1
|
Munaron L, Chinigò G, Scarpellino G, Ruffinatti FA. The fallacy of functional nomenclature in the kingdom of biological multifunctionality: physiological and evolutionary considerations on ion channels. J Physiol 2024; 602:2367-2381. [PMID: 37635695 DOI: 10.1113/jp284422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Living organisms are multiscale complex systems that have evolved high degrees of multifunctionality and redundancy in the structure-function relationship. A number of factors, only in part determined genetically, affect the jobs of proteins. The overall structural organization confers unique molecular properties that provide the potential to perform a pattern of activities, some of which are co-opted by specific environments. The variety of multifunctional proteins is expanding, but most cases are handled individually and according to the still dominant 'one structure-one function' approach, which relies on the attribution of canonical names typically referring to the first task identified for a given protein. The present topical review focuses on the multifunctionality of ion channels as a paradigmatic example. Mounting evidence reports the ability of many ion channels (including members of voltage-dependent, ligand-gated and transient receptor potential families) to exert biological effects independently of their ion conductivity. 'Functionally based' nomenclature (the practice of naming a protein or family of proteins based on a single purpose) is a conceptual bias for three main reasons: (i) it increases the amount of ambiguity, deceiving our understanding of the multiple contributions of biomolecules that is the heart of the complexity; (ii) it is in stark contrast to protein evolution dynamics, largely based on multidomain arrangement; and (iii) it overlooks the crucial role played by the microenvironment in adjusting the actions of cell structures and in tuning protein isoform diversity to accomplish adaptational requirements. Biological information in protein physiology is distributed among different entwined layers working as the primary 'locus' of natural selection and of evolutionary constraints.
Collapse
Affiliation(s)
- Luca Munaron
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Giorgia Chinigò
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Giorgia Scarpellino
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | | |
Collapse
|
2
|
Mulholland PJ, Padula AE, Wilhelm LJ, Park B, Grant KA, Ferguson BM, Cervera-Juanes R. Cross-species epigenetic regulation of nucleus accumbens KCNN3 transcripts by excessive ethanol drinking. Transl Psychiatry 2023; 13:364. [PMID: 38012158 PMCID: PMC10682415 DOI: 10.1038/s41398-023-02676-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/29/2023] Open
Abstract
The underlying genetic and epigenetic mechanisms driving functional adaptations in neuronal excitability and excessive alcohol intake are poorly understood. Small-conductance Ca2+-activated K+ (KCa2 or SK) channels encoded by the KCNN family of genes have emerged from preclinical studies as a key contributor to alcohol-induced functional neuroadaptations in alcohol-drinking monkeys and alcohol-dependent mice. Here, this cross-species analysis focused on KCNN3 DNA methylation, gene expression, and single nucleotide polymorphisms, including alternative promoters in KCNN3, that could influence surface trafficking and function of KCa2 channels. Bisulfite sequencing analysis of the nucleus accumbens tissue from alcohol-drinking monkeys and alcohol-dependent mice revealed a differentially methylated region in exon 1A of KCNN3 that overlaps with a predicted promoter sequence. The hypermethylation of KCNN3 in the accumbens paralleled an increase in the expression of alternative transcripts that encode apamin-insensitive and dominant-negative KCa2 channel isoforms. A polymorphic repeat in macaque KCNN3 encoded by exon 1 did not correlate with alcohol drinking. At the protein level, KCa2.3 channel expression in the accumbens was significantly reduced in very heavy-drinking monkeys. Together, our cross-species findings on epigenetic dysregulation of KCNN3 represent a complex mechanism that utilizes alternative promoters to potentially impact the firing of accumbens neurons. Thus, these results provide support for hypermethylation of KCNN3 as a possible key molecular mechanism underlying harmful alcohol intake and alcohol use disorder.
Collapse
Affiliation(s)
- Patrick J Mulholland
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Audrey E Padula
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Larry J Wilhelm
- Department of Translational Neuroscience, Atrium Health Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Byung Park
- Department of Public Health and Preventive Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Kathleen A Grant
- Department of Neurosciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Betsy M Ferguson
- Department of Neurosciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Rita Cervera-Juanes
- Department of Translational Neuroscience, Atrium Health Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
- Center for Precision Medicine, Atrium Health Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
3
|
Juanes RC, Mulholland P, Padula A, Wilhelm L, Park B, Grant K, Ferguson B. Cross-species epigenetic regulation of nucleus accumbens KCNN3 transcripts by excessive ethanol drinking. RESEARCH SQUARE 2023:rs.3.rs-3315122. [PMID: 37790552 PMCID: PMC10543433 DOI: 10.21203/rs.3.rs-3315122/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
The underlying genetic and epigenetic mechanisms driving functional adaptations in neuronal excitability and excessive alcohol intake are poorly understood. Small-conductance Ca2+-activated K+ (KCa2 or SK) channels encoded by the KCNN family of genes have emerged from preclinical studies as a key contributor to alcohol-induced functional neuroadaptations in alcohol-drinking monkeys and alcohol dependent mice. Here, this cross-species analysis focused on KCNN3 DNA methylation, gene expression, and single nucleotide polymorphisms including alternative promoters in KCNN3 that could influence surface trafficking and function of KCa2 channels. Bisulfite sequencing analysis of the nucleus accumbens tissue from alcohol-drinking monkeys and alcohol dependent mice revealed a differentially methylated region in exon 1A of KCNN3 that overlaps with a predicted promoter sequence. The hypermethylation of KCNN3 in the accumbens paralleled an increase in expression of alternative transcripts that encode apamin-insensitive and dominant-negative KCa2 channel isoforms. A polymorphic repeat in macaque KCNN3 encoded by exon 1 did not correlate with alcohol drinking. At the protein level, KCa2.3 channel expression in the accumbens was significantly reduced in very heavy drinking monkeys. Together, our cross-species findings on epigenetic dysregulation of KCNN3 represent a complex mechanism that utilizes alternative promoters to impact firing of accumbens neurons. Thus, these results provide support for hypermethylation of KCNN3 as a possible key molecular mechanism underlying harmful alcohol intake and alcohol use disorder.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Betsy Ferguson
- Oregon Health & Sciences University/Oregon National Primate Research Center
| |
Collapse
|
4
|
Engelhard CA, Khani S, Derdak S, Bilban M, Kornfeld JW. Nanopore sequencing unveils the complexity of the cold-activated murine brown adipose tissue transcriptome. iScience 2023; 26:107190. [PMID: 37564700 PMCID: PMC10410515 DOI: 10.1016/j.isci.2023.107190] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/28/2023] [Accepted: 06/16/2023] [Indexed: 08/12/2023] Open
Abstract
Alternative transcription increases transcriptome complexity by expression of multiple transcripts per gene. Annotation and quantification of transcripts using short-read sequencing is non-trivial. Long-read sequencing aims at overcoming these problems by sequencing full-length transcripts. Activation of brown adipose tissue (BAT) thermogenesis involves major transcriptomic remodeling and positively affects metabolism via increased energy expenditure. We benchmark Oxford Nanopore Technology (ONT) long-read sequencing protocols to Illumina short-read sequencing assessing alignment characteristics, gene and transcript detection and quantification, differential gene and transcript expression, transcriptome reannotation, and differential transcript usage (DTU). We find ONT sequencing is superior to Illumina for transcriptome reassembly, reducing the risk of false-positive events by unambiguously mapping reads to transcripts. We identified novel isoforms of genes undergoing DTU in cold-activated BAT including Cars2, Adtrp, Acsl5, Scp2, Aldoa, and Pde4d, validated by real-time PCR. The reannotated murine BAT transcriptome established here provides a framework for future investigations into the regulation of BAT.
Collapse
Affiliation(s)
- Christoph Andreas Engelhard
- Department for Biochemistry and Molecular Biology (BMB), University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Sajjad Khani
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Sophia Derdak
- Core Facilities, Medical University of Vienna, Lazarettgasse 14, 1090 Vienna, Austria
| | - Martin Bilban
- Department of Laboratory Medicine & Core Facilities, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Jan-Wilhelm Kornfeld
- Department for Biochemistry and Molecular Biology (BMB), University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| |
Collapse
|
5
|
Nam YW, Downey M, Rahman MA, Cui M, Zhang M. Channelopathy of small- and intermediate-conductance Ca 2+-activated K + channels. Acta Pharmacol Sin 2023; 44:259-267. [PMID: 35715699 PMCID: PMC9889811 DOI: 10.1038/s41401-022-00935-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 05/31/2022] [Indexed: 02/04/2023]
Abstract
Small- and intermediate-conductance Ca2+-activated K+ (KCa2.x/KCa3.1 also called SK/IK) channels are gated exclusively by intracellular Ca2+. The Ca2+ binding protein calmodulin confers sub-micromolar Ca2+ sensitivity to the channel-calmodulin complex. The calmodulin C-lobe is constitutively associated with the proximal C-terminus of the channel. Interactions between calmodulin N-lobe and the channel S4-S5 linker are Ca2+-dependent, which subsequently trigger conformational changes in the channel pore and open the gate. KCNN genes encode four subtypes, including KCNN1 for KCa2.1 (SK1), KCNN2 for KCa2.2 (SK2), KCNN3 for KCa2.3 (SK3), and KCNN4 for KCa3.1 (IK). The three KCa2.x channel subtypes are expressed in the central nervous system and the heart. The KCa3.1 subtype is expressed in the erythrocytes and the lymphocytes, among other peripheral tissues. The impact of dysfunctional KCa2.x/KCa3.1 channels on human health has not been well documented. Human loss-of-function KCa2.2 mutations have been linked with neurodevelopmental disorders. Human gain-of-function mutations that increase the apparent Ca2+ sensitivity of KCa2.3 and KCa3.1 channels have been associated with Zimmermann-Laband syndrome and hereditary xerocytosis, respectively. This review article discusses the physiological significance of KCa2.x/KCa3.1 channels, the pathophysiology of the diseases linked with KCa2.x/KCa3.1 mutations, the structure-function relationship of the mutant KCa2.x/KCa3.1 channels, and potential pharmacological therapeutics for the KCa2.x/KCa3.1 channelopathy.
Collapse
Affiliation(s)
- Young-Woo Nam
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Myles Downey
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Mohammad Asikur Rahman
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Meng Cui
- Department of Pharmaceutical Sciences, Northeastern University School of Pharmacy, Boston, MA, 02115, USA
| | - Miao Zhang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA.
| |
Collapse
|
6
|
Fuest S, Post C, Balbach ST, Jabar S, Neumann I, Schimmelpfennig S, Sargin S, Nass E, Budde T, Kailayangiri S, Altvater B, Ranft A, Hartmann W, Dirksen U, Rössig C, Schwab A, Pethő Z. Relevance of Abnormal KCNN1 Expression and Osmotic Hypersensitivity in Ewing Sarcoma. Cancers (Basel) 2022; 14:4819. [PMID: 36230742 PMCID: PMC9564116 DOI: 10.3390/cancers14194819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 12/02/2022] Open
Abstract
Ewing sarcoma (EwS) is a rare and highly malignant bone tumor occurring mainly in childhood and adolescence. Physiologically, the bone is a central hub for Ca2+ homeostasis, which is severely disturbed by osteolytic processes in EwS. Therefore, we aimed to investigate how ion transport proteins involved in Ca2+ homeostasis affect EwS pathophysiology. We characterized the expression of 22 candidate genes of Ca2+-permeable or Ca2+-regulated ion channels in three EwS cell lines and found the Ca2+-activated K+ channel KCa2.1 (KCNN1) to be exceptionally highly expressed. We revealed that KCNN1 expression is directly regulated by the disease-driving oncoprotein EWSR1-FL1. Due to its consistent overexpression in EwS, KCNN1 mRNA could be a prognostic marker in EwS. In a large cohort of EwS patients, however, KCNN1 mRNA quantity does not correlate with clinical parameters. Several functional studies including patch clamp electrophysiology revealed no evidence for KCa2.1 function in EwS cells. Thus, elevated KCNN1 expression is not translated to KCa2.1 channel activity in EwS cells. However, we found that the low K+ conductance of EwS cells renders them susceptible to hypoosmotic solutions. The absence of a relevant K+ conductance in EwS thereby provides an opportunity for hypoosmotic therapy that can be exploited during tumor surgery.
Collapse
Affiliation(s)
- Sebastian Fuest
- Institute of Physiology II, University Münster, 48149 Münster, Germany
| | - Christoph Post
- Institute of Physiology II, University Münster, 48149 Münster, Germany
| | - Sebastian T Balbach
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149 Münster, Germany
| | - Susanne Jabar
- Pediatrics III, University Hospital Essen, 45147 Essen, Germany
| | - Ilka Neumann
- Institute of Physiology II, University Münster, 48149 Münster, Germany
| | | | - Sarah Sargin
- Institute of Physiology II, University Münster, 48149 Münster, Germany
| | - Elke Nass
- Institute of Physiology I, University Münster, 48149 Münster, Germany
| | - Thomas Budde
- Institute of Physiology I, University Münster, 48149 Münster, Germany
| | - Sareetha Kailayangiri
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149 Münster, Germany
| | - Bianca Altvater
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149 Münster, Germany
| | - Andreas Ranft
- Pediatrics III, University Hospital Essen, 45147 Essen, Germany
| | - Wolfgang Hartmann
- Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, University Münster, 48149 Münster, Germany
| | - Uta Dirksen
- Pediatrics III, University Hospital Essen, 45147 Essen, Germany
| | - Claudia Rössig
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149 Münster, Germany
| | - Albrecht Schwab
- Institute of Physiology II, University Münster, 48149 Münster, Germany
| | - Zoltán Pethő
- Institute of Physiology II, University Münster, 48149 Münster, Germany
| |
Collapse
|
7
|
Nashed MG, Waye S, Hasan SMN, Nguyen D, Wiseman M, Zhang J, Lau H, Dinesh OC, Raymond R, Greig IR, Bambico FR, Nobrega JN. Antidepressant activity of pharmacological and genetic deactivation of the small-conductance calcium-activated potassium channel subtype-3. Psychopharmacology (Berl) 2022; 239:253-266. [PMID: 34982171 DOI: 10.1007/s00213-021-06045-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 12/13/2021] [Indexed: 11/25/2022]
Abstract
RATIONALE The voltage-insensitive, small-conductance calcium-activated potassium (SK) channel is a key regulator of neuronal depolarization and is implicated in the pathophysiology of depressive disorders. OBJECTIVE We ascertained whether the SK channel is impaired in the chronic unpredictable stress (CUS) model and whether it can serve as a molecular target of antidepressant action. METHODS We assessed the depressive-like behavioral phenotype of CUS-exposed rats and performed post-mortem SK channel binding and activity-dependent zif268 mRNA analyses on their brains. To begin an assessment of SK channel subtypes involved, we examined the effects of genetic and pharmacological inhibition of the SK3 channel using conditional knockout mice and selective SK3 channel negative allosteric modulators (NAMs). RESULTS We found that [125I]apamin binding to SK channels is increased in the prefrontal cortex and decreased in the hippocampus, an effect that was associated with reciprocal levels of zif268 mRNA transcripts indicating abnormal regional cell activity in this model. We found that genetic and pharmacological manipulations significantly decreased immobility in the forced swim test without altering general locomotor activity, a hallmark of antidepressant-like activity. CONCLUSIONS Taken together, these findings link depression-related neural and behavioral pathophysiology with abnormal SK channel functioning and suggest that this can be reversed by the selective inhibition of SK3 channels.
Collapse
Affiliation(s)
- Mina G Nashed
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| | - Shannon Waye
- Department of Psychology, Memorial University of Newfoundland, St. John's, Newfoundland & Labrador, A1B 3X9, Canada
| | - S M Nageeb Hasan
- Department of Psychology, Memorial University of Newfoundland, St. John's, Newfoundland & Labrador, A1B 3X9, Canada
| | - Diana Nguyen
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| | - Micaela Wiseman
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| | - Jing Zhang
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| | - Harry Lau
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| | - O Chandani Dinesh
- Department of Psychology, Memorial University of Newfoundland, St. John's, Newfoundland & Labrador, A1B 3X9, Canada
| | - Roger Raymond
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| | - Iain R Greig
- Institute of Medical Sciences, University of Aberdeen, King's College, Aberdeen, AB25 2ZD, Scotland
| | - Francis Rodriguez Bambico
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada. .,Department of Psychology, Memorial University of Newfoundland, St. John's, Newfoundland & Labrador, A1B 3X9, Canada.
| | - José N Nobrega
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| |
Collapse
|
8
|
Dwivedi D, Bhalla US. Physiology and Therapeutic Potential of SK, H, and M Medium AfterHyperPolarization Ion Channels. Front Mol Neurosci 2021; 14:658435. [PMID: 34149352 PMCID: PMC8209339 DOI: 10.3389/fnmol.2021.658435] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/13/2021] [Indexed: 12/19/2022] Open
Abstract
SK, HCN, and M channels are medium afterhyperpolarization (mAHP)-mediating ion channels. The three channels co-express in various brain regions, and their collective action strongly influences cellular excitability. However, significant diversity exists in the expression of channel isoforms in distinct brain regions and various subcellular compartments, which contributes to an equally diverse set of specific neuronal functions. The current review emphasizes the collective behavior of the three classes of mAHP channels and discusses how these channels function together although they play specialized roles. We discuss the biophysical properties of these channels, signaling pathways that influence the activity of the three mAHP channels, various chemical modulators that alter channel activity and their therapeutic potential in treating various neurological anomalies. Additionally, we discuss the role of mAHP channels in the pathophysiology of various neurological diseases and how their modulation can alleviate some of the symptoms.
Collapse
Affiliation(s)
- Deepanjali Dwivedi
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, GKVK Campus, Bengaluru, India.,Department of Neurobiology, Harvard Medical School, Boston, MA, United States.,Stanley Center at the Broad, Cambridge, MA, United States
| | - Upinder S Bhalla
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, GKVK Campus, Bengaluru, India
| |
Collapse
|
9
|
Weisbrod D. Small and Intermediate Calcium Activated Potassium Channels in the Heart: Role and Strategies in the Treatment of Cardiovascular Diseases. Front Physiol 2020; 11:590534. [PMID: 33329039 PMCID: PMC7719780 DOI: 10.3389/fphys.2020.590534] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/02/2020] [Indexed: 12/11/2022] Open
Abstract
Calcium-activated potassium channels are a heterogeneous family of channels that, despite their different biophysical characteristics, structures, and pharmacological signatures, play a role of transducer between the ubiquitous intracellular calcium signaling and the electric variations of the membrane. Although this family of channels was extensively described in various excitable and non-excitable tissues, an increasing amount of evidences shows their functional role in the heart. This review aims to focus on the physiological role and the contribution of the small and intermediate calcium-activated potassium channels in cardiac pathologies.
Collapse
|
10
|
Maldonado O, Jenkins A, Belalcazar HM, Hernandez-Cuervo H, Hyman KM, Ladaga G, Padilla L, de Erausquin GA. Age-dependent neuroprotective effect of an SK3 channel agonist on excitotoxity to dopaminergic neurons in organotypic culture. PLoS One 2020; 15:e0223633. [PMID: 32701951 PMCID: PMC7377472 DOI: 10.1371/journal.pone.0223633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 07/06/2020] [Indexed: 11/19/2022] Open
Abstract
Background Small conductance, calcium-activated (SK3) potassium channels control the intrinsic excitability of dopaminergic neurons (DN) in the midbrain and modulate their susceptibility to toxic insults during development. Methods We evaluated the age-dependency of the neuroprotective effect of an SK3 agonist, 1-Ethyl-1,3-dihydro-2H-benzimidazol-2-one (1-EBIO), on Amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) excitotoxicity to DN in ventral mesencephalon (VM) organotypic cultures. Results Most tyrosine hydroxylase (TH)+ neurons were also SK3+; SK3+/TH- cells (DN+) were common at each developmental stage but more prominently at day in vitro (DIV) 8. Young DN+ neurons were small bipolar and fusiform, whereas mature ones were large and multipolar. Exposure of organotypic cultures to AMPA (100 μm, 16 h) had no effect on the survival of DN+ at DIV 8, but caused significant toxicity at DIV 15 (n = 15, p = 0.005) and DIV 22 (n = 15, p<0.001). These results indicate that susceptibility of DN to AMPA excitotoxicity is developmental stage-dependent in embryonic VM organotypic cultures. Immature DN+ (small, bipolar) were increased after AMPA (100 μm, 16 h) at DIV 8, at the expense of the number of differentiated (large, multipolar) DN+ (p = 0.039). This effect was larger at DIV 15 (p<<<0.0001) and at DIV 22 (p<<<0.0001). At DIV 8, 30 μM 1-EBIO resulted in a large increase in DN+. At DIV 15, AMPA toxicity was prevented by exposure to 30 μM, but not 100 μM 1-EBIO. At DIV 22, excitotoxicity was unaffected by 30 μM 1-EBIO, and partially reduced by 100 μM 1-EBIO. Conclusion The effects of the SK3 channel agonist 1-EBIO on the survival of SK3-expressing dopaminergic neurons were concentration-dependent and influenced by neuronal developmental stage.
Collapse
Affiliation(s)
- Oscar Maldonado
- Laboratory of Brain Development, and Repair, Biggs Institute for Alzheimer and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, Texas, United States of America
| | - Alexandra Jenkins
- Department of Molecular Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Helen M. Belalcazar
- Department of Genetics, Albert Einstein College of Medicine, New York City, New York, United States of America
| | - Helena Hernandez-Cuervo
- Department of Molecular Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Katelynn M. Hyman
- Department of Molecular Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Giannina Ladaga
- Laboratory of Brain Development, and Repair, Biggs Institute for Alzheimer and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, Texas, United States of America
| | - Lucia Padilla
- Laboratory of Brain Development, and Repair, Biggs Institute for Alzheimer and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, Texas, United States of America
| | - Gabriel A. de Erausquin
- Laboratory of Brain Development, and Repair, Biggs Institute for Alzheimer and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|
11
|
Hypobaric Hypoxia-Induced Learning and Memory Impairment: Elucidating the Role of Small Conductance Ca2+-Activated K+ Channels. Neuroscience 2018; 388:418-429. [DOI: 10.1016/j.neuroscience.2018.07.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/11/2018] [Accepted: 07/13/2018] [Indexed: 11/19/2022]
|
12
|
Honrath B, Krabbendam IE, Culmsee C, Dolga AM. Small conductance Ca 2+-activated K + channels in the plasma membrane, mitochondria and the ER: Pharmacology and implications in neuronal diseases. Neurochem Int 2017; 109:13-23. [PMID: 28511953 DOI: 10.1016/j.neuint.2017.05.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/24/2017] [Accepted: 05/08/2017] [Indexed: 12/14/2022]
Abstract
Ca2+-activated K+ (KCa) channels regulate after-hyperpolarization in many types of neurons in the central and peripheral nervous system. Small conductance Ca2+-activated K+ (KCa2/SK) channels, a subfamily of KCa channels, are widely expressed in the nervous system, and in the cardiovascular system. Voltage-independent SK channels are activated by alterations in intracellular Ca2+ ([Ca2+]i) which facilitates the opening of these channels through binding of Ca2+ to calmodulin that is constitutively bound to the SK2 C-terminus. In neurons, SK channels regulate synaptic plasticity and [Ca2+]i homeostasis, and a number of recent studies elaborated on the emerging neuroprotective potential of SK channel activation in conditions of excitotoxicity and cerebral ischemia, as well as endoplasmic reticulum (ER) stress and oxidative cell death. Recently, SK channels were discovered in the inner mitochondrial membrane and in the membrane of the endoplasmic reticulum which sheds new light on the underlying molecular mechanisms and pathways involved in SK channel-mediated protective effects. In this review, we will discuss the protective properties of pharmacological SK channel modulation with particular emphasis on intracellularly located SK channels as potential therapeutic targets in paradigms of neuronal dysfunction.
Collapse
Affiliation(s)
- Birgit Honrath
- Institute of Pharmacology and Clinical Pharmacy, University of Marburg, 35043 Marburg, Germany; Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Inge E Krabbendam
- Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Carsten Culmsee
- Institute of Pharmacology and Clinical Pharmacy, University of Marburg, 35043 Marburg, Germany
| | - Amalia M Dolga
- Institute of Pharmacology and Clinical Pharmacy, University of Marburg, 35043 Marburg, Germany; Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands.
| |
Collapse
|
13
|
González C, Baez-Nieto D, Valencia I, Oyarzún I, Rojas P, Naranjo D, Latorre R. K(+) channels: function-structural overview. Compr Physiol 2013; 2:2087-149. [PMID: 23723034 DOI: 10.1002/cphy.c110047] [Citation(s) in RCA: 157] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Potassium channels are particularly important in determining the shape and duration of the action potential, controlling the membrane potential, modulating hormone secretion, epithelial function and, in the case of those K(+) channels activated by Ca(2+), damping excitatory signals. The multiplicity of roles played by K(+) channels is only possible to their mammoth diversity that includes at present 70 K(+) channels encoding genes in mammals. Today, thanks to the use of cloning, mutagenesis, and the more recent structural studies using x-ray crystallography, we are in a unique position to understand the origins of the enormous diversity of this superfamily of ion channels, the roles they play in different cell types, and the relations that exist between structure and function. With the exception of two-pore K(+) channels that are dimers, voltage-dependent K(+) channels are tetrameric assemblies and share an extremely well conserved pore region, in which the ion-selectivity filter resides. In the present overview, we discuss in the function, localization, and the relations between function and structure of the five different subfamilies of K(+) channels: (a) inward rectifiers, Kir; (b) four transmembrane segments-2 pores, K2P; (c) voltage-gated, Kv; (d) the Slo family; and (e) Ca(2+)-activated SK family, SKCa.
Collapse
Affiliation(s)
- Carlos González
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | | | | | | | | | | | | |
Collapse
|
14
|
Definitive Endoderm Formation from Plucked Human Hair-Derived Induced Pluripotent Stem Cells and SK Channel Regulation. Stem Cells Int 2013; 2013:360573. [PMID: 23710194 PMCID: PMC3654369 DOI: 10.1155/2013/360573] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 03/13/2013] [Indexed: 11/25/2022] Open
Abstract
Pluripotent stem cells present an extraordinary powerful tool to investigate embryonic development in humans. Essentially, they provide a unique platform for dissecting the distinct mechanisms underlying pluripotency and subsequent lineage commitment. Modest information currently exists about the expression and the role of ion channels during human embryogenesis, organ development, and cell fate determination. Of note, small and intermediate conductance, calcium-activated potassium channels have been reported to modify stem cell behaviour and differentiation. These channels are broadly expressed throughout human tissues and are involved in various cellular processes, such as the after-hyperpolarization in excitable cells, and also in differentiation processes. To this end, human induced pluripotent stem cells (hiPSCs) generated from plucked human hair keratinocytes have been exploited in vitro to recapitulate endoderm formation and, concomitantly, used to map the expression of the SK channel (SKCa) subtypes over time. Thus, we report the successful generation of definitive endoderm from hiPSCs of ectodermal origin using a highly reproducible and robust differentiation system. Furthermore, we provide the first evidence that SKCas subtypes are dynamically regulated in the transition from a pluripotent stem cell to a more lineage restricted, endodermal progeny.
Collapse
|
15
|
Linta L, Boeckers TM, Kleger A, Liebau S. Calcium activated potassium channel expression during human iPS cell-derived neurogenesis. Ann Anat 2013; 195:303-311. [PMID: 23587809 DOI: 10.1016/j.aanat.2013.02.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 02/05/2013] [Accepted: 02/27/2013] [Indexed: 01/11/2023]
Abstract
The family of calcium activated potassium channels of low and intermediate conductance, known as SK channels, consists of four members (SK1-4). These channels are widely expressed throughout the organism and involved in various cellular processes, such as the afterhyperpolarization in excitable cells but also in differentiation processes of various tissues. To date, the role of SK channels in developmental processes has been merely a marginal focus of investigation, although it is well accepted that cell differentiation and maturation affect the expression patterns of certain ion channels. Recently, several studies from our laboratory delineated the influence of SK channel expression and their respective activity on cytoskeletal reorganization in neural and pluripotent stem cells and regulation of cell fate determination toward the cardiac lineage in human and mouse pluripotent stem cells. Herein, we have now analyzed SK channel expression patterns and distribution at various stages of human induced pluripotent stem cell-derived neurogenesis particularly focusing on undifferentiated iPS cells, neural progenitors and mature neurons. All family members could be detected starting at the iPS cell level and were differentially expressed during the subsequent maturation process. Intriguingly, we found obvious discrepancies between mRNA and protein expression pointing toward a complex regulatory mechanism. Inhibition of SK channels with either apamin or clotrimazol did not have any significant effects on the speed or amount of neurogenesis in vitro. The abundance and specific regulation of SK channel expression during iPS cell differentiation indicates distinct roles of these ion channels not only for the cardiac but also for neuronal cell differentiation and in vitro neurogenesis.
Collapse
Affiliation(s)
- Leonhard Linta
- Institute for Anatomy & Cell Biology, Ulm University, Ulm, Germany
| | | | - Alexander Kleger
- Department of Internal Medicine I, Ulm University, Ulm, Germany.
| | - Stefan Liebau
- Institute for Anatomy & Cell Biology, Ulm University, Ulm, Germany.
| |
Collapse
|
16
|
Andres MA. Glucose-sensitivity of the afterhyperpolarization potential: role of SK1 channel in insulin-secreting cells. Gen Comp Endocrinol 2012; 178:459-62. [PMID: 22809667 DOI: 10.1016/j.ygcen.2012.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 07/08/2012] [Indexed: 11/19/2022]
Abstract
The role of the small-conductance, calcium-activated SK potassium channel in regulating pancreatic β cell function remains controversial with conflicting pharmacological results. In this study, we used current clamp recordings to further characterize the function of SK channels in INS-1 cell line. We compared afterhyperpolarization potential (AHP) responses of SK1-downregulated cells with those of control INS-1 cells. They were tested with and without the presence of glucose. We found that cells in which SK1 channel subunit expression had been downregulated exhibited AHPs in the presence of 20mM glucose while control INS-1 cells had AHPs only in the absence of glucose. Our findings show that the glucose-dependence of the AHP in the rat INS-1 cell line depends only on SK1 channel subunit expression.
Collapse
Affiliation(s)
- Marilou A Andres
- Pacific Biosciences Research Center, University of Hawaii, Honolulu, HI 96822, USA.
| |
Collapse
|
17
|
Balut CM, Hamilton KL, Devor DC. Trafficking of intermediate (KCa3.1) and small (KCa2.x) conductance, Ca(2+)-activated K(+) channels: a novel target for medicinal chemistry efforts? ChemMedChem 2012; 7:1741-55. [PMID: 22887933 DOI: 10.1002/cmdc.201200226] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 07/09/2012] [Indexed: 12/22/2022]
Abstract
Ca(2+)-activated K(+) (KCa) channels play a pivotal role in the physiology of a wide variety of tissues and disease states, including vascular endothelia, secretory epithelia, certain cancers, red blood cells (RBC), neurons, and immune cells. Such widespread involvement has generated an intense interest in elucidating the function and regulation of these channels, with the goal of developing pharmacological strategies aimed at selective modulation of KCa channels in various disease states. Herein we give an overview of the molecular and functional properties of these channels and their therapeutic importance. We discuss the achievements made in designing pharmacological tools that control the function of KCa channels by modulating their gating properties. Moreover, this review discusses the recent advances in our understanding of KCa channel assembly and anterograde trafficking toward the plasma membrane, the micro-domains in which these channels are expressed within the cell, and finally the retrograde trafficking routes these channels take following endocytosis. As the regulation of intracellular trafficking by agonists as well as the protein-protein interactions that modify these events continue to be explored, we anticipate this will open new therapeutic avenues for the targeting of these channels based on the pharmacological modulation of KCa channel density at the plasma membrane.
Collapse
Affiliation(s)
- Corina M Balut
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | | |
Collapse
|
18
|
Differential expression of genes encoding neuronal ion-channel subunits in major depression, bipolar disorder and schizophrenia: implications for pathophysiology. Int J Neuropsychopharmacol 2012; 15:869-82. [PMID: 22008100 DOI: 10.1017/s1461145711001428] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Evidence concerning ion-channel abnormalities in the pathophysiology of common psychiatric disorders is still limited. Given the significance of ion channels in neuronal activity, neurotransmission and neuronal plasticity we hypothesized that the expression patterns of genes encoding different ion channels may be altered in schizophrenia, bipolar and unipolar disorders. Frozen samples of striatum including the nucleus accumbens (Str-NAc) and the lateral cerebellar hemisphere of 60 brains from depressed (MDD), bipolar (BD), schizophrenic and normal subjects, obtained from the Stanley Foundation Brain Collection, were assayed. mRNA of 72 different ion-channel subunits were determined by qRT-PCR and alteration in four genes were verified by immunoblotting. In the Str-NAc the prominent change was observed in the MDD group, in which there was a significant up-regulation in genes encoding voltage-gated potassium-channel subunits. However, in the lateral cerebellar hemisphere (cerebellum), the main change was observed in schizophrenia specimens, as multiple genes encoding various ion-channel subunits were significantly down-regulated. The impaired expression of genes encoding ion channels demonstrates a disease-related neuroanatomical pattern. The alterations observed in Str-NAc of MDD may imply electrical hypo-activity of this region that could be of relevance to MDD symptoms and treatment. The robust unidirectional alteration of both excitatory and inhibitory ion channels in the cerebellum may suggests cerebellar general hypo-transcriptional activity in schizophrenia.
Collapse
|
19
|
Mulholland PJ. K(Ca)2 channels: novel therapeutic targets for treating alcohol withdrawal and escalation of alcohol consumption. Alcohol 2012; 46:309-15. [PMID: 22464787 DOI: 10.1016/j.alcohol.2011.11.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 11/18/2011] [Accepted: 11/28/2011] [Indexed: 01/07/2023]
Abstract
Small-conductance, calcium-activated potassium (K(Ca)2) channels influence neuronal firing properties, intrinsic excitability, and NMDA receptor-dependent synaptic responses and plasticity. In this mini-review, we discuss new evidence that chronic alcohol-associated plasticity critically involves K(Ca)2 channels in hippocampus, ventral tegmental area, and nucleus accumbens. K(Ca)2 channel activity can modulate the magnitude of excitation of midbrain dopamine neurons induced by acute alcohol exposure. Emerging evidence indicates that K(Ca)2 channels regulate neuroadaptations to chronic alcohol that contribute to withdrawal hyperexcitability and escalation of voluntary alcohol consumption. Restoring K(Ca)2 channel activity can attenuate the severity of the alcohol withdrawal syndrome in vivo and withdrawal-associated neurotoxicity in vitro. Pharmacological modulation of K(Ca)2 channels can bi-directionally influence drinking behavior in rat and mouse models of voluntary alcohol consumption. Collectively, these studies using various rodent models have clearly indicated a central role for K(Ca)2 channels in the neuroplasticity of chronic alcohol exposure. In addition, accumulating evidence suggests that K(Ca)2 channels are a novel therapeutic target to alleviate the symptoms of alcohol withdrawal and reduce high amounts of alcohol drinking.
Collapse
|
20
|
Liebau S, Tischendorf M, Ansorge D, Linta L, Stockmann M, Weidgang C, Iacovino M, Boeckers T, von Wichert G, Kyba M, Kleger A. An inducible expression system of the calcium-activated potassium channel 4 to study the differential impact on embryonic stem cells. Stem Cells Int 2011; 2011:456815. [PMID: 21941566 PMCID: PMC3173888 DOI: 10.4061/2011/456815] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 06/14/2011] [Indexed: 11/20/2022] Open
Abstract
Rationale. The family of calcium-activated potassium channels consists of four members with varying biological functions and conductances. Besides membrane potential modulation, SK channels have been found to be involved in cardiac pacemaker cell development from ES cells and morphological shaping of neural stem cells. Objective. Distinct SK channel subtype expression in ES cells might elucidate their precise impact during cardiac development. We chose SK channel subtype 4 as a potential candidate influencing embryonic stem cell differentiation. Methods. We generated a doxycycline inducible mouse ES cell line via targeted homologous recombination of a cassette expressing a bicistronic construct encoding SK4 and a fluorophore from the murine HPRT locus. Conclusion. We characterized the mouse ES cell line iSK4-AcGFP. The cassette is readily expressed under the control of doxycycline, and the overexpression of SK4 led to an increase in cardiac and pacemaker cell differentiation thereby serving as a unique tool to characterize the cell biological variances due to specific SK channel overexpression.
Collapse
Affiliation(s)
- Stefan Liebau
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Schijndel JEV, Martens GJM. Gene expression profiling in rodent models for schizophrenia. Curr Neuropharmacol 2011; 8:382-93. [PMID: 21629445 PMCID: PMC3080594 DOI: 10.2174/157015910793358132] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Revised: 04/15/2010] [Accepted: 04/30/2010] [Indexed: 12/12/2022] Open
Abstract
The complex neurodevelopmental disorder schizophrenia is thought to be induced by an interaction between predisposing genes and environmental stressors. In order to get a better insight into the aetiology of this complex disorder, animal models have been developed. In this review, we summarize mRNA expression profiling studies on neurodevelopmental, pharmacological and genetic animal models for schizophrenia. We discuss parallels and contradictions among these studies, and propose strategies for future research.
Collapse
Affiliation(s)
- Jessica E Van Schijndel
- Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Centre for Neuroscience & Nijmegen Centre for Molecular Life Sciences (NCMLS), Faculty of Science, Radboud University Nijmegen, 6525 GA, Nijmegen, The Netherlands
| | | |
Collapse
|
22
|
Calcium-Activated Potassium Channels, Cardiogenesis of Pluripotent Stem Cells, and Enrichment of Pacemaker-Like Cells. Trends Cardiovasc Med 2011; 21:74-83. [DOI: 10.1016/j.tcm.2012.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
23
|
Ohya S, Niwa S, Yanagi A, Fukuyo Y, Yamamura H, Imaizumi Y. Involvement of dominant-negative spliced variants of the intermediate conductance Ca2+-activated K+ channel, K(Ca)3.1, in immune function of lymphoid cells. J Biol Chem 2011; 286:16940-52. [PMID: 21345794 DOI: 10.1074/jbc.m110.184192] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The intermediate conductance Ca(2+)-activated K(+) channel (IK(Ca) channel) encoded by K(Ca)3.1 is responsible for the control of proliferation and differentiation in various types of cells. We identified novel spliced variants of K(Ca)3.1 (human (h) K(Ca)3.1b) from the human thymus, which were lacking the N-terminal domains of the original hK(Ca)3.1a as a result of alternative splicing events. hK(Ca)3.1b was significantly expressed in human lymphoid tissues. Western blot analysis showed that hK(Ca)3.1a proteins were mainly expressed in the plasma membrane fraction, whereas hK(Ca)3.1b was in the cytoplasmic fraction. We also identified a similar N terminus lacking K(Ca)3.1 variants from mice and rat lymphoid tissues (mK(Ca)3.1b and rK(Ca)3.1b). In the HEK293 heterologous expression system, the cellular distribution of cyan fluorescent protein-tagged hK(Ca)3.1a and/or YFP-tagged hK(Ca)3.1b isoforms showed that hK(Ca)3.1b suppressed the localization of hK(Ca)3.1a to the plasma membrane. In the Xenopus oocyte translation system, co-expression of hK(Ca)3.1b with hK(Ca)3.1a suppressed IK(Ca) channel activity of hK(Ca)3.1a in a dominant-negative manner. In addition, this study indicated that up-regulation of mK(Ca)3.1b in mouse thymocytes differentiated CD4(+)CD8(+) phenotype thymocytes into CD4(-)CD8(-) ones and suppressed concanavalin-A-stimulated thymocyte growth by down-regulation of mIL-2 transcripts. Anti-proliferative effects and down-regulation of mIL-2 transcripts were also observed in mK(Ca)3.1b-overexpressing mouse thymocytes. These suggest that the N-terminal domain of K(Ca)3.1 is critical for channel trafficking to the plasma membrane and that the fine-tuning of IK(Ca) channel activity modulated through alternative splicing events may be related to the control in physiological and pathophysiological conditions in T-lymphocytes.
Collapse
Affiliation(s)
- Susumu Ohya
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | | | | | | | | | | |
Collapse
|
24
|
Nijholt IM, Granic I, Luiten PGM, Eisel ULM. TNFR2 - target for therapeutics against neurodegenerative diseases? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 691:567-73. [PMID: 21153362 DOI: 10.1007/978-1-4419-6612-4_59] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Ingrid M Nijholt
- Department of Molecular Neurobiology, University of Groningen, Groningen, The Netherlands.
| | | | | | | |
Collapse
|
25
|
Berkefeld H, Fakler B, Schulte U. Ca2+-activated K+ channels: from protein complexes to function. Physiol Rev 2010; 90:1437-59. [PMID: 20959620 DOI: 10.1152/physrev.00049.2009] [Citation(s) in RCA: 193] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Molecular research on ion channels has demonstrated that many of these integral membrane proteins associate with partner proteins, often versatile in their function, or even assemble into stable macromolecular complexes that ensure specificity and proper rate of the channel-mediated signal transduction. Calcium-activated potassium (K(Ca)) channels that link excitability and intracellular calcium concentration are responsible for a wide variety of cellular processes ranging from regulation of smooth muscle tone to modulation of neurotransmission and control of neuronal firing pattern. Most of these functions are brought about by interaction of the channels' pore-forming subunits with distinct partner proteins. In this review we summarize recent insights into protein complexes associated with K(Ca) channels as revealed by proteomic research and discuss the results available on structure and function of these complexes and on the underlying protein-protein interactions. Finally, the results are related to their significance for the function of K(Ca) channels under cellular conditions.
Collapse
Affiliation(s)
- Henrike Berkefeld
- Institute of Physiology II, University of Freiburg, and Centre for Biological Signalling Studies (Bioss),Freiburg, Germany.
| | | | | |
Collapse
|
26
|
Tuteja D, Rafizadeh S, Timofeyev V, Wang S, Zhang Z, Li N, Mateo RK, Singapuri A, Young JN, Knowlton AA, Chiamvimonvat N. Cardiac small conductance Ca2+-activated K+ channel subunits form heteromultimers via the coiled-coil domains in the C termini of the channels. Circ Res 2010; 107:851-9. [PMID: 20689065 DOI: 10.1161/circresaha.109.215269] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Ca(2+)-activated K(+) channels are present in a wide variety of cells. We have previously reported the presence of small conductance Ca(2+)-activated K(+) (SK or K(Ca)) channels in human and mouse cardiac myocytes that contribute functionally toward the shape and duration of cardiac action potentials. Three isoforms of SK channel subunits (SK1, SK2, and SK3) are found to be expressed. Moreover, there is differential expression with more abundant SK channels in the atria and pacemaking tissues compared with the ventricles. SK channels are proposed to be assembled as tetramers similar to other K(+) channels, but the molecular determinants driving their subunit interaction and assembly are not defined in cardiac tissues. OBJECTIVE To investigate the heteromultimeric formation and the domain necessary for the assembly of 3 SK channel subunits (SK1, SK2, and SK3) into complexes in human and mouse hearts. METHODS AND RESULTS Here, we provide evidence to support the formation of heteromultimeric complexes among different SK channel subunits in native cardiac tissues. SK1, SK2, and SK3 subunits contain coiled-coil domains (CCDs) in the C termini. In vitro interaction assay supports the direct interaction between CCDs of the channel subunits. Moreover, specific inhibitory peptides derived from CCDs block the Ca(2+)-activated K(+) current in atrial myocytes, which is important for cardiac repolarization. CONCLUSIONS The data provide evidence for the formation of heteromultimeric complexes among different SK channel subunits in atrial myocytes. Because SK channels are predominantly expressed in atrial myocytes, specific ligands of the different isoforms of SK channel subunits may offer a unique therapeutic opportunity to directly modify atrial cells without interfering with ventricular myocytes.
Collapse
Affiliation(s)
- Dipika Tuteja
- Department of Medicine, University of California, Davis, CA 95616, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Benes FM. Amygdalocortical circuitry in schizophrenia: from circuits to molecules. Neuropsychopharmacology 2010; 35:239-57. [PMID: 19727065 PMCID: PMC3055447 DOI: 10.1038/npp.2009.116] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2009] [Revised: 07/09/2009] [Accepted: 07/16/2009] [Indexed: 12/19/2022]
Abstract
Schizophrenia is a disorder in which disturbances in the integration of emotion with cognition plays a central role and probably involves several different regions, including the dorsolateral prefrontal cortex, the rostral anterior cingulate cortex, the hippocampal formation, and basolateral amygdala (BLA). Recent brain imaging studies have reported changes in volume, whereas postmortem studies point to dysfunction of the GABA and glutamate systems in these regions. Microarray-based profiles indicate that complex changes in the expression of genes associated with synaptic transmission and ion channels are involved in GABA cell dysfunction in schizophrenics. Molecular abnormalities vary considerably on the basis of sector and layer, suggesting that the unique connectivity of intrinsic and extrinsic afferents may critical in regulating the activity of genes in specific subpopulations of GABA cells. Projections of the BLA may be of particular importance to the induction of abnormal circuitry in schizophrenia, as their ingrowth during late adolescence and early adulthood may help to 'trigger' the onset of illness in susceptible individuals. A preponderance of cellular and molecular abnormalities has been found in the stratum oriens (SO) of sectors CA3/2 in which BLA afferents provide a robust innervation. These observations have lead to the development of a rodent model for the study of abnormal circuitry in this disorder. For example, single-cell recordings in hippocampal slices exposed to increased activation from the BLA have shown decreases in GABA currents in pyramidal neurons in SO of CA3/2, but not CA1, and support the validity of this model. Overall, the postmortem studies of neural circuitry abnormalities in schizophrenia are beginning to implicate specific cellular, molecular, and electrophysiological mechanism in specific subtypes of cortical neurons defined by their afferent and efferent connectivity within key corticolimbic regions.
Collapse
Affiliation(s)
- Francine M Benes
- Program in Structural and Molecular Neuroscience, McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
28
|
Functions and modulation of neuronal SK channels. Cell Biochem Biophys 2009; 55:127-39. [PMID: 19655101 DOI: 10.1007/s12013-009-9062-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Accepted: 07/15/2009] [Indexed: 10/20/2022]
Abstract
Small conductance (SK) channels are calcium-activated potassium channels that, when cloned in 1996, were thought solely to contribute to the afterhyperpolarisation that follows action potentials, and to control repetitive firing patterns of neurons. However, discoveries over the past few years have identified novel roles for SK channels in controlling dendritic excitability, synaptic transmission and synaptic plasticity. More recently, modulation of SK channel calcium sensitivity by casein kinase 2, and of SK channel trafficking by protein kinase A, have been demonstrated. This article will discuss recent findings regarding the function and modulation of SK channels in central neurons.
Collapse
|
29
|
Pedarzani P, Stocker M. Molecular and cellular basis of small--and intermediate-conductance, calcium-activated potassium channel function in the brain. Cell Mol Life Sci 2008; 65:3196-217. [PMID: 18597044 PMCID: PMC2798969 DOI: 10.1007/s00018-008-8216-x] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Small conductance calcium-activated potassium (SK or K(Ca)2) channels link intracellular calcium transients to membrane potential changes. SK channel subtypes present different pharmacology and distribution in the nervous system. The selective blocker apamin, SK enhancers and mice lacking specific SK channel subunits have revealed multifaceted functions of these channels in neurons, glia and cerebral blood vessels. SK channels regulate neuronal firing by contributing to the afterhyperpolarization following action potentials and mediating I(AHP), and partake in a calcium-mediated feedback loop with NMDA receptors, controlling the threshold for induction of hippocampal long-term potentiation. The function of distinct SK channel subtypes in different neurons often results from their specific coupling to different calcium sources. The prominent role of SK channels in the modulation of excitability and synaptic function of limbic, dopaminergic and cerebellar neurons hints at their possible involvement in neuronal dysfunction, either as part of the causal mechanism or as potential therapeutic targets.
Collapse
Affiliation(s)
- P Pedarzani
- Research Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.
| | | |
Collapse
|
30
|
Murthy SRK, Teodorescu G, Nijholt IM, Dolga AM, Grissmer S, Spiess J, Blank T. Identification and characterization of a novel, shorter isoform of the small conductance Ca2+-activated K+channel SK2. J Neurochem 2008; 106:2312-21. [DOI: 10.1111/j.1471-4159.2008.05557.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
31
|
Mercado GE, Xia SJ, Zhang C, Ahn EH, Gustafson DM, Laé M, Ladanyi M, Barr FG. Identification of PAX3-FKHR-regulated genes differentially expressed between alveolar and embryonal rhabdomyosarcoma: focus on MYCN as a biologically relevant target. Genes Chromosomes Cancer 2008; 47:510-20. [PMID: 18335505 DOI: 10.1002/gcc.20554] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Rhabdomyosarcoma is a family of myogenic soft tissue tumors subdivided into two main subtypes: alveolar (ARMS) and embryonal (ERMS). ARMS is characterized by a frequent 2;13 chromosomal translocation that creates a PAX3-FKHR fusion transcription factor. To identify downstream targets of PAX3-FKHR, we introduced an inducible form of PAX3-FKHR into human RD ERMS cells. Microarray analysis identified 39 genes (29 upregulated and 10 downregulated) that are modulated by PAX3-FKHR in RD cells and differentially expressed between ERMS and PAX3-FKHR-positive ARMS tumors. Functional annotation demonstrated that genes involved in regulation of transcription and development, particularly neurogenesis, are represented in this group. MYCN was one notable neural-related transcription factor-encoding gene identified in this set, and its regulation by PAX3-FKHR was further confirmed at the RNA and protein levels. The findings of cycloheximide inhibition and time-course studies are consistent with the hypothesis that the PAX3-FKHR protein acts directly on the MYCN gene at the transcriptional level. Functional studies established that MYCN cooperates with PAX3-FKHR to enhance oncogenic activity. In conclusion, we identified a selected set of biologically relevant genes modulated by PAX3-FKHR, and demonstrated that PAX3-FKHR contributes to the expression of MYCN and in turn MYCN collaborates with PAX3-FKHR in tumorigenesis.
Collapse
Affiliation(s)
- Gabriela E Mercado
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Wulff H, Zhorov BS. K+ channel modulators for the treatment of neurological disorders and autoimmune diseases. Chem Rev 2008; 108:1744-73. [PMID: 18476673 PMCID: PMC2714671 DOI: 10.1021/cr078234p] [Citation(s) in RCA: 176] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Heike Wulff
- Department of Pharmacology, University of California, Davis, California 95616, USA.
| | | |
Collapse
|
33
|
The expression and function of Ca(2+)-sensing receptors in rat mesenteric artery; comparative studies using a model of type II diabetes. Br J Pharmacol 2008; 154:652-62. [PMID: 18414396 DOI: 10.1038/bjp.2008.108] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND AND PURPOSE The extracellular calcium-sensing receptor (CaR) in vascular endothelial cells activates endothelial intermediate-conductance, calcium-sensitive K(+) channels (IK(Ca)) indirectly leading to myocyte hyperpolarization. We determined whether CaR expression and function was modified in a rat model of type II diabetes. EXPERIMENTAL APPROACH Pressure myography, western blotting, sharp microelectrode and K(+)-selective electrode recordings were used to investigate the functional expression of the CaR and IK(Ca) in rat mesenteric arteries. KEY RESULTS Myocyte hyperpolarization to the CaR activator calindol was inhibited by Calhex 231. U46619-induced vessel contraction elevated the extracellular [K(+)] around the myocytes, and inhibition of this 'K(+) cloud' by iberiotoxin was needed to reveal calindol-induced vasodilatations. These were antagonized by Calhex 231 and significantly smaller in Zucker diabetic fatty rat (ZDF) vessels than in Zucker lean (ZL) controls. Myocyte hyperpolarizations to calindol were also smaller in ZDF than in ZL arteries. In ZDF vessels, endothelial cell CaR protein expression was reduced; IK(Ca) expression was also diminished, but IK(Ca)-generated hyperpolarizations mediated by 1-EBIO were unaffected. CONCLUSIONS AND IMPLICATIONS The reduced CaR-mediated hyperpolarizing and vasodilator responses in ZDF arteries result from a decrease in CaR expression, rather than from a modification of IK(Ca) channels. Detection of CaR-mediated vasodilatation required the presence of iberiotoxin, suggesting a CaR contribution to vascular diameter, that is, inversely related to the degree of vasoconstriction. Compromise of the CaR pathway would favour the long-term development of a higher basal vascular tone and could contribute to the vascular complications associated with type II diabetes.
Collapse
|
34
|
Abstract
1. SK channels are small-conductance calcium-activated potassium channels that are widely expressed in neurons. The traditional view of the functional role of SK channels is in mediating one component of the after-hyperpolarization that follows action potentials. Calcium influx via voltage-gated calcium channels active during action potentials opens SK channels and the resultant hyperpolarization lowers the firing frequency of action potentials in many neurons. 2. Recent advances have shown that, in addition to controlling action potential firing frequency, SK channels are also important in regulating dendritic excitability, synaptic transmission and synaptic plasticity. 3. In accordance with their role in modulating synaptic plasticity, SK channels are also important in regulating several learning and memory tasks and may also play a role in a number of neurological disorders. 4. The present review discusses recent findings on the role of SK channels in central neurons.
Collapse
Affiliation(s)
- E S Louise Faber
- Queensland Brain Institute, University of Queensland, St Lucia, Queensland, Australia.
| | | |
Collapse
|
35
|
Brainard AM, Korovkina VP, England SK. Potassium channels and uterine function. Semin Cell Dev Biol 2007; 18:332-9. [PMID: 17596977 PMCID: PMC2012947 DOI: 10.1016/j.semcdb.2007.05.008] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2007] [Accepted: 05/03/2007] [Indexed: 01/14/2023]
Abstract
Ion channels are effector proteins that mediate uterine excitability throughout gestation. This review will focus primarily on the role of potassium channels in regulating myometrial tone in pregnancy and labor contractions. During gestation, potassium channels maintain the uterus in a state of quiescence by contributing to the resting membrane potential and counteracting contractile stimuli. This review summarizes the current knowledge about the significance of the potassium channels in maintaining a normal gestational period and initiating labor contractions at term.
Collapse
Affiliation(s)
- Adam M. Brainard
- University of Iowa Carver College of Medicine, Department of Molecular Physiology and Biophysics, Iowa City, IA 52242, Phone: (319) 335-7860, FAX: (319) 335-7330,
| | - Victoria P. Korovkina
- University of Iowa Carver College of Medicine, Department of Molecular Physiology and Biophysics, Iowa City, IA 52242, Phone: (319) 335-7860, FAX: (319) 335-7330,
| | - Sarah K. England
- University of Iowa Carver College of Medicine, Department of Molecular Physiology and Biophysics, Iowa City, IA 52242, Phone: (319) 335-7860, FAX: (319) 335-7330,
| |
Collapse
|
36
|
Decimo I, Roncarati R, Grasso S, Clemens M, Chiamulera C, Fumagalli G. SK3 trafficking in hippocampal cells: the role of different molecular domains. Biosci Rep 2007; 26:399-412. [PMID: 17061167 DOI: 10.1007/s10540-006-9029-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
The regulative steps that control trafficking of ion channels are fundamental determinants of their qualitative and quantitative expression on the cell membrane. In this work the trafficking of the small conductance calcium-activated potassium channel, SK3 was studied in neurons in order to identify relevant molecular domains involved in this process. Hippocampal cell cultures were transfected with fusion proteins of green fluorescent protein (GFP) and different SK3 subunit truncations. The differential distribution of the mutants was analyzed by confocal microscopy and compared to the localization of the control fusion protein with full length SK3. The transport of chimeric proteins was quantified from fluorescence images by developing a morphometric analytical method. We found that the full length SK3 was distributed in cell body, axon and dendrites, whereas the deleted forms GFPDelta578-736 (deletion of the entire C-terminal domain), GFPDeltaCaMBD (deletion of the calmodulin-binding site) and GFPDeltaN (deletion of the N-terminal domain) were not transported into cell processes but accumulated in the cell body. The GFPDelta640-736 (deletion of the distal C-terminal domain) showed a distribution similar to control. The quantification and statistical analysis confirmed the differences in distribution across the three groups. In conclusion, the current work provides evidence for a fundamental role of the N-terminal domain and the calmodulin binding domain in SK3 trafficking in neurons.
Collapse
Affiliation(s)
- Ilaria Decimo
- Department of Medicine and Public Health, University of Verona, P.le L. Scuro 10, 37100 Verona, Italy.
| | | | | | | | | | | |
Collapse
|
37
|
Potier M, Joulin V, Roger S, Besson P, Jourdan ML, Leguennec JY, Bougnoux P, Vandier C. Identification of SK3 channel as a new mediator of breast cancer cell migration. Mol Cancer Ther 2006; 5:2946-53. [PMID: 17121942 DOI: 10.1158/1535-7163.mct-06-0194] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Potassium channels have been involved in epithelial tumorigenesis but the role of small-conductance Ca(2+)-activated K(+) channels is unknown. We report here that small-conductance Ca(2+)-activated K(+) channels are expressed in a highly metastasizing mammary cancer cell line, MDA-MB-435s. Patch-clamp recordings showed typical small-conductance Ca(2+)-activated K(+) channel-mediated currents sensitive to apamin, 4-aminopyridine, and tetraethylammonium. Moreover, the cells displayed a high intracellular calcium concentration, which was decreased after 24 hours of apamin treatment. By regulating membrane potential and intracellular calcium concentration, these channels were involved in MDA-MB-435s cell migration, but not in proliferation. Only SK3 protein expression was observed in these cells in contrast to SK2, which was expressed both in cancer and noncancer cell lines. Whereas small interfering RNA directed against SK3 almost totally abolished MDA-MB-435s cell migration, transient expression of SK3 increased migration of the SK3-deficient cell lines, MCF-7 and 184A1. SK3 channel was solely expressed in tumor breast biopsies and not in nontumor breast tissues. Thus, SK3 protein channel seems to be a new mediator of breast cancer cell migration and represents a potential target for a new class of anticancer agents.
Collapse
Affiliation(s)
- Marie Potier
- Inserm E-0211, Nutrition Croissance et Cancer, University Francois Rabelais, 10 boulevard Tonnelle, Tours, F-37000 France
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Gargus JJ. Ion channel functional candidate genes in multigenic neuropsychiatric disease. Biol Psychiatry 2006; 60:177-85. [PMID: 16497276 DOI: 10.1016/j.biopsych.2005.12.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2005] [Revised: 11/15/2005] [Accepted: 12/15/2005] [Indexed: 10/25/2022]
Abstract
Scores of monogenic Mendelian ion channel diseases serve to anchor the pathophysiology of the channelopathies, but there are also now clear examples of environmental, pharmacogenetic, and acquired channelopathy mechanisms. The cardinal feature of heritable ion channel disease is a periodic disturbance of rhythmic function in constitutionally hyperexcitable tissue. While the complexity of neuroanatomy obscures functional analysis of mutations causing monogenic seizure, ataxia, or migraine syndromes, extrapolation from the cardiac (Long QT [LQT]) and muscle (Periodic Paralysis) channelopathy syndromes provides a simplified predictive framework of molecular pathology: electrically stabilizing potassium ion (K(+)) and chloride ion (Cl(-)) channels, likely having lesions that diminish their current, and excitatory Na(+) channels, likely having gain-of-function lesions. The voltage-gated calcium channel gene family that contains CACNA1C, the newest LQT locus, causing Timothy Syndrome with a phenotype including autism, has proven to be particularly informative for its members' ability to tie the various central nervous system (CNS) phenotypes together in an interpretable fashion, now including direct extension to the classically multigenic neuropsychiatric phenotypes. Features of a promising ion channel candidate gene arise from its broad locus, gene family, nature of alleles, physiology and pharmacology, tissue expression profile, and phenotype in model organisms. KCNN3 is explored as a paradigm to consider.
Collapse
Affiliation(s)
- J Jay Gargus
- Department of Physiology, Section of Human Genetics, University of California, Irvine, California 92697-4034, USA.
| |
Collapse
|
39
|
Abstract
Decades of research have demonstrated that anorexia nervosa (AN) may be associated with aberrant cognition, yet, its role in maintaining stringent dieting has received relatively little attention from mainstream researchers of eating disorders. The purpose of the present article is to highlight cognitive ('top-down') factors that are considered responsible for anticipatory anxiety of stoutness and frank fat-phobia (laparophobia). A cognitive model proposed departs from the formulation suggesting that phobia of over-eating is superimposed on avoidant tendencies ('environmental autonomy syndrome'), whereas excessive exercising becomes a natural coping strategy with laparophobia, an instrument of reward. AN ideation involves complex neuronal circuitries and multiple neurochemical components that may conceivably represent a mirror image of those underlying obesity. The emphasis on phobia and aberrant membrane excitability akin to channelopathies behoves the clinicians to be aware of potential uses of drugs acting at the gamma-aminobutyric acid and the N-methyl-D-aspartate/AMPA [2-amino-3-(3-hydroxy-5-methylisoxazol-4-yl) propionic acid] receptors sites as the adjuncts to conventional agents in managing AN.
Collapse
|
40
|
Tuteja D, Xu D, Timofeyev V, Lu L, Sharma D, Zhang Z, Xu Y, Nie L, Vázquez AE, Young JN, Glatter KA, Chiamvimonvat N. Differential expression of small-conductance Ca2+-activated K+ channels SK1, SK2, and SK3 in mouse atrial and ventricular myocytes. Am J Physiol Heart Circ Physiol 2005; 289:H2714-23. [PMID: 16055520 DOI: 10.1152/ajpheart.00534.2005] [Citation(s) in RCA: 187] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Small-conductance Ca2+-activated K+ channels (SK channels, KCa channels) have been reported in excitable cells, where they aid in integrating changes in intracellular Ca2+ with membrane potential. We recently reported for the first time the functional existence of SK2 (KCa2.2) channels in human and mouse cardiac myocytes. Here, we report cloning of SK1 (KCa2.1) and SK3 (KCa2.3) channels from mouse atria and ventricles using RT-PCR. Full-length transcripts and their variants were detected for both SK1 and SK3 channels. Variants of mouse SK1 channel (mSK1) differ mainly in the COOH-terminal structure, affecting a portion of the sixth transmembrane segment (S6) and the calmodulin binding domain (CaMBD). Mouse SK3 channel (mSK3) differs not only in the number of polyglutamine repeats in the NH2 terminus but also in the intervening sequences between the polyglutamine repeats. Full-length cardiac mSK1 and mSK3 show 99 and 91% nucleotide identity with those of mouse colon SK1 and SK3, respectively. Quantification of SK1, SK2, and SK3 transcripts between atria and ventricles was performed using real-time quantitative RT-PCR from single, isolated cardiomyocytes. SK1 transcript was found to be more abundant in atria compared with ventricles, similar to the previously reported finding for SK2 channel. In contrast, SK3 showed similar levels of expression in atria and ventricles. Together, our data are the first to indicate the presence of the three different isoforms of SK channels in heart and the differential expression of SK1 and SK2 in mouse atria and ventricles. Because of the marked differential expression of SK channel isoforms in heart, specific ligands for Ca2+-activated K+ currents may offer a unique therapeutic opportunity to modify atrial cells without interfering with ventricular myocytes.
Collapse
Affiliation(s)
- Dipika Tuteja
- Division of Cardiovascular Medicine, Dept. of Medicine, Univ. of California, Davis, Genome and Biomedical Sciences Facility, 451 East Health Sciences Drive, Rm. 6315, Davis, CA 95616, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Roncarati R, Decimo I, Fumagalli G. Assembly and trafficking of human small conductance Ca2+-activated K+ channel SK3 are governed by different molecular domains. Mol Cell Neurosci 2005; 28:314-25. [PMID: 15691712 DOI: 10.1016/j.mcn.2004.09.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2004] [Revised: 04/17/2004] [Accepted: 07/12/2004] [Indexed: 12/30/2022] Open
Abstract
Intracellular trafficking is an important event in the control of type and number of ion channels expressed on the cell surface. In this study, we have identified molecular domains involved in assembly and trafficking of the human small conductance Ca2+-activated K+ channel SK3. Deletion of the N-terminus, the C-terminus, or the calmodulin-binding domain (CaMBD) led to retention of SK3 channels in the endoplasmic reticulum. Presence of the CaMBD allowed trafficking to the Golgi complex, and sequences downstream were required for efficient transport to the plasma membrane, suggesting several steps in the control of SK3 forward trafficking. Co-immunoprecipitation studies demonstrated that SK3 subunits lacking the N-terminus, the CaMBD, or the distal C-terminus, but not the entire C-terminus, were able to oligomerize with wild-type SK3 subunits. Thus, these two C-terminal regions of SK3 seem to contribute to assembly and trafficking of channels whereas the N-terminus is necessary for trafficking but not sufficient for oligomerization.
Collapse
Affiliation(s)
- Renza Roncarati
- Department of Medicine and Public Health, Section of Pharmacology, University of Verona, 37134 Verona, Italy.
| | | | | |
Collapse
|
42
|
Stocker M. Ca2+-activated K+ channels: molecular determinants and function of the SK family. Nat Rev Neurosci 2004; 5:758-70. [PMID: 15378036 DOI: 10.1038/nrn1516] [Citation(s) in RCA: 407] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ca(2+)-activated K(+) (K(Ca)) channels of small (SK) and intermediate (IK) conductance are present in a wide range of excitable and non-excitable cells. On activation by low concentrations of Ca(2+), they open, which results in hyperpolarization of the membrane potential and changes in cellular excitability. K(Ca)-channel activation also counteracts further increases in intracellular Ca(2+), thereby regulating the concentration of this ubiquitous intracellular messenger in space and time. K(Ca) channels have various functions, including the regulation of neuronal firing properties, blood flow and cell proliferation. The cloning of SK and IK channels has prompted investigations into their gating, pharmacology and organization into calcium-signalling domains, and has provided a framework that can be used to correlate molecularly identified K(Ca) channels with their native currents.
Collapse
Affiliation(s)
- Martin Stocker
- Wellcome Laboratory for Molecular Pharmacology, Department of Pharmacology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
43
|
Stankovich L, Wicks D, Despotovski S, Liang D. Atomic Absorption Spectroscopy in Ion Channel Screening. Assay Drug Dev Technol 2004; 2:569-74. [PMID: 15671655 DOI: 10.1089/adt.2004.2.569] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
This article examines the utility of atomic absorption spectroscopy, in conjunction with cold flux assays, to ion channel screening. The multiplicity of ion channels that can be interrogated using cold flux assays and atomic absorption spectroscopy is summarized. The importance of atomic absorption spectroscopy as a screening tool is further elaborated upon by providing examples of the relevance of ion channels to various physiological processes and targeted diseases.
Collapse
Affiliation(s)
- Larisa Stankovich
- Aurora Biomed Inc., 1001 East Pender Street, Vancouver, BC, Canada V6A 1W2.
| | | | | | | |
Collapse
|
44
|
Villalobos C, Shakkottai VG, Chandy KG, Michelhaugh SK, Andrade R. SKCa channels mediate the medium but not the slow calcium-activated afterhyperpolarization in cortical neurons. J Neurosci 2004; 24:3537-42. [PMID: 15071101 PMCID: PMC6729743 DOI: 10.1523/jneurosci.0380-04.2004] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Many neurons, including pyramidal cells of the cortex, express a slow afterhyperpolarization (sAHP) that regulates their firing. Although initial findings suggested that the current underlying the sAHP could be carried through SK(Ca) channels, recent work has uncovered anomalies that are not congruent with this idea. Here, we used overexpression and dominant-negative strategies to assess the involvement of SK(Ca) channels in mediating the current underlying the sAHP in pyramidal cells of the cerebral cortex. Pyramidal cells of layer V exhibit robust AHP currents composed of two kinetically and pharmacologically distinguishable currents known as the medium AHP current (I(mAHP)) and the slow AHP current (I(sAHP)). I(mAHP) is blocked by the SK(Ca) channel blockers apamin and bicuculline, whereas I(sAHP) is resistant to these agents but is inhibited by activation of muscarinic receptors. To test for a role for SK(Ca) channels, we overexpressed K(Ca)2.1 (SK1) and K(Ca)2.2 (SK2), the predominant SK(Ca) subunits expressed in the cortex, in pyramidal cells of cultured brain slices. Overexpression of K(Ca)2.1 and K(Ca)2.2 resulted in a fourfold to fivefold increase in the amplitude of I(mAHP) but had no detectable effect on I(sAHP). As an additional test, we examined I(sAHP) in a transgenic mouse expressing a truncated SK(Ca) subunit (SK3-1B) capable of acting as a dominant negative for the entire family of SK(Ca)-IK(Ca) channels. Expression of SK3-1B profoundly inhibited I(mAHP) but again had no discernable effect on I(sAHP). These results are inconsistent with the proposal that SK(Ca) channels mediate I(sAHP) in pyramidal cells and indicate that a different potassium channel mediates this current.
Collapse
Affiliation(s)
- Claudio Villalobos
- Department of Psychiatry and Behavioral Neuroscience, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | | | | | | | |
Collapse
|
45
|
Manaves V, Qin W, Bauer AL, Rossie S, Kobayashi M, Rane SG. Calcium and Vitamin D increase mRNA levels for the growth control hIK1 channel in human epidermal keratinocytes but functional channels are not observed. BMC DERMATOLOGY 2004; 4:7. [PMID: 15200683 PMCID: PMC446203 DOI: 10.1186/1471-5945-4-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2004] [Accepted: 06/16/2004] [Indexed: 11/17/2022]
Abstract
Background Intermediate-conductance, calcium-activated potassium channels (IKs) modulate proliferation and differentiation in mesodermal cells by enhancing calcium influx, and they contribute to the physiology of fluid movement in certain epithelia. Previous reports suggest that IK channels stimulate proliferative growth in a keratinocyte cell line; however, because these channels indirectly promote calcium influx, a critically unique component of the keratinocyte differentiation program, an alternative hypothesis is that they would be anti-proliferative and pro-differentiating. This study addresses these hypotheses. Methods Real-time PCR, patch clamp electrophysiology, and proliferation assays were used to determine if human IK1 (hIK1) expression and function are correlated with either proliferation or differentiation in cultured human skin epidermal keratinocytes, and skin biopsies grown in explant culture. Results hIK1 mRNA expression in human keratinocytes and skin was increased in response to anti-proliferative/pro-differentiating stimuli (elevated calcium and Vitamin D). Correspondingly, the hIK1 agonist 1-EBIO inhibited keratinocyte proliferation suggesting that the channel could be anti-proliferative and pro-differentiating. However, this proliferative inhibition by 1-EBIO was not reversed by a panel of hIK1 blockers, calling into question the mechanism of 1-EBIO action. Subsequent patch clamp electrophysiological analysis failed to detect hIK1 channel currents in keratinocytes, even those expressing substantial hIK1 mRNA in response to calcium and Vitamin D induced differentiation. Identical electrophysiological recording conditions were then used to observe robust IK1 currents in fibroblasts which express IK1 mRNA levels comparable to those of keratinocytes. Thus, the absence of observable hIK1 currents in keratinocytes was not a function of the electrophysiological techniques. Conclusion Human keratinocyte differentiation is stimulated by calcium mobilization and influx, and differentiation stimuli coordinately upregulate mRNA levels of the calcium-activated hIK1 channel. This upregulation is paradoxical in that functional hIK1 channels are not observed in cultured keratinocytes. It appears, therefore, that hIK1 does not contribute to the functional electrophysiology of primary human keratinocytes, nor intact human skin. Further, the results indicate caution is required when interpreting experiments utilizing pharmacological hIK1 modulators in human keratinocytes.
Collapse
Affiliation(s)
- Vlasios Manaves
- Molecular Human Physiology Group, Fujisawa Research Institute of America, Evanston, IL 60201 USA
| | - Wuxuan Qin
- Molecular Human Physiology Group, Fujisawa Research Institute of America, Evanston, IL 60201 USA
| | - Amy L Bauer
- Molecular Human Physiology Group, Fujisawa Research Institute of America, Evanston, IL 60201 USA
| | - Sandra Rossie
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907 USA
| | | | - Stanley G Rane
- Molecular Human Physiology Group, Fujisawa Research Institute of America, Evanston, IL 60201 USA
- Depts. of Medicine, Evanston Northwestern Healthcare, Evanston, IL 60201, and Northwestern University Medical School, Chicago, IL 60611 USA
| |
Collapse
|
46
|
Abstract
Ataxia is a lethal neurological disease characterized by incoordination, postural abnormalities, difficulties with gait, and problems with clarity of speech. The etiology of ataxia is divided equally between hereditary and sporadic forms. Regardless of cause, the cerebellar cortex is often a target in ataxia. Thus, how a disruption in cerebellar cortex might lead to ataxia is of considerable interest. A report in this issue of the JCI links ataxia to enhanced hyperexcitability of neurons in the deep cerebellar nuclei.
Collapse
Affiliation(s)
- Harry T Orr
- Institute of Human Genetics, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
47
|
Shakkottai VG, Chou CH, Oddo S, Sailer CA, Knaus HG, Gutman GA, Barish ME, LaFerla FM, Chandy KG. Enhanced neuronal excitability in the absence of neurodegeneration induces cerebellar ataxia. J Clin Invest 2004; 113:582-90. [PMID: 14966567 PMCID: PMC338266 DOI: 10.1172/jci20216] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2003] [Accepted: 11/25/2003] [Indexed: 11/17/2022] Open
Abstract
Cerebellar ataxia, a devastating neurological disease, may be initiated by hyperexcitability of deep cerebellar nuclei (DCN) secondary to loss of inhibitory input from Purkinje neurons that frequently degenerate in this disease. This mechanism predicts that intrinsic DCN hyperexcitability would cause ataxia in the absence of upstream Purkinje degeneration. We report the generation of a transgenic (Tg) model that supports this mechanism of disease initiation. Small-conductance calcium-activated potassium (SK) channels, regulators of firing frequency, were silenced in the CNS of Tg mice with the dominant-inhibitory construct SK3-1B-GFP. Transgene expression was restricted to the DCN within the cerebellum and was detectable beginning on postnatal day 10, concomitant with the onset of cerebellar ataxia. Neurodegeneration was not evident up to the sixth month of age. Recordings from Tg DCN neurons revealed loss of the apamin-sensitive after-hyperpolarization current (IAHP) and increased spontaneous firing through SK channel suppression, indicative of DCN hyperexcitability. Spike duration and other electrogenic conductance were unaffected. Thus, a purely electrical alteration is sufficient to cause cerebellar ataxia, and SK openers such as the neuroprotective agent riluzole may reduce neuronal hyperexcitability and have therapeutic value. This dominant-inhibitory strategy may help define the in vivo role of SK channels in other neuronal pathways.
Collapse
Affiliation(s)
- Vikram G Shakkottai
- Department of Physiology and Biophysics, University of California Irvine, 92697, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Shakkottai VG, Chou CH, Oddo S, Sailer CA, Knaus HG, Gutman GA, Barish ME, LaFerla FM, Chandy KG. Enhanced neuronal excitability in the absence of neurodegeneration induces cerebellar ataxia. J Clin Invest 2004. [DOI: 10.1172/jci200420216] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
49
|
|
50
|
Kolski-Andreaco A, Tomita H, Shakkottai VG, Gutman GA, Cahalan MD, Gargus JJ, Chandy KG. SK3-1C, a Dominant-negative Suppressor of SKCa and IKCa Channels. J Biol Chem 2004; 279:6893-904. [PMID: 14638680 DOI: 10.1074/jbc.m311725200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Small conductance Ca2+-activated K+ channels, products of the SK1-SK3 genes, regulate membrane excitability both within and outside the nervous system. We report the characterization of a SK3 variant (SK3-1C) that differs from SK3 by utilizing an alternative first exon (exon 1C) in place of exon 1A used by SK3, but is otherwise identical to SK3. Quantitative RT-PCR detected abundant expression of SK3-1C transcripts in human lymphoid tissues, skeletal muscle, trachea, and salivary gland but not the nervous system. SK3-1C did not produce functional channels when expressed alone in mammalian cells, but suppressed SK1, SK2, SK3, and IKCa1 channels, but not BKCa or KV channels. Confocal microscopy revealed that SK3-1C sequestered SK3 protein intracellularly. Dominant-inhibitory activity of SK3-1C was not due to a nonspecific calmodulin sponge effect since overexpression of calmodulin did not reverse SK3-1C-mediated intracellular trapping of SK3 protein, and calmodulin-Ca2+-dependent inactivation of CaV channels was not affected by SK3-1C overexpression. Deletion analysis identified a dominant-inhibitory segment in the SK3-1C C terminus that resembles tetramerization-coiled-coiled domains reported to enhance tetramer stability and selectivity of multimerization of many K+ channels. SK3-1C may therefore suppress calmodulin-gated SKCa/IKCa channels by trapping these channel proteins intracellularly via subunit interactions mediated by the dominant-inhibitory segment and thereby reduce functional channel expression on the cell surface. Such family-wide dominant-negative suppression by SK3-1C provides a powerful mechanism to titrate membrane excitability and is a useful approach to define the functional in vivo role of these channels in diverse tissues by their targeted silencing.
Collapse
Affiliation(s)
- Aaron Kolski-Andreaco
- Department of Physiology and Biophysics, University of California Irvine, Irvine, California 92697, USA
| | | | | | | | | | | | | |
Collapse
|