamma glycines greatly weakened amiloride block, but, surprisingly, mutation of the serine of the alpha subunit resulted in moderate (<5-fold) weakening of amiloride K(i). We investigated the role of alphaSer-583 in amiloride binding by systematically mutating alphaSer-583 and analyzing the mutant channels with two-electrode voltage clamp. We observed that most mutations had moderate effects on amiloride block, whereas those introducing rings showed dramatic effects on amiloride block. In addition, mutations introducing a beta-methyl group at this site altered the electric field of ENaC, affecting both amiloride binding and the voltage dependence of channel gating. We also found that the His mutation, in addition to greatly weakening amiloride binding, appends a voltage-sensitive gate within the pore of ENaC at low pH. Because diverse residues at alpha583, such as Asn, Gln, Ser, Gly, Thr, and Ala, have similar amiloride binding affinities, our results suggest that the wild type Ser side chain is not important for amiloride binding. However, given that some alphaSer-583 mutations affect the electrical properties of the channel whereas those introducing rings greatly weaken amiloride block, we conclude that amiloride binds at or near this site and that alphaSer-583 may have a role in ion permeation through ENaC.
- Published
- 2005
- Full Text
- View/download PDF
46. Rational polynomial representation of ribonucleotide reductase activity.
- Author
-
Radivoyevitch T, Kashlan OB, and Cooperman BS
- Subjects
- Enzyme Activation physiology, Least-Squares Analysis, Models, Biological, Models, Statistical, Ribonucleotide Reductases metabolism
- Abstract
Background: Recent data suggest that ribonucleotide reductase (RNR) exists not only as a heterodimer R12R22 of R12 and R22 homodimers, but also as tetramers R14R24 and hexamers R16R26. Recent data also suggest that ATP binds the R1 subunit at a previously undescribed hexamerization site, in addition to its binding to previously described dimerization and tetramerization sites. Thus, the current view is that R1 has four NDP substrate binding possibilities, four dimerization site binding possibilities (dATP, ATP, dGTP, or dTTP), two tetramerization site binding possibilities (dATP or ATP), and one hexamerization site binding possibility (ATP), in addition to possibilities of unbound site states. This large number of internal R1 states implies an even larger number of quaternary states. A mathematical model of RNR activity which explicitly represents the states of R1 currently exists, but it is complicated in several ways: (1) it includes up to six-fold nested sums; (2) it uses different mathematical structures under different substrate-modulator conditions; and (3) it requires root solutions of high order polynomials to determine R1 proportions in mono-, di-, tetra- and hexamer states and thus RNR activity as a function of modulator and total R1 concentrations., Results: We present four (one for each NDP) rational polynomial models of RNR activity as a function of substrate and reaction rate modifier concentrations. The new models avoid the complications of the earlier model without compromising curve fits to recent data., Conclusion: Compared to the earlier model of recent data, the new rational polynomial models are simpler, adequately fitting, and likely better suited for biochemical network simulations.
- Published
- 2005
- Full Text
- View/download PDF
47. Side chain orientation of residues lining the selectivity filter of epithelial Na+ channels.
- Author
-
Sheng S, Perry CJ, Kashlan OB, and Kleyman TR
- Subjects
- Alanine chemistry, Amiloride pharmacology, Amino Acid Sequence, Animals, Binding Sites, Cations, Cysteine chemistry, Dithiothreitol pharmacology, Dose-Response Relationship, Drug, Electrophysiology, Epithelial Sodium Channels, Mice, Models, Biological, Models, Molecular, Molecular Sequence Data, Mutagenesis, Site-Directed, Mutation, Oocytes metabolism, Protein Binding, Protein Structure, Tertiary, RNA, Complementary metabolism, Sequence Homology, Amino Acid, Serine chemistry, Time Factors, Xenopus, Cadmium chemistry, Sodium Channels chemistry
- Abstract
Epithelial Na(+) channels (ENaCs) selectively conduct Na(+) and Li(+) but exclude K(+). A three-residue tract ((G/S)XS) present within all three subunits has been identified as a key structure forming a putative selectivity filter. We investigated the side chain orientation of residues within this tract by analyzing accessibility of the introduced sulfhydryl groups to thiophilic Cd(2+). Xenopus oocytes were used to express wild-type or mutant mouse alphabetagammaENaCs. The blocking effect of external Cd(2+) was examined by comparing amiloride-sensitive Na(+) currents measured by two-electrode voltage clamp in the absence and presence of Cd(2+) in the bath solution. The currents in mutant channels containing a single Cys substitution at the first or third position within the (G/S)XS tract (alphaG587C, alphaS589C, betaG529C, betaS531C, gammaS546C, and gammaS548C) were blocked by Cd(2+) with varying inhibitory constants (0.06-13 mm), whereas the currents in control channels were largely insensitive to Cd(2+) at concentrations up to 10 mm. The Cd(2+) blocking effects were fast, with time constants in the range of seconds, and were only partially reversible. The blocked currents were restored by 10 mm dithiothreitol. Mutant channels containing alanine or serine substitutions at these sites within the alpha subunit were only poorly and reversibly blocked by 10 mm Cd(2+). These results indicate that the introduced sulfhydryl groups face the conduction pore and suggest that serine hydroxyl groups within the selectivity filter in wild-type ENaCs face the conduction pore and may contribute to cation selectivity by participating in coordination of permeating cations.
- Published
- 2005
- Full Text
- View/download PDF
48. The enantioselectivities of the active and allosteric sites of mammalian ribonucleotide reductase.
- Author
-
He J, Roy B, Périgaud C, Kashlan OB, and Cooperman BS
- Subjects
- Adenosine Diphosphate chemistry, Adenosine Diphosphate metabolism, Adenosine Triphosphate chemistry, Allosteric Site, Animals, Binding Sites, Cytidine Diphosphate chemistry, Cytidine Diphosphate metabolism, Mice, Protein Binding, Protein Subunits metabolism, Recombinant Proteins chemistry, Recombinant Proteins isolation & purification, Recombinant Proteins metabolism, Ribonucleoside Diphosphate Reductase metabolism, Substrate Specificity genetics, Adenosine Triphosphate metabolism, Allosteric Regulation, Protein Subunits chemistry, Ribonucleoside Diphosphate Reductase chemistry
- Abstract
Here we examine the enantioselectivity of the allosteric and substrate binding sites of murine ribonucleotide reductase (mRR). L-ADP binds to the active site and L-ATP binds to both the s- and a-allosteric sites of mR1 with affinities that are only three- to 10-fold weaker than the values for the corresponding D-enantiomers. These results demonstrate the potential of L-nucleotides for interacting with and modulating the activity of mRR, a cancer chemotherapeutic and antiviral target. On the other hand, we detect no substrate activity for L-ADP and no inhibitory activity for N3-L-dUDP, demonstrating the greater stereochemical stringency at the active site with respect to catalytic activity.
- Published
- 2005
- Full Text
- View/download PDF
49. Mechanisms of action of peptide inhibitors of mammalian ribonucleotide reductase targeting quaternary structure.
- Author
-
Gao Y, Kashlan OB, Kaur J, Tan C, and Cooperman BS
- Subjects
- Animals, Dimerization, Kinetics, Mammals, Models, Theoretical, Nucleic Acid Conformation, Oligopeptides chemistry, Oligopeptides pharmacology, Ribonucleotide Reductases chemistry, Scattering, Radiation, Enzyme Inhibitors pharmacology, Peptides chemistry, Peptides pharmacology, Ribonucleotide Reductases antagonists & inhibitors
- Abstract
Mammalian ribonucleotide reductase (mRR) is a chemotherapeutic target. The enzyme is composed of 2 subunits (mR1 and mR2) and is inhibited by Ac-FTLDADF (denoted P7), corresponding to the C-terminus of mR2, which competes with mR2 for binding to mR1. mRR has 2 physiologically important active forms, mR12mR22 and mR16(mR22)j (j = 1-3). Here we report on the mechanism of action of recently identified peptide derivatives having higher activities than P7 toward inhibition of one or both active forms. A significant feature of both P7 and these new inhibitors is that they are more potent vs. mR12mR22 than mR16(mR22)j. For some of these peptides, this is due in part to their preferential binding to the mR1 monomer. The possible application of these peptide derivatives in cancer chemotherapy is discussed., (2004 Wiley Periodicals, Inc.)
- Published
- 2005
- Full Text
- View/download PDF
50. Peptide inhibitors of mammalian ribonucleotide reductase.
- Author
-
Cooperman BS, Gao Y, Tan C, Kashlan OB, and Kaur J
- Subjects
- Allosteric Regulation physiology, Animals, Enzyme Inhibitors isolation & purification, Enzyme Inhibitors pharmacology, Mice, Models, Molecular, Oligopeptides isolation & purification, Peptide Library, Ribonucleotide Reductases pharmacology, Oligopeptides pharmacology, Peptide Fragments pharmacology, Ribonucleotide Reductases antagonists & inhibitors
- Abstract
Mammalian ribonucleotide reductase (mRR) is a chemotherapeutic target. In common with other class Ia RRs, the enzyme is composed of two subunits (mR1 and mR2), with mR1 containing both the active site and allosteric effector sites and mR2 containing a stable tyrosyl radical that is essential for enzymatic activity. mRR is inhibited by Ac-FTLDADF (denoted P7), corresponding to the C-terminus of mR2, which competes with mR2 for binding to mR1. The enzyme has two physiologically important active forms, mR12mR22 and mR16(mR22)j (j=1-3), with high ATP concentrations favoring the latter. Here, we report on our progress in using structural and functional studies in conjunction with library screening to identify derivatives of tri-, tetra- and hexapeptides, and cyclic heptapeptides, having equal or significantly higher activities than P7 toward inhibition of one or both active forms. These identifications were made by screening candidate peptides both for their abilities to bind to mR1 competitively with P7 and to inhibit ribonucleotide reductase activity. A significant feature of both P7 and the newly identified derivatives is that they are stronger inhibitors of mR12mR22 than of mR16(mR22)j. For the tetrapeptides, this is due in part to their preferential binding to mR1 monomer. The possible application of these peptide derivatives in cancer chemotherapy, exploiting their preferential inhibition of mR12mR22, is considered.
- Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.