1
|
A cytoplasmic Slo3 isoform is expressed in somatic tissues. Mol Biol Rep 2019; 46:5561-5567. [PMID: 31270758 DOI: 10.1007/s11033-019-04943-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 06/26/2019] [Indexed: 12/19/2022]
Abstract
Slo3 is a pH-sensitive and weakly voltage-sensitive potassium channel that is essential for male fertility in mouse and whose expression is regarded as sperm-specific. These properties have proposed Slo3 as a candidate target for male contraceptive drugs. Nonetheless, the tissue distribution of Slo3 expression has not been rigorously studied yet. Applying computational and RT-PCR approaches, we identified expression of two short Slo3 isoforms in somatic mouse tissues such as brain, kidney and eye. These isoforms, which seem to result of transcription starting sites between exons 20 and 21, have an identical open reading frame, both encoding the terminal 381 amino acids of the cytosolic Slo3 domain. We corroborated the expression of these isoforms in mouse brain and testis by Western-blot. The complete isoform encoding the Slo3 ion channel was uniquely detected in testis, both at transcript and protein level. Although the functional role of the cytosolic Slo3 isoforms remains to be established, we propose that they may have a functional effect by modulating Slo channels trafficking and/or activity. This study confirms that expression of full-length Slo3 is sperm-specific but warns against developing contraceptive drugs targeting the C-terminal tail of Slo3 channels.
Collapse
|
2
|
Rajendran VM, Sandle GI. Colonic Potassium Absorption and Secretion in Health and Disease. Compr Physiol 2018; 8:1513-1536. [PMID: 30215859 PMCID: PMC9769410 DOI: 10.1002/cphy.c170030] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The colon has large capacities for K+ absorption and K+ secretion, but its role in maintaining K+ homeostasis is often overlooked. For many years, passive diffusion and/or solvent drag were thought to be the primary mechanisms for K+ absorption in human and animal colon. However, it is now clear that apical H+ ,K+ -ATPase, in coordination with basolateral K+ -Cl- cotransport and/or K+ and Cl- channels operating in parallel, mediate electroneutral K+ absorption in animal colon. We now know that K+ absorption in rat colon reflects ouabain-sensitive and ouabain-insensitive apical H+ ,K+ -ATPase activities. Ouabain-insensitive and ouabain-sensitive H+ ,K+ -ATPases are localized in surface and crypt cells, respectively. Colonic H+ ,K+ -ATPase consists of α- (HKCα ) and β- (HKCβ ) subunits which, when coexpressed, exhibit ouabain-insensitive H+ ,K+ -ATPase activity in HEK293 cells, while HKCα coexpressed with the gastric β-subunit exhibits ouabain-sensitive H+ ,K+ -ATPase activity in Xenopus oocytes. Aldosterone enhances apical H+ ,K+ -ATPase activity, HKCα specific mRNA and protein expression, and K+ absorption. Active K+ secretion, on the other hand, is mediated by apical K+ channels operating in a coordinated way with the basolateral Na+ -K+ -2Cl- cotransporter. Both Ca2+ -activated intermediate conductance K+ (IK) and large conductance K+ (BK) channels are located in the apical membrane of colonic epithelia. IK channel-mediated K+ efflux provides the driving force for Cl- secretion, while BK channels mediate active (e.g., cAMP-activated) K+ secretion. BK channel expression and activity are increased in patients with end-stage renal disease and ulcerative colitis. This review summarizes the role of apical H+ ,K+ -ATPase in K+ absorption, and apical BK channel function in K+ secretion in health and disease. © 2018 American Physiological Society. Compr Physiol 8:1513-1536, 2018.
Collapse
Affiliation(s)
| | - Geoffrey I. Sandle
- Leeds Institute of Biomedical and Clinical Sciences, St James’s University Hospital, Leeds LS9 7TF, UK
| |
Collapse
|
3
|
The Slo(w) path to identifying the mitochondrial channels responsible for ischemic protection. Biochem J 2017; 474:2067-2094. [PMID: 28600454 DOI: 10.1042/bcj20160623] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/10/2017] [Accepted: 02/13/2017] [Indexed: 12/19/2022]
Abstract
Mitochondria play an important role in tissue ischemia and reperfusion (IR) injury, with energetic failure and the opening of the mitochondrial permeability transition pore being the major causes of IR-induced cell death. Thus, mitochondria are an appropriate focus for strategies to protect against IR injury. Two widely studied paradigms of IR protection, particularly in the field of cardiac IR, are ischemic preconditioning (IPC) and volatile anesthetic preconditioning (APC). While the molecular mechanisms recruited by these protective paradigms are not fully elucidated, a commonality is the involvement of mitochondrial K+ channel opening. In the case of IPC, research has focused on a mitochondrial ATP-sensitive K+ channel (mitoKATP), but, despite recent progress, the molecular identity of this channel remains a subject of contention. In the case of APC, early research suggested the existence of a mitochondrial large-conductance K+ (BK, big conductance of potassium) channel encoded by the Kcnma1 gene, although more recent work has shown that the channel that underlies APC is in fact encoded by Kcnt2 In this review, we discuss both the pharmacologic and genetic evidence for the existence and identity of mitochondrial K+ channels, and the role of these channels both in IR protection and in regulating normal mitochondrial function.
Collapse
|
4
|
Maqoud F, Cetrone M, Mele A, Tricarico D. Molecular structure and function of big calcium-activated potassium channels in skeletal muscle: pharmacological perspectives. Physiol Genomics 2017; 49:306-317. [DOI: 10.1152/physiolgenomics.00121.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 02/08/2017] [Accepted: 04/10/2017] [Indexed: 11/22/2022] Open
Abstract
The large-conductance Ca2+-activated K+ (BK) channel is broadly expressed in various mammalian cells and tissues such as neurons, skeletal muscles (sarco-BK), and smooth muscles. These channels are activated by changes in membrane electrical potential and by increases in the concentration of intracellular calcium ion (Ca2+). The BK channel is subjected to many mechanisms that add diversity to the BK channel α-subunit gene. These channels are indeed subject to alternative splicing, auxiliary subunits modulation, posttranslational modifications, and protein-protein interactions. BK channels can be modulated by diverse molecules that may induce either an increase or decrease in channel activity. The linkage of these channels to many intracellular metabolites and pathways, as well as their modulation by extracellular natural agents, have been found to be relevant in many physiological processes. BK channel diversity is obtained by means of alternative splicing and modulatory β- and γ-subunits. The association of the α-subunit with β- or with γ-subunits can change the BK channel phenotype, functional diversity, and pharmacological properties in different tissues. In the case of the skeletal muscle BK channel (sarco-BK channel), we established that the main mechanism regulating BK channel diversity is the alternative splicing of the KCNMA1/slo1 gene encoding for the α-subunit generating different splicing isoform in the muscle phenotypes. This finding helps to design molecules selectively targeting the skeletal muscle subtypes. The use of drugs selectively targeting the skeletal muscle BK channels is a promising strategy in the treatment of familial disorders affecting muscular skeletal apparatus including hyperkalemia and hypokalemia periodic paralysis.
Collapse
Affiliation(s)
- Fatima Maqoud
- Department of Pharmacy-Drug Science, University of Bari, Bari, Italy
- Faculty of Science, Chouaib Doukkali University, El Jadida, Morocco
| | - Michela Cetrone
- Istituto Tumori Giovanni Paolo II, Istituto di Ricovero e Cura a Carattere Scientifico, National Cancer Institute, Bari, Italy; and
| | - Antonietta Mele
- Department of Pharmacy-Drug Science, University of Bari, Bari, Italy
| | | |
Collapse
|
5
|
Latorre R, Castillo K, Carrasquel-Ursulaez W, Sepulveda RV, Gonzalez-Nilo F, Gonzalez C, Alvarez O. Molecular Determinants of BK Channel Functional Diversity and Functioning. Physiol Rev 2017; 97:39-87. [DOI: 10.1152/physrev.00001.2016] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Large-conductance Ca2+- and voltage-activated K+ (BK) channels play many physiological roles ranging from the maintenance of smooth muscle tone to hearing and neurosecretion. BK channels are tetramers in which the pore-forming α subunit is coded by a single gene ( Slowpoke, KCNMA1). In this review, we first highlight the physiological importance of this ubiquitous channel, emphasizing the role that BK channels play in different channelopathies. We next discuss the modular nature of BK channel-forming protein, in which the different modules (the voltage sensor and the Ca2+ binding sites) communicate with the pore gates allosterically. In this regard, we review in detail the allosteric models proposed to explain channel activation and how the models are related to channel structure. Considering their extremely large conductance and unique selectivity to K+, we also offer an account of how these two apparently paradoxical characteristics can be understood consistently in unison, and what we have learned about the conduction system and the activation gates using ions, blockers, and toxins. Attention is paid here to the molecular nature of the voltage sensor and the Ca2+ binding sites that are located in a gating ring of known crystal structure and constituted by four COOH termini. Despite the fact that BK channels are coded by a single gene, diversity is obtained by means of alternative splicing and modulatory β and γ subunits. We finish this review by describing how the association of the α subunit with β or with γ subunits can change the BK channel phenotype and pharmacology.
Collapse
Affiliation(s)
- Ramon Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso and Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Universidad Andres Bello, Facultad de Ciencias Biologicas, Center for Bioinformatics and Integrative Biology, Avenida Republica 239, Santiago, Chile and Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Karen Castillo
- Centro Interdisciplinario de Neurociencia de Valparaíso and Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Universidad Andres Bello, Facultad de Ciencias Biologicas, Center for Bioinformatics and Integrative Biology, Avenida Republica 239, Santiago, Chile and Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Willy Carrasquel-Ursulaez
- Centro Interdisciplinario de Neurociencia de Valparaíso and Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Universidad Andres Bello, Facultad de Ciencias Biologicas, Center for Bioinformatics and Integrative Biology, Avenida Republica 239, Santiago, Chile and Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Romina V. Sepulveda
- Centro Interdisciplinario de Neurociencia de Valparaíso and Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Universidad Andres Bello, Facultad de Ciencias Biologicas, Center for Bioinformatics and Integrative Biology, Avenida Republica 239, Santiago, Chile and Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Fernando Gonzalez-Nilo
- Centro Interdisciplinario de Neurociencia de Valparaíso and Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Universidad Andres Bello, Facultad de Ciencias Biologicas, Center for Bioinformatics and Integrative Biology, Avenida Republica 239, Santiago, Chile and Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Carlos Gonzalez
- Centro Interdisciplinario de Neurociencia de Valparaíso and Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Universidad Andres Bello, Facultad de Ciencias Biologicas, Center for Bioinformatics and Integrative Biology, Avenida Republica 239, Santiago, Chile and Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Osvaldo Alvarez
- Centro Interdisciplinario de Neurociencia de Valparaíso and Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Universidad Andres Bello, Facultad de Ciencias Biologicas, Center for Bioinformatics and Integrative Biology, Avenida Republica 239, Santiago, Chile and Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| |
Collapse
|
6
|
Kwon SH, Oh S, Nacke M, Mostov KE, Lipschutz JH. Adaptor Protein CD2AP and L-type Lectin LMAN2 Regulate Exosome Cargo Protein Trafficking through the Golgi Complex. J Biol Chem 2016; 291:25462-25475. [PMID: 27765817 DOI: 10.1074/jbc.m116.729202] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 09/27/2016] [Indexed: 01/30/2023] Open
Abstract
Exosomes, 40-150-nm extracellular vesicles, transport biological macromolecules that mediate intercellular communications. Although exosomes are known to originate from maturation of endosomes into multivesicular endosomes (also known as multivesicular bodies) with subsequent fusion of the multivesicular endosomes with the plasma membrane, it remains unclear how cargos are selected for exosomal release. Using an inducible expression system for the exosome cargo protein GPRC5B and following its trafficking trajectory, we show here that newly synthesized GPRC5B protein accumulates in the Golgi complex prior to its release into exosomes. The L-type lectin LMAN2 (also known as VIP36) appears to be specifically required for the accumulation of GPRC5B in the Golgi complex and restriction of GPRC5B transport along the exosomal pathway. This may occur due to interference with the adaptor protein GGA1-mediated trans Golgi network-to-endosome transport of GPRC5B. The adaptor protein CD2AP-mediated internalization following cell surface delivery appears to contribute to the Golgi accumulation of GPRC5B, possibly in parallel with biosynthetic/secretory trafficking from the endoplasmic reticulum. Our data thus reveal a Golgi-traversing pathway for exosomal release of the cargo protein GPRC5B in which CD2AP facilitates the entry and LMAN2 impedes the exit of the flux, respectively.
Collapse
Affiliation(s)
- Sang-Ho Kwon
- From the Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, South Carolina 29425,
| | - Sekyung Oh
- the Department of Radiology, Stanford Cardiovascular Institute, and.,Department of Neurology and Neurological Sciences,Stanford University School of Medicine, Stanford, California 94305
| | - Marisa Nacke
- the Departments of Anatomy and Biochemistry/Biophysics, University of California, San Francisco, California 94143, and
| | - Keith E Mostov
- the Departments of Anatomy and Biochemistry/Biophysics, University of California, San Francisco, California 94143, and
| | - Joshua H Lipschutz
- From the Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, South Carolina 29425.,the Department of Medicine, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29401
| |
Collapse
|
7
|
Suzuki Y, Ohya S, Yamamura H, Giles WR, Imaizumi Y. A New Splice Variant of Large Conductance Ca2+-activated K+ (BK) Channel α Subunit Alters Human Chondrocyte Function. J Biol Chem 2016; 291:24247-24260. [PMID: 27758860 DOI: 10.1074/jbc.m116.743302] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 10/06/2016] [Indexed: 02/06/2023] Open
Abstract
Large conductance Ca2+-activated K+ (BK) channels play essential roles in both excitable and non-excitable cells. For example, in chondrocytes, agonist-induced Ca2+ release from intracellular store activates BK channels, and this hyperpolarizes these cells, augments Ca2+ entry, and forms a positive feed-back mechanism for Ca2+ signaling and stimulation-secretion coupling. In the present study, functional roles of a newly identified splice variant in the BK channel α subunit (BKαΔe2) were examined in a human chondrocyte cell line, OUMS-27, and in a HEK293 expression system. Although BKαΔe2 lacks exon2, which codes the intracellular S0-S1 linker (Glu-127-Leu-180), significant expression was detected in several tissues from humans and mice. Molecular image analyses revealed that BKαΔe2 channels are not expressed on plasma membrane but can traffic to the plasma membrane after forming hetero-tetramer units with wild-type BKα (BKαWT). Single-channel current analyses demonstrated that BKα hetero-tetramers containing one, two, or three BKαΔe2 subunits are functional. These hetero-tetramers have a smaller single channel conductance and exhibit lower trafficking efficiency than BKαWT homo-tetramers in a stoichiometry-dependent manner. Site-directed mutagenesis of residues in exon2 identified Helix2 and the linker to S1 (Trp-158-Leu-180, particularly Arg-178) as an essential segment for channel function including voltage dependence and trafficking. BKαΔe2 knockdown in OUMS-27 chondrocytes increased BK current density and augmented the responsiveness to histamine assayed as cyclooxygenase-2 gene expression. These findings provide significant new evidence that BKαΔe2 can modulate cellular responses to physiological stimuli in human chondrocyte and contribute under pathophysiological conditions, such as osteoarthritis.
Collapse
Affiliation(s)
- Yoshiaki Suzuki
- From the Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabedori, Mizuhoku, Nagoya 467-8603, Japan
| | - Susumu Ohya
- From the Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabedori, Mizuhoku, Nagoya 467-8603, Japan.,the Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan, and
| | - Hisao Yamamura
- From the Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabedori, Mizuhoku, Nagoya 467-8603, Japan
| | - Wayne R Giles
- the Faculties of Kinesiology and Medicine, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Yuji Imaizumi
- From the Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabedori, Mizuhoku, Nagoya 467-8603, Japan,
| |
Collapse
|
8
|
Carrisoza-Gaytan R, Carattino MD, Kleyman TR, Satlin LM. An unexpected journey: conceptual evolution of mechanoregulated potassium transport in the distal nephron. Am J Physiol Cell Physiol 2015; 310:C243-59. [PMID: 26632600 DOI: 10.1152/ajpcell.00328.2015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Flow-induced K secretion (FIKS) in the aldosterone-sensitive distal nephron (ASDN) is mediated by large-conductance, Ca(2+)/stretch-activated BK channels composed of pore-forming α-subunits (BKα) and accessory β-subunits. This channel also plays a critical role in the renal adaptation to dietary K loading. Within the ASDN, the cortical collecting duct (CCD) is a major site for the final renal regulation of K homeostasis. Principal cells in the ASDN possess a single apical cilium whereas the surfaces of adjacent intercalated cells, devoid of cilia, are decorated with abundant microvilli and microplicae. Increases in tubular (urinary) flow rate, induced by volume expansion, diuretics, or a high K diet, subject CCD cells to hydrodynamic forces (fluid shear stress, circumferential stretch, and drag/torque on apical cilia and presumably microvilli/microplicae) that are transduced into increases in principal (PC) and intercalated (IC) cell cytoplasmic Ca(2+) concentration that activate apical voltage-, stretch- and Ca(2+)-activated BK channels, which mediate FIKS. This review summarizes studies by ourselves and others that have led to the evolving picture that the BK channel is localized in a macromolecular complex at the apical membrane, composed of mechanosensitive apical Ca(2+) channels and a variety of kinases/phosphatases as well as other signaling molecules anchored to the cytoskeleton, and that an increase in tubular fluid flow rate leads to IC- and PC-specific responses determined, in large part, by the cell-specific composition of the BK channels.
Collapse
Affiliation(s)
| | - Marcelo D Carattino
- Renal-Electrolyte Division, Department of Medicine, Pittsburgh, Pennsylvania
| | - Thomas R Kleyman
- Renal-Electrolyte Division, Department of Medicine, Pittsburgh, Pennsylvania
| | - Lisa M Satlin
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York; and
| |
Collapse
|
9
|
Toro L, Li M, Zhang Z, Singh H, Wu Y, Stefani E. MaxiK channel and cell signalling. Pflugers Arch 2014; 466:875-86. [PMID: 24077696 DOI: 10.1007/s00424-013-1359-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 09/09/2013] [Accepted: 09/10/2013] [Indexed: 01/23/2023]
Abstract
The large-conductance Ca2+- and voltage-activated K+ (MaxiK, BK, BKCa, Slo1, KCa1.1) channel role in cell signalling is becoming apparent as we learn how the channel interacts with a multiplicity of proteins not only at the plasma membrane but also in intracellular organelles including the endoplasmic reticulum, nucleus, and mitochondria. In this review, we focus on the interactions of MaxiK channels with seven-transmembrane G protein-coupled receptors and discuss information suggesting that, the channel big C-terminus may act as the nucleus of signalling molecules including kinases relevant for cell death and survival. Increasing evidence indicates that the channel is able to associate with a variety of receptors including β-adrenergic receptors, G protein-coupled estrogen receptors, acetylcholine receptors, thromboxane A2 receptors, and angiotensin II receptors, which highlights the varied functions that the channel has (or may have) not only in regulating contraction/relaxation of muscle cells or neurotransmission in the brain but also in cell metabolism, proliferation, migration, and gene expression. In line with this view, MaxiK channels have been implicated in obesity and in brain, prostate, and mammary cancers. A better understanding on the molecular mechanisms underlying or triggered by MaxiK channel abnormalities like overexpression in certain cancers may lead to new therapeutics to prevent devastating diseases.
Collapse
|
10
|
Liu Y, Song X, Shi Y, Shi Z, Niu W, Feng X, Gu D, Bao HF, Ma HP, Eaton DC, Zhuang J, Cai H. WNK1 activates large-conductance Ca2+-activated K+ channels through modulation of ERK1/2 signaling. J Am Soc Nephrol 2014; 26:844-54. [PMID: 25145935 DOI: 10.1681/asn.2014020186] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
With no lysine (WNK) kinases are members of the serine/threonine kinase family. We previously showed that WNK4 inhibits renal large-conductance Ca(2+)-activated K(+) (BK) channel activity by enhancing its degradation through a lysosomal pathway. In this study, we investigated the effect of WNK1 on BK channel activity. In HEK293 cells stably expressing the α subunit of BK (HEK-BKα cells), siRNA-mediated knockdown of WNK1 expression significantly inhibited both BKα channel activity and open probability. Knockdown of WNK1 expression also significantly inhibited BKα protein expression and increased ERK1/2 phosphorylation, whereas overexpression of WNK1 significantly enhanced BKα expression and decreased ERK1/2 phosphorylation in a dose-dependent manner in HEK293 cells. Knockdown of ERK1/2 prevented WNK1 siRNA-mediated inhibition of BKα expression. Similarly, pretreatment of HEK-BKα cells with the lysosomal inhibitor bafilomycin A1 reversed the inhibitory effects of WNK1 siRNA on BKα expression in a dose-dependent manner. Knockdown of WNK1 expression also increased the ubiquitination of BKα channels. Notably, mice fed a high-K(+) diet for 10 days had significantly higher renal protein expression levels of BKα and WNK1 and lower levels of ERK1/2 phosphorylation compared with mice fed a normal-K(+) diet. These data suggest that WNK1 enhances BK channel function by reducing ERK1/2 signaling-mediated lysosomal degradation of the channel.
Collapse
Affiliation(s)
- Yingli Liu
- Renal Division, Department of Medicine, and Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine
| | - Xiang Song
- Department of Cardiology, The Fourth Affiliated Hospital, Harbin Medical University, Heilongjiang, China; and
| | | | - Zhen Shi
- Department of Nephrology, The Second Affiliated Hospital, Wenzhou Medical University, Zhejiang, China
| | - Weihui Niu
- Department of Nephrology, The Second Affiliated Hospital, Wenzhou Medical University, Zhejiang, China
| | - Xiuyan Feng
- Renal Division, Department of Medicine, and Renal Section, Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - Dingying Gu
- Department of Nephrology, The Second Affiliated Hospital, Wenzhou Medical University, Zhejiang, China
| | - Hui-Fang Bao
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| | - He-Ping Ma
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| | - Douglas C Eaton
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| | - Jieqiu Zhuang
- Department of Nephrology, The Second Affiliated Hospital, Wenzhou Medical University, Zhejiang, China;
| | - Hui Cai
- Renal Division, Department of Medicine, and Renal Section, Atlanta Veterans Affairs Medical Center, Decatur, Georgia Department of Physiology, Emory University School of Medicine, Atlanta, Georgia;
| |
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW Potassium channels in the distal nephron are precisely controlled to regulate potassium secretion in accord with physiological demands. In recent years, it has become evident that membrane trafficking processes play a fundamental role. This short review highlights recent developments in elucidating the underlying mechanisms. RECENT FINDINGS Novel sorting signals in the renal potassium channels, and the elusive intracellular trafficking machinery that read and act on these signals have recently been identified. These new discoveries reveal that independent signals sequentially interact with different intracellular sorting, retention and internalization machineries to appropriately ferry the channels to and from the apical and basolateral membrane domains in sufficient numbers to regulate potassium balance. SUMMARY A new understanding of the basic mechanisms that control potassium channel density at polarized membrane domains has emerged, providing new insights into how potassium balance is achieved and how it goes awry in disease.
Collapse
Affiliation(s)
- Paul A Welling
- Department of Physiology, University of Maryland Medical School, Baltimore, Maryland 21201, USA.
| |
Collapse
|
12
|
|
13
|
Zhang J, Halm ST, Halm DR. Role of the BK channel (KCa1.1) during activation of electrogenic K+ secretion in guinea pig distal colon. Am J Physiol Gastrointest Liver Physiol 2012; 303:G1322-34. [PMID: 23064759 PMCID: PMC3532550 DOI: 10.1152/ajpgi.00325.2012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Secretagogues acting at a variety of receptor types activate electrogenic K(+) secretion in guinea pig distal colon, often accompanied by Cl(-) secretion. Distinct blockers of K(Ca)1.1 (BK, Kcnma1), iberiotoxin (IbTx), and paxilline inhibited the negative short-circuit current (I(sc)) associated with K(+) secretion. Mucosal addition of IbTx inhibited epinephrine-activated I(sc) ((epi)I(sc)) and transepithelial conductance ((epi)G(t)) consistent with K(+) secretion occurring via apical membrane K(Ca)1.1. The concentration dependence of IbTx inhibition of (epi)I(sc) yielded an IC(50) of 193 nM, with a maximal inhibition of 51%. Similarly, IbTx inhibited (epi)G(t) with an IC(50) of 220 nM and maximal inhibition of 48%. Mucosally added paxilline (10 μM) inhibited (epi)I(sc) and (epi)G(t) by ∼50%. IbTx and paxilline also inhibited I(sc) activated by mucosal ATP, supporting apical K(Ca)1.1 as a requirement for this K(+) secretagogue. Responses to IbTx and paxilline indicated that a component of K(+) secretion occurred during activation of Cl(-) secretion by prostaglandin-E(2) and cholinergic stimulation. Analysis of K(Ca)1.1α mRNA expression in distal colonic epithelial cells indicated the presence of the ZERO splice variant and three splice variants for the COOH terminus. The presence of the regulatory β-subunits K(Ca)β1 and K(Ca)β4 also was demonstrated. Immunolocalization supported the presence of K(Ca)1.1α in apical and basolateral membranes of surface and crypt cells. Together these results support a cellular mechanism for electrogenic K(+) secretion involving apical membrane K(Ca)1.1 during activation by several secretagogue types, but the observed K(+) secretion likely required the activity of additional K(+) channel types in the apical membrane.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Neuroscience, Cell Biology and Physiology, Wright State University Boonshoft School of Medicine, Dayton, Ohio
| | - Susan T. Halm
- Department of Neuroscience, Cell Biology and Physiology, Wright State University Boonshoft School of Medicine, Dayton, Ohio
| | - Dan R. Halm
- Department of Neuroscience, Cell Biology and Physiology, Wright State University Boonshoft School of Medicine, Dayton, Ohio
| |
Collapse
|
14
|
Abstract
The large conductance calcium- and voltage-activated potassium channel (BK(Ca)) is widely expressed at the plasma membrane. This channel is involved in a variety of fundamental cellular functions including excitability, smooth muscle contractility, and Ca(2+) homeostasis, as well as in pathological situations like proinflammatory responses in rheumatoid arthritis, and cancer cell proliferation. Immunochemical, biochemical and pharmacological studies from over a decade have intermittently shown the presence of BK(Ca) in intracellular organelles. To date, intracellular BK(Ca) (iBK(Ca)) has been localized in the mitochondria, endoplasmic reticulum, nucleus and Golgi apparatus but its functional role remains largely unknown except for the mitochondrial BK(Ca) whose opening is thought to play a role in protecting the heart from ischaemic injury. In the nucleus, pharmacology suggests a role in regulating nuclear Ca(2+), membrane potential and eNOS expression. Establishing the molecular correlates of iBK(Ca), the mechanisms defining iBK(Ca) organelle-specific targeting, and their modulation are challenging questions. This review summarizes iBK(Ca) channels, their possible functions, and efforts to identify their molecular correlates.
Collapse
Affiliation(s)
- Harpreet Singh
- Department of Anesthesiology, University of California, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
15
|
Dinardo MM, Camerino G, Mele A, Latorre R, Conte Camerino D, Tricarico D. Splicing of the rSlo gene affects the molecular composition and drug response of Ca2+-activated K+ channels in skeletal muscle. PLoS One 2012; 7:e40235. [PMID: 22808126 PMCID: PMC3393747 DOI: 10.1371/journal.pone.0040235] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 06/03/2012] [Indexed: 12/02/2022] Open
Abstract
The molecular composition and drug responses of calcium-activated K+ (BK) channels of skeletal muscle are unknown. Patch-clamp experiments combined with transcript scanning of the Kcnma1 gene encoding the alpha subunit of the BK channel were performed in rat slow-twitch soleus (Sol) and fast-twitch flexor digitorum brevis (FDB) skeletal muscles. Five splicing products of the Kcnma1 gene were isolated from Sol and FDB: the e17, e22, +29 aa, Slo27 and Slo0 variants. RT-PCR analysis demonstrated that the expression of e22 and Slo0 were 80–90% higher in FDB than Sol, whereas the expression of Slo27 was 60% higher in Sol than FDB, and the +29 aa variant was equally expressed in both muscle types. No beta 1-4 subunits were detected. In Sol, a large BK current with low Ca2+ sensitivity was recorded. The BK channel of Sol also showed a reduced response to BK channel openers, such as NS1619, acetazolamide and related drugs. In FDB, a reduced BK current with high Ca2+ sensitivity and an enhanced drug response was recorded. The total BK RNA content, which was 200% higher in Sol than in FDB, correlated with the BK currents in both muscles. Drug responses primarily correlated with e22 and Slo0 expression levels in FDB and to Slo27 expression in Sol muscle. In conclusion, phenotype-dependent BK channel biophysical and pharmacological properties correlated with the expression levels of the variants in muscles. These findings may be relevant to conditions affecting postural muscles, such as prolonged bed-rest, and to diseases affecting fast-twitch muscles, such as periodic paralysis. Down-regulation or up-regulation of the variants associated with pathological conditions may affect channel composition and drug responses.
Collapse
Affiliation(s)
| | - Giulia Camerino
- Departments of Pharmacobiology, Faculty of Pharmacy, University of Bari, Bari, Italy
| | - Antonietta Mele
- Departments of Pharmacobiology, Faculty of Pharmacy, University of Bari, Bari, Italy
| | - Ramon Latorre
- Centro Interdisciplinario de Neurociencias de Valparaíso, Facultad de Ciencias, Universidad de Valparaiso, Valparaiso, Chile
| | - Diana Conte Camerino
- Departments of Pharmacobiology, Faculty of Pharmacy, University of Bari, Bari, Italy
| | - Domenico Tricarico
- Departments of Pharmacobiology, Faculty of Pharmacy, University of Bari, Bari, Italy
- * E-mail:
| |
Collapse
|
16
|
Chen L, Jeffries O, Rowe ICM, Liang Z, Knaus HG, Ruth P, Shipston MJ. Membrane trafficking of large conductance calcium-activated potassium channels is regulated by alternative splicing of a transplantable, acidic trafficking motif in the RCK1-RCK2 linker. J Biol Chem 2010; 285:23265-75. [PMID: 20479001 PMCID: PMC2906319 DOI: 10.1074/jbc.m110.139758] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2010] [Indexed: 01/26/2023] Open
Abstract
Trafficking of the pore-forming alpha-subunits of large conductance calcium- and voltage-activated potassium (BK) channels to the cell surface represents an important regulatory step in controlling BK channel function. Here, we identify multiple trafficking signals within the intracellular RCK1-RCK2 linker of the cytosolic C terminus of the channel that are required for efficient cell surface expression of the channel. In particular, an acidic cluster-like motif was essential for channel exit from the endoplasmic reticulum and subsequent cell surface expression. This motif could be transplanted onto a heterologous nonchannel protein to enhance cell surface expression by accelerating endoplasmic reticulum export. Importantly, we identified a human alternatively spliced BK channel variant, hSloDelta(579-664), in which these trafficking signals are excluded because of in-frame exon skipping. The hSloDelta(579-664) variant is expressed in multiple human tissues and cannot form functional channels at the cell surface even though it retains the putative RCK domains and downstream trafficking signals. Functionally, the hSloDelta(579-664) variant acts as a dominant negative subunit to suppress cell surface expression of BK channels. Thus alternative splicing of the intracellular RCK1-RCK2 linker plays a critical role in determining cell surface expression of BK channels by controlling the inclusion/exclusion of multiple trafficking motifs.
Collapse
Affiliation(s)
- Lie Chen
- From the
Centre for Integrative Physiology, College of Medicine & Veterinary Medicine, University of Edinburgh, Edinburgh EH8 9XD, Scotland, United Kingdom
| | - Owen Jeffries
- From the
Centre for Integrative Physiology, College of Medicine & Veterinary Medicine, University of Edinburgh, Edinburgh EH8 9XD, Scotland, United Kingdom
| | - Iain C. M. Rowe
- From the
Centre for Integrative Physiology, College of Medicine & Veterinary Medicine, University of Edinburgh, Edinburgh EH8 9XD, Scotland, United Kingdom
| | - Zhi Liang
- From the
Centre for Integrative Physiology, College of Medicine & Veterinary Medicine, University of Edinburgh, Edinburgh EH8 9XD, Scotland, United Kingdom
| | - Hans-Guenther Knaus
- the
Division for Molecular and Cellular Pharmacology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University Innsbruck, Peter-Mayr Strasse 1, 6020 Innsbruck, Austria, and
| | - Peter Ruth
- Pharmacology and Toxicology, Institute of Pharmacy, University Tuebingen, 72076 Tuebingen, Germany
| | - Michael J. Shipston
- From the
Centre for Integrative Physiology, College of Medicine & Veterinary Medicine, University of Edinburgh, Edinburgh EH8 9XD, Scotland, United Kingdom
| |
Collapse
|
17
|
Errasti-Murugarren E, Casado FJ, Pastor-Anglada M. Different N-terminal motifs determine plasma membrane targeting of the human concentrative nucleoside transporter 3 in polarized and nonpolarized cells. Mol Pharmacol 2010; 78:795-803. [PMID: 20643903 DOI: 10.1124/mol.110.065920] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Human concentrative nucleoside transporter 3 (hCNT3) is a broad-selectivity, high-affinity protein implicated in the uptake of most nucleoside-derived anticancer and antiviral drugs. Regulated trafficking of hCNT3 has been recently postulated as a suitable way to improve nucleoside-based therapies. Moreover, the recent identification of a putative novel hCNT3-type transporter lacking the first 69 amino acids and retained at the endoplasmic reticulum anticipated that the N terminus of hCNT3 contains critical motifs implicated in trafficking. In the current study, we have addressed this issue by using deletions and site-directed mutagenesis and plasma membrane expression and nucleoside uptake kinetic analysis. Data reveal that 1) a segment between amino acids 50 and 62 contains plasma membrane-sorting determinants in nonpolarized cells; 2) in particular, the Val(57)-Thr(58)-Val(59) tripeptide seems to be the core of the export signal, whereas acidic motifs upstream and downstream of it seem to be important for the kinetics of the process; and 3) in polarized epithelia, the β-turn-forming motif (17)VGFQ(20) is necessary for proper apical expression of the protein.
Collapse
Affiliation(s)
- Ekaitz Errasti-Murugarren
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona and CIBER EHD, Avda Diagonal 645, Edifici annex, Planta-1, 08028 Barcelona, Spain
| | | | | |
Collapse
|
18
|
Highly specific alternative splicing of transcripts encoding BK channels in the chicken's cochlea is a minor determinant of the tonotopic gradient. Mol Cell Biol 2010; 30:3646-60. [PMID: 20479127 DOI: 10.1128/mcb.00073-10] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The frequency sensitivity of auditory hair cells in the inner ear varies with their longitudinal position in the sensory epithelium. Among the factors that determine the differential cellular response to sound is the resonance of a hair cell's transmembrane electrical potential, whose frequency correlates with the kinetic properties of the high-conductance Ca(2+)-activated K(+) (BK) channels encoded by a Slo (kcnma1) gene. It has been proposed that the inclusion of specific alternative axons in the Slo transcripts along the cochlea underlies the gradient of BK-channel kinetics. By analyzing the complete sequences of chicken Slo gene (cSlo) cDNAs from the chicken's cochlea, we show that most transcripts lack alternative exons. Transcripts with more than one alternative exon constitute only 10% of the total. Although the fraction of transcripts containing alternative exons increases from the cochlear base to the apex, the combination of alternative exons is not regulated. There is also a clear increase in the expression of BK transcripts with long carboxyl termini toward the apex. When long and short BK transcripts are expressed in HEK-293 cells, the kinetics of single-channel currents differ only slightly, but they are substantially slowed when the channels are coexpressed with the auxiliary beta subunit that occurs more widely at the apex. These results argue that the tonotopic gradient is not established by the selective inclusion of highly specific cSlo exons. Instead, a gradient in the expression of beta subunits slows BK channels toward the low-frequency apex of the cochlea.
Collapse
|
19
|
Manna PT, Smith AJ, Taneja TK, Howell GJ, Lippiat JD, Sivaprasadarao A. Constitutive endocytic recycling and protein kinase C-mediated lysosomal degradation control K(ATP) channel surface density. J Biol Chem 2010; 285:5963-73. [PMID: 20026601 PMCID: PMC2820821 DOI: 10.1074/jbc.m109.066902] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Revised: 12/07/2009] [Indexed: 11/06/2022] Open
Abstract
Pancreatic ATP-sensitive potassium (K(ATP)) channels control insulin secretion by coupling the excitability of the pancreatic beta-cell to glucose metabolism. Little is currently known about how the plasma membrane density of these channels is regulated. We therefore set out to examine in detail the endocytosis and recycling of these channels and how these processes are regulated. To achieve this goal, we expressed K(ATP) channels bearing an extracellular hemagglutinin epitope in human embryonic kidney cells and followed their fate along the endocytic pathway. Our results show that K(ATP) channels undergo multiple rounds of endocytosis and recycling. Further, activation of protein kinase C (PKC) with phorbol 12-myristate 13-acetate significantly decreases K(ATP) channel surface density by reducing channel recycling and diverting the channel to lysosomal degradation. These findings were recapitulated in the model pancreatic beta-cell line INS1e, where activation of PKC leads to a decrease in the surface density of native K(ATP) channels. Because sorting of internalized channels between lysosomal and recycling pathways could have opposite effects on the excitability of pancreatic beta-cells, we propose that PKC-regulated K(ATP) channel trafficking may play a role in the regulation of insulin secretion.
Collapse
Affiliation(s)
- Paul T. Manna
- From the Multidisciplinary Cardiovascular Research Centre, Institute of Membrane and Systems Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, United Kingdom
| | - Andrew J. Smith
- From the Multidisciplinary Cardiovascular Research Centre, Institute of Membrane and Systems Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, United Kingdom
| | - Tarvinder K. Taneja
- From the Multidisciplinary Cardiovascular Research Centre, Institute of Membrane and Systems Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, United Kingdom
| | - Gareth J. Howell
- From the Multidisciplinary Cardiovascular Research Centre, Institute of Membrane and Systems Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, United Kingdom
| | - Jonathan D. Lippiat
- From the Multidisciplinary Cardiovascular Research Centre, Institute of Membrane and Systems Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, United Kingdom
| | - Asipu Sivaprasadarao
- From the Multidisciplinary Cardiovascular Research Centre, Institute of Membrane and Systems Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, United Kingdom
| |
Collapse
|
20
|
Chiu YH, Alvarez-Baron C, Kim EY, Dryer SE. Dominant-negative regulation of cell surface expression by a pentapeptide motif at the extreme COOH terminus of an Slo1 calcium-activated potassium channel splice variant. Mol Pharmacol 2010; 77:497-507. [PMID: 20051533 DOI: 10.1124/mol.109.061929] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels regulate the physiology of many cell types. A single vertebrate gene variously known as Slo1, KCa1.1, or KCNMA1 encodes the pore-forming subunits of BK(Ca) channel but is expressed in a potentially very large number of alternative splice variants. Two splice variants of Slo1, Slo1(VEDEC) and Slo1(QEERL), which differ at the extreme COOH terminus, show markedly different steady-state expression levels on the cell surface. Here we show that Slo1(VEDEC) and Slo1(QEERL) can reciprocally coimmunoprecipitate, indicating that they form heteromeric complexes. Moreover, coexpression of even small amounts of Slo1(VEDEC) markedly reduces surface expression of Slo1(QEERL) and total Slo1 as indicated by cell-surface biotinylation assays. The effects of Slo1(VEDEC) on steady-state surface expression can be attributed primarily to the last five residues of the protein based on surface expression of motif-swapped constructs of Slo1 in human embryonic kidney (HEK) 293T cells. In addition, the presence of the VEDEC motif at the COOH terminus of Slo1 channels is sufficient to confer a dominant-negative effect on cell surface expression of itself or other types of Slo1 subunits. Treating cells with short peptides containing the VEDEC motif increased surface expression of Slo1(VEDEC) channels transiently expressed in HEK293T cells and increased current through endogenous BK(Ca) channels in mouse podocytes. Slo1(VEDEC) and Slo1(QEERL) channels are removed from the HEK293T cell surface with similar kinetics and to a similar extent, which suggests that the inhibitory effect of the VEDEC motif is exerted primarily on forward trafficking into the plasma membrane.
Collapse
Affiliation(s)
- Yu-Hsin Chiu
- Department of Biology and Biochemistry, University of Houston, 4800 Calhoun, Houston, TX 77204-5001, USA
| | | | | | | |
Collapse
|
21
|
Jha S, Dryer SE. The beta1 subunit of Na+/K+-ATPase interacts with BKCa channels and affects their steady-state expression on the cell surface. FEBS Lett 2009; 583:3109-14. [PMID: 19729011 PMCID: PMC2757478 DOI: 10.1016/j.febslet.2009.08.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Revised: 08/21/2009] [Accepted: 08/24/2009] [Indexed: 02/05/2023]
Abstract
Large conductance Ca2+-activated K+ channels (BKCa) encoded by the Slo1 gene play a role in the physiological regulation of many cell types. Here, we show that the beta1 subunit of Na+/K+-ATPase (NKbeta1) interacts with the cytoplasmic COOH-terminal region of Slo1 proteins. Reduced expression of endogenous NKbeta1 markedly inhibits evoked BKCa currents with no apparent effect on their gating. In addition, NKbeta1 down-regulated cells show decreased density of Slo1 subunits on the cell surface.
Collapse
Affiliation(s)
- Smita Jha
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204-5001
| | - Stuart E. Dryer
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204-5001
| |
Collapse
|
22
|
Hong YH, Kim JY, Lee JH, Chae HG, Jang SS, Jeon JH, Kim CH, Kim J, Kim SJ. Agonist-induced internalization of mGluR1alpha is mediated by caveolin. J Neurochem 2009; 111:61-71. [PMID: 19627451 DOI: 10.1111/j.1471-4159.2009.06289.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Agonist-induced internalization of metabotropic glutamate receptors (mGluRs) plays an important role in neuronal signaling. Although internalization of mGluRs has been reported to be mediated by clathrin-dependent pathway, studies describing clathrin-independent pathways are emerging. Here, we report that agonist-induced internalization of mGluR1alpha is mediated by caveolin. We show that two caveolin-binding motifs of mGluR1alpha interact with caveolin1/2. Using cell surface-immunoprecipitation and total internal reflection fluorescence imaging, we found that agonist-induced internalization of mGluR1alpha is regulated by caveolin-binding motifs of the receptor in heterologous cells. Moreover, in the cerebellum, group I mGluR agonist dihydroxyphenylglycol increased the interaction of phosphorylated caveolin with mGluR1alpha. This interaction was blocked by methyl-beta-cyclodextrin, known to disrupt caveolin/caveolae-dependent signaling by cholesterol depletion. Methyl-beta-cyclodextrin also blocked the agonist-induced internalization of mGluR1alpha. Thus, these findings represent the evidence for agonist-induced internalization of mGluR1alpha via caveolin and suggest that caveolin might play a role in synaptic metaplasticity by regulating internalization of mGluR1alpha in the cerebellum.
Collapse
Affiliation(s)
- Yun Hwa Hong
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Zou J, Zhang Y, Yin S, Wu H, Pyykkö I. Mitochondrial dysfunction disrupts trafficking of Kir4.1 in spiral ganglion satellite cells. J Neurosci Res 2009; 87:141-9. [DOI: 10.1002/jnr.21842] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
24
|
Klose A, Huth T, Alzheimer C. 1-[6-[[(17beta)-3-methoxyestra-1,3,5(10)-trien-17-yl]amino]hexyl]-1H-pyrrole-2,5-dione (U73122) selectively inhibits Kir3 and BK channels in a phospholipase C-independent fashion. Mol Pharmacol 2008; 74:1203-14. [PMID: 18682550 DOI: 10.1124/mol.108.047837] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
1-[6-[[(17beta)-3-methoxyestra-1,3,5(10)-trien-17-yl]amino]hexyl]-1H-pyrrole-2,5-dione (U73122) is widely used to inhibit phospholipase C (PLC)-mediated signaling, but we and others have also reported a PLC-independent block of Kir3 channels in native cells. To elaborate on this major side effect, we examined the action of U73122 and 1-[6-[[(17beta)-3-methoxyestra-1,3,5(10)-trien-17-yl]amino]hexyl]-2,5-pyrollidinedione (U73343), a structurally related but not PLC-inhibiting analog, on Kir1.1, Kir2.1, or Kir3.1/3.2 channels expressed in HEK293 cells. Both compounds (10 microM) displayed an unusual degree of selectivity for Kir3, superior even to that of tertiapin, which discriminates between Kir3 and Kir2 but also inhibits Kir1.1. Recordings from mutant Kir2 and Kir3 channels showed that U73122 is unlikely to block Kir3 by interfering with binding of phosphatidylinositol 4,5-bisphosphate, and U73122 did not seem to act like a pore blocker. U73122 and U73343 also unexpectedly suppressed Ca(2+)-activated K(+) channels of the large-conductance type (MaxiK, BK) in a PLC-independent fashion. In single-channel recordings, both compounds significantly decreased open probability of BK channels and slowed their ultrafast gating ("flickering") at very depolarized potentials. Alignment of the amino acid sequences of Kir3 and BK channels suggested that the highly selective effect of U73122/U73343 is mediated by a homologous domain within the long C-terminal ends. In fact, mutations in the C-terminal region of Kir2 and Kir3 channels significantly altered their sensitivity to the two compounds. Our data strongly caution against the use of U73122 when exploring signaling pathways involving Kir3 and BK channels. However, the apparent binding of U73122/U73343 to a common structural motif might be exploited to develop drugs selectively targeting Kir3 and BK channels.
Collapse
Affiliation(s)
- Angelika Klose
- Department of Physiology, University of Kiel, Olshausenstr. 40, 24098 Kiel, Germany
| | | | | |
Collapse
|
25
|
Estilo G, Liu W, Pastor-Soler N, Mitchell P, Carattino MD, Kleyman TR, Satlin LM. Effect of aldosterone on BK channel expression in mammalian cortical collecting duct. Am J Physiol Renal Physiol 2008; 295:F780-8. [PMID: 18579708 DOI: 10.1152/ajprenal.00002.2008] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Apical large-conductance Ca(2+)-activated K(+) (BK) channels in the cortical collecting duct (CCD) mediate flow-stimulated K(+) secretion. Dietary K(+) loading for 10-14 days leads to an increase in BK channel mRNA abundance, enhanced flow-stimulated K(+) secretion in microperfused CCDs, and a redistribution of immunodetectable channels from an intracellular pool to the apical membrane (Najjar F, Zhou H, Morimoto T, Bruns JB, Li HS, Liu W, Kleyman TR, Satlin LM. Am J Physiol Renal Physiol 289: F922-F932, 2005). To test whether this adaptation was mediated by a K(+)-induced increase in aldosterone, New Zealand White rabbits were fed a low-Na(+) (LS) or high-Na(+) (HS) diet for 7-10 days to alter circulating levels of aldosterone but not serum K(+) concentration. Single CCDs were isolated for quantitation of BK channel subunit (total, alpha-splice variants, beta-isoforms) mRNA abundance by real-time PCR and measurement of net transepithelial Na(+) (J(Na)) and K(+) (J(K)) transport by microperfusion; kidneys were processed for immunolocalization of BK alpha-subunit by immunofluorescence microscopy. At the time of death, LS rabbits excreted no urinary Na(+) and had higher circulating levels of aldosterone than HS animals. The relative abundance of BK alpha-, beta(2)-, and beta(4)-subunit mRNA and localization of immunodetectable alpha-subunit were similar in CCDs from LS and HS animals. In response to an increase in tubular flow rate from approximately 1 to 5 nl.min(-1).mm(-1), the increase in J(Na) was greater in LS vs. HS rabbits, yet the flow-stimulated increase in J(K) was similar in both groups. These data suggest that aldosterone does not contribute to the regulation of BK channel expression/activity in response to dietary K(+) loading.
Collapse
Affiliation(s)
- Genevieve Estilo
- Division of Pediatric Nephrology, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Alioua A, Lu R, Kumar Y, Eghbali M, Kundu P, Toro L, Stefani E. Slo1 caveolin-binding motif, a mechanism of caveolin-1-Slo1 interaction regulating Slo1 surface expression. J Biol Chem 2007; 283:4808-17. [PMID: 18079116 DOI: 10.1074/jbc.m709802200] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The large conductance, voltage- and Ca2+-activated potassium (MaxiK, BK) channel and caveolin-1 play important roles in regulating vascular contractility. Here, we hypothesized that the MaxiK alpha-subunit (Slo1) and caveolin-1 may interact with each other. Slo1 and caveolin-1 physiological association in native vascular tissue is strongly supported by (i) detergent-free purification of caveolin-1-rich domains demonstrating a pool of aortic Slo1 co-migrating with caveolin-1 to light density sucrose fractions, (ii) reverse co-immunoprecipitation, and (iii) double immunolabeling of freshly isolated myocytes revealing caveolin-1 and Slo1 proximity at the plasmalemma. In HEK293T cells, Slo1-caveolin-1 association was unaffected by the smooth muscle MaxiK beta1-subunit. Sequence analysis revealed two potential caveolin-binding motifs along the Slo1 C terminus, one equivalent, 1007YNMLCFGIY1015, and another mirror image, 537YTEYLSSAF545, to the consensus sequence, varphiXXXXvarphiXXvarphi. Deletion of 1007YNMLCFGIY1015 caused approximately 80% loss of Slo1-caveolin-1 association while preserving channel normal folding and overall Slo1 and caveolin-1 intracellular distribution patterns. 537YTEYLSSAF545 deletion had an insignificant dissociative effect. Interestingly, caveolin-1 coexpression reduced Slo1 surface and functional expression near 70% without affecting channel voltage sensitivity, and deletion of 1007YNMLCFGIY1015 motif obliterated channel surface expression. The results suggest 1007YNMLCFGIY1015 possible participation in Slo1 plasmalemmal targeting and demonstrate its role as a main mechanism for caveolin-1 association with Slo1 potentially serving a dual role: (i) maintaining channels in intracellular compartments downsizing their surface expression and/or (ii) serving as anchor of plasma membrane resident channels to caveolin-1-rich membranes. Because the caveolin-1 scaffolding domain is juxtamembrane, it is tempting to suggest that Slo1-caveolin-1 interaction facilitates the tethering of the Slo1 C-terminal end to the membrane.
Collapse
Affiliation(s)
- Abderrahmane Alioua
- Department of Anesthesiology, Brain Research Institute, Cardiovascular Research Laboratories, UCLA, Los Angeles, CA 90095-1778, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Zou S, Jha S, Kim EY, Dryer SE. A novel actin-binding domain on Slo1 calcium-activated potassium channels is necessary for their expression in the plasma membrane. Mol Pharmacol 2007; 73:359-68. [PMID: 17989352 DOI: 10.1124/mol.107.039743] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels regulate the physiological properties of many cell types. The gating properties of BK(Ca) channels are Ca(2+)-, voltage- and stretch-sensitive, and stretch-sensitive gating of these channels requires interactions with actin microfilaments subjacent to the plasma membrane. Moreover, we have previously shown that trafficking of BK(Ca) channels to the plasma membrane is associated with processes that alter cytoskeletal dynamics. Here, we show that the Slo1 subunits of BK(Ca) channels contain a novel cytoplasmic actin-binding domain (ABD) close to the C terminus, considerably downstream from regions of the channel molecule that play a major role in determining channel-gating properties. Binding of actin to the ABD can occur in a binary mixture in the absence of other proteins. Coexpression of a small ABD-green fluorescent protein fusion protein that competes with full-length Slo1 channels for binding to actin markedly suppresses trafficking of full-length Slo1 channels to the plasma membrane. In addition, Slo1 channels containing deletions of the ABD that eliminate actin binding are retained in intracellular pools, and they are not expressed on the cell surface. At least one point mutation within the ABD (L1020A) reduces surface expression of Slo1 channels to approximately 25% of wild type, but it does not cause a marked effect on the gating of point mutant channels that reach the cell surface. These data suggest that Slo1-actin interactions are necessary for normal trafficking of BK(Ca) channels to the plasma membrane and that the mechanisms of this interaction may be different from those that underlie F-actin and stretch-sensitive gating.
Collapse
Affiliation(s)
- Shengwei Zou
- Department of Biology and Biochemistry, University of Houston, 4800 Calhoun, Houston, TX 77204-5001, USA
| | | | | | | |
Collapse
|
28
|
Kim EY, Ridgway LD, Dryer SE. Interactions with filamin A stimulate surface expression of large-conductance Ca2+-activated K+ channels in the absence of direct actin binding. Mol Pharmacol 2007; 72:622-30. [PMID: 17586600 DOI: 10.1124/mol.107.038026] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels play an important role in the regulation of cell physiology in a wide variety of excitable and nonexcitable tissues. Filamin A is a conserved and ubiquitous actin-binding protein that forms perpendicular actin cross-links and contributes to changes in cell shape, stiffness, and motility. A variety of membrane proteins bind to filamin A, which regulates their trafficking in and out of the plasma membrane. Filamin A is therefore believed to couple membrane dynamics with those of the underlying cytoskeleton. Filamin A was identified in a yeast two-hybrid screen of a neuronal transcriptome using a subunit of BK(Ca) channels as bait, and the interaction was confirmed by a variety of biochemical assays in native neuronal cells and in human embryonic kidney 293T cells expressing BK(Ca) channels. BK(Ca) channels do not traffic to the plasma membrane in M2 melanoma cells, which lack filamin A, but normal trafficking is seen in A7 cells, which express filamin A, or in M2 cells transiently transfected with filamin A. It is noteworthy that stimulation of plasma membrane expression of BK(Ca) channels also occurs when M2 cells are transfected with filamin A constructs that lack the actin binding domain and that do not bind actin in vivo or in vitro. Filamin A is necessary for normal trafficking of BK(Ca) channels to the plasma membrane, but this effect does not require interactions with actin microfilaments, and it is possible that other actions of the filamin family of scaffolding proteins are independent of effects on actin.
Collapse
Affiliation(s)
- Eun Young Kim
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204-5001, USA
| | | | | |
Collapse
|
29
|
Kim EY, Ridgway LD, Zou S, Chiu YH, Dryer SE. Alternatively spliced C-terminal domains regulate the surface expression of large conductance calcium-activated potassium channels. Neuroscience 2007; 146:1652-61. [PMID: 17478049 PMCID: PMC1995407 DOI: 10.1016/j.neuroscience.2007.03.038] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2007] [Revised: 03/09/2007] [Accepted: 03/13/2007] [Indexed: 01/31/2023]
Abstract
The Slo1 gene, also known as KCNMA1, encodes the pore-forming subunits of large-conductance Ca2+-activated K+ (BK(Ca)) channels. Products of this gene are widely expressed in vertebrate tissues, and occur in a large number (>or=20) of alternatively spliced variants that vary in their gating properties, susceptibility to modulation, and trafficking to the plasma membrane. Motifs in the large cytoplasmic C-terminal are especially important in determining the functional properties of BK(Ca) channels. Here we report that chick ciliary ganglion neurons express transcripts and proteins of two Slo1 splice variants that differ at the extreme C-terminal. We refer to these variants as VEDEC and QEDRL (or QEERL for the orthologous mammalian versions), after the five terminal amino acid residues in each isoform. Individual ciliary ganglion neurons preferentially express these variants in different subcellular compartments. Moreover, QEERL channels show markedly higher levels of constitutive expression on the plasma membrane than VEDEC channels in HEK293T and NG108-15 cells. However, growth factor treatment can stimulate surface expression of VEDEC channels to levels comparable to those seen with QEERL. In addition, we show that co-expression of a soluble protein composed of VEDEC C-terminal tail residues markedly increases cell surface expression of full-length VEDEC channels, suggesting that this region binds to proteins that cause retention of the these channels in intracellular stores.
Collapse
Affiliation(s)
| | | | | | | | - Stuart E. Dryer
- Author for correspondence: , +1 713-743-2697 (ph), +1 713-743-2632 (FAX)
| |
Collapse
|
30
|
MacLeish PR, Nurse CA. Ion channel compartments in photoreceptors: evidence from salamander rods with intact and ablated terminals. J Neurophysiol 2007; 98:86-95. [PMID: 17460105 DOI: 10.1152/jn.00775.2006] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Vertebrate photoreceptors are highly polarized sensory cells in which several different ionic currents have been characterized. In the present study we used whole cell voltage-clamp and optical imaging techniques, the former combined with microsurgical manipulations, and simultaneous recording of membrane current and intracellular calcium signals to investigate the spatial distribution of ion channels within isolated salamander rods. In recordings from intact rods with visible terminals, evidence for five previously identified ionic currents was obtained. These include two Ca(2+)-dependent, i.e., a Ca(2+)-dependent chloride current [I(Cl(Ca))] and a large-conductance Ca(2+)- and voltage-dependent K(+) or BK current [I(K(Ca))], and three voltage-dependent currents, i.e., a delayed-rectifier type current [I(K(V))], a hyperpolarization-activated cation current (I(h)), and a dihydropyridine-sensitive L-type calcium current (I(Ca)). Of these, I(Cl(Ca)) was highly correlated with the presence of a terminal; rods with visible terminals expressed I(Cl(Ca)) without exception (n = 125), whereas approximately 71% of rods (40/56) without visible terminals lacked I(Cl(Ca)). More significantly, I(Cl(Ca)) was absent from all rods (n = 33) that had their terminals ablated, and recordings from the same cell before and after terminal ablation led, in all cases (n =10), to the loss of I(Cl(Ca)). In contrast, I(K(Ca)), I(K(V)), and I(h) remained largely intact after terminal ablation, suggesting that they arose principally from ion channels located in the soma and/or inner segment. The outward I(K)((Ca)) in terminal-ablated rods was reversibly suppressed on "puffing" a Ca(2+)-free extracellular solution over the soma and was appreciably enhanced by the L-type Ca(2+) channel agonist, Bay K 8644 (0.1-2 microM). These data indicate that rod photoreceptors possess discrete targeting mechanisms that preferentially sort ion channels mediating I(Cl(Ca)) to the terminal.
Collapse
Affiliation(s)
- Peter R MacLeish
- Department of Anatomy and Neurobiology, Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA 30310-1495, USA.
| | | |
Collapse
|
31
|
Kim EY, Zou S, Ridgway LD, Dryer SE. Beta1-subunits increase surface expression of a large-conductance Ca2+-activated K+ channel isoform. J Neurophysiol 2007; 97:3508-16. [PMID: 17329633 DOI: 10.1152/jn.00009.2007] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Auxiliary (beta) subunits of large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels regulate the gating properties of the functional channel complex. Here we show that an avian beta1-subunit also stimulates the trafficking of BK(Ca) channels to the plasma membrane in HEK293T cells and in a native population of developing vertebrate neurons. One C-terminal variant of BK(Ca) alpha-subunits, called the VEDEC isoform after its five last residues, is largely retained in intracellular compartments when it is heterologously expressed in HEK293T cells. A closely related splice variant, called QEERL, shows high levels of constitutive trafficking to the plasma membrane. Co-expression of beta1-subunits with the VEDEC isoform resulted in a large increase in surface BK(Ca) channels as assessed by cell-surface biotinylation assays, whole cell recordings of membrane current, and confocal microscopy in HEK293T cells. Co-expression of beta1-subunits slowed the gating kinetics of BK(Ca) channels, as reported previously. Consistent with this, overexpression of beta1-subunits in a native cell type that expresses intracellular VEDEC channels, embryonic day 9 chick ciliary ganglion neurons, resulted in a significant increase in macroscopic Ca(2+)-activated K(+) current. Both the cytoplasmic N- and C-terminal domains of avian beta1 are able to bind directly to VEDEC and QEERL channels. However, overexpression of the N-terminal domain by itself is sufficient to stimulate trafficking of VEDEC channels to the plasma membrane, whereas overexpression of either the cytoplasmic C-terminal domain or the extracellular loop domain did not affect surface expression of VEDEC.
Collapse
Affiliation(s)
- Eun Young Kim
- Dept. of Biology and Biochemistry, University of Houston, Houston, TX 77204-5001, USA
| | | | | | | |
Collapse
|
32
|
Davies KP, Stanevsky Y, Tar MT, Moses T, Chang JS, Chance MR, Melman A. Ageing causes cytoplasmic retention of MaxiK channels in rat corporal smooth muscle cells. Int J Impot Res 2007; 19:371-7. [PMID: 17287835 PMCID: PMC2043378 DOI: 10.1038/sj.ijir.3901541] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The MaxiK channel plays a critical role in the regulation of corporal smooth muscle tone and thereby erectile function. Given that ageing results in a decline in erectile function, we determined changes in the expression of MaxiK, which might impact erectile function. Quantitative-polymerase chain reaction demonstrated that although there is no significant change in transcription of the alpha- and beta-subunits that comprise the MaxiK channel, there are significant changes in the expression of transcripts encoding different splice variants. One transcript, SV1, is 13-fold increased in expression in the ageing rat corpora. SV1 has previously been reported to trap other isoforms of the MaxiK channel in the cytoplasm. Correlating with increased expression of SV1, we observed in older rats there is approximately a 13-fold decrease in MaxiK protein in the corpora cell membrane and a greater proportion is retained in the cytoplasm (approximately threefold). These experiments demonstrate that ageing of the corpora is accompanied by changes in alternative splicing and cellular localization of the MaxiK channel.
Collapse
Affiliation(s)
- K P Davies
- Department of Urology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Caruso-Neves C, Pinheiro AAS, Cai H, Souza-Menezes J, Guggino WB. PKB and megalin determine the survival or death of renal proximal tubule cells. Proc Natl Acad Sci U S A 2006; 103:18810-5. [PMID: 17121993 PMCID: PMC1693744 DOI: 10.1073/pnas.0605029103] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Renal proximal tubule cells have a remarkable ability to reabsorb large quantities of albumin through megalin-mediated endocytosis. This is an essential process for overall body homeostasis. Overstressing this endocytic system with a prolonged excess of albumin is injurious to proximal tubule cells. How these cells function and protect themselves from injury is unknown. Here, we show that megalin is the sensor that determines whether cells will be protected or injured by albumin. Megalin, through a novel mechanism, binds PKB in a D-3-phosphorylated phospholipid-insensitive manner, anchoring PKB in the luminal plasma membrane. Whereas low doses of albumin are protective, an overload of albumin decreases megalin expression followed by a reduction of plasma membrane PKB, PKB activity, and Bad phosphorylation induced by PKB. The result is albumin-induced apoptosis. These results reveal a model for PKB distribution in the plasma membrane and elucidate mechanisms involved in both the protective and toxic effects of albumin on proximal tubule cells. In addition, our findings suggest a mechanism for the progression of chronic kidney disease to end-stage renal disease.
Collapse
Affiliation(s)
- Celso Caruso-Neves
- *Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil 21949-900
- Departments of Physiology and
| | - Ana Acacia S. Pinheiro
- Department of Molecular Microbiology and Immunology, Johns Hopkins University School of Public Health, Baltimore, MD 21205
| | - Hui Cai
- Departments of Physiology and
- Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, MD 21205; and
| | - Jackson Souza-Menezes
- *Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil 21949-900
- Departments of Physiology and
| | - William B. Guggino
- Departments of Physiology and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
34
|
Hodson CA, Ambrogi IG, Scott RO, Mohler PJ, Milgram SL. Polarized apical sorting of guanylyl cyclase C is specified by a cytosolic signal. Traffic 2006; 7:456-64. [PMID: 16536743 DOI: 10.1111/j.1600-0854.2006.00398.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Receptor guanylyl cyclases respond to ligand stimulation by increasing intracellular cGMP, thereby initiating a variety of cell-signaling pathways. Furthermore, these proteins are differentially localized at the apical and basolateral membranes of epithelial cells. We have identified a region of 11 amino acids in the cytosolic COOH terminus of guanylyl cyclase C (GCC) required for normal apical localization in Madin-Darby canine kidney (MDCK) cells. These amino acids share no significant sequence homology with previously identified cytosolic apical sorting determinants. However, these amino acids are highly conserved and are sufficient to confer apical polarity to the interleukin-2 receptor alpha-chain (Tac). Additionally, we find two molecular weight species of GCC in lysates prepared from MDCK cells over-expressing GCC but observe only the fully mature species on the cell surface. Using pulse-chase analysis in polarized MDCK cells, we followed the generation of this mature species over time finding it to be detectable only at the apical cell surface. These data support the hypothesis that selective apical sorting can be determined using short, cytosolic amino acid motifs and argue for the existence of apical sorting machinery comparable with the machinery identified for basolateral protein traffic.
Collapse
Affiliation(s)
- Caleb A Hodson
- Graduate Program in Cell and Molecular Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|
35
|
Misonou H, Menegola M, Buchwalder L, Park EW, Meredith A, Rhodes KJ, Aldrich RW, Trimmer JS. Immunolocalization of the Ca2+-activated K+ channel Slo1 in axons and nerve terminals of mammalian brain and cultured neurons. J Comp Neurol 2006; 496:289-302. [PMID: 16566008 PMCID: PMC2605666 DOI: 10.1002/cne.20931] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Ca(2+)-activated voltage-dependent K(+) channels (Slo1, KCa1.1, Maxi-K, or BK channel) play a crucial role in controlling neuronal signaling by coupling channel activity to both membrane depolarization and intracellular Ca(2+) signaling. In mammalian brain, immunolabeling experiments have shown staining for Slo1 channels predominantly localized to axons and presynaptic terminals of neurons. We have developed anti-Slo1 mouse monoclonal antibodies that have been extensively characterized for specificity of staining against recombinant Slo1 in heterologous cells, and native Slo1 in mammalian brain, and definitively by the lack of detectable immunoreactivity against brain samples from Slo1 knockout mice. Here we provide precise immunolocalization of Slo1 in rat brain with one of these monoclonal antibodies and show that Slo1 is accumulated in axons and synaptic terminal zones associated with glutamatergic synapses in hippocampus and GABAergic synapses in cerebellum. By using cultured hippocampal pyramidal neurons as a model system, we show that heterologously expressed Slo1 is initially targeted to the axonal surface membrane, and with further development in culture, become localized in presynaptic terminals. These studies provide new insights into the polarized localization of Slo1 channels in mammalian central neurons and provide further evidence for a key role in regulating neurotransmitter release in glutamatergic and GABAergic terminals.
Collapse
Affiliation(s)
- Hiroaki Misonou
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616
- Department of Biochemistry and Cell Biology, State University of New York at Stony Brook, Stony Brook, NY 11794
| | - Milena Menegola
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616
| | - Lynn Buchwalder
- Department of Biochemistry and Cell Biology, State University of New York at Stony Brook, Stony Brook, NY 11794
| | - Eunice W. Park
- Department of Biochemistry and Cell Biology, State University of New York at Stony Brook, Stony Brook, NY 11794
| | - Andrea Meredith
- Department of Molecular and Cellular Physiology and Howard Hughes Medical Institute, Stanford School of Medicine, Stanford, CA 94305
| | | | - Richard W. Aldrich
- Department of Molecular and Cellular Physiology and Howard Hughes Medical Institute, Stanford School of Medicine, Stanford, CA 94305
| | - James S. Trimmer
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616
- Department of Biochemistry and Cell Biology, State University of New York at Stony Brook, Stony Brook, NY 11794
| |
Collapse
|
36
|
Chen L, Tian L, MacDonald SHF, McClafferty H, Hammond MSL, Huibant JM, Ruth P, Knaus HG, Shipston MJ. Functionally Diverse Complement of Large Conductance Calcium- and Voltage-activated Potassium Channel (BK) α-Subunits Generated from a Single Site of Splicing. J Biol Chem 2005; 280:33599-609. [PMID: 16081418 DOI: 10.1074/jbc.m505383200] [Citation(s) in RCA: 136] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The pore-forming alpha-subunits of large conductance calcium- and voltage-activated potassium (BK) channels are encoded by a single gene that undergoes extensive alternative pre-mRNA splicing. However, the extent to which differential exon usage at a single site of splicing may confer functionally distinct properties on BK channels is largely unknown. Here we demonstrated that alternative splicing at site of splicing C2 in the mouse BK channel C terminus generates five distinct splice variants: ZERO, e20, e21(STREX), e22, and a novel variant deltae23. Splice variants display distinct patterns of tissue distribution with e21(STREX) expressed at the highest levels in adult endocrine tissues and e22 at embryonic stages of mouse development. deltae23 is not functionally expressed at the cell surface and acts as a dominant negative of cell surface expression by trapping other BK channel splice variant alpha-subunits in the endoplasmic reticulum and perinuclear compartments. Splice variants display a range of biophysical properties. e21(STREX) and e22 variants display a significant left shift (>20 mV at 1 microM [Ca2+]i) in half-maximal voltage of activation compared with ZERO and e20 as well as considerably slower rates of deactivation. Splice variants are differentially sensitive to phosphorylation by endogenous cAMP-dependent protein kinase; ZERO, e20, and e22 variants are all activated, whereas e21 (STREX) is the only variant that is inhibited. Thus alternative pre-mRNA splicing from a single site of splicing provides a mechanism to generate a physiologically diverse complement of BK channel alpha-subunits that differ dramatically in their tissue distribution, trafficking, and regulation.
Collapse
Affiliation(s)
- Lie Chen
- Centre for Integrative Physiology, Membrane Biology Group, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, Scotland, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Lim HH, Park CS. Identification and functional characterization of ankyrin-repeat family protein ANKRA as a protein interacting with BKCa channel. Mol Biol Cell 2004; 16:1013-25. [PMID: 15616191 PMCID: PMC551470 DOI: 10.1091/mbc.e04-06-0537] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Ankyrin-repeat family A protein (ANKRA) was originally cloned in mouse as an interacting protein to megalin, a member of low-density lipoprotein receptor superfamily. Here, we report that the isolation of rat ANKRA as a new binding partner for the alpha-subunit of rat large-conductance Ca2+-activated K+ channel (rSlo). We mapped the binding region of each protein by using yeast two-hybrid and in vitro binding assays. ANKRA expressed together with rSlo channels were colocalized near the plasma membrane and coimmunoprecipitated in transfected cells. We also showed that BKCa channel in rat cerebral cortex coprecipitated with rANKRA and colocalized in cultured rat hippocampal neuron. Although the coexpression of ANKRA did not affect the surface expression of rSlo, the gating kinetics of rSlo channel was significantly altered and the effects were highly dependent on the intracellular calcium. These results indicate that ANKRA could modulate the excitability of neurons by binding directly to endogenous BKCa channel and altering its gating kinetics in a calcium-dependent manner.
Collapse
Affiliation(s)
- Hyun-Ho Lim
- Department of Life Science, Gwangju Institute of Science and Technology, Gwangju 500-712, Korea
| | | |
Collapse
|