1
|
Jansen LAR, Welch MA, Plant LD, Baro DJ. Crosstalk between PKA and PIAS3 regulates cardiac Kv4 channel SUMOylation. Cell Commun Signal 2024; 22:422. [PMID: 39223673 PMCID: PMC11367828 DOI: 10.1186/s12964-024-01795-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Post-translational SUMOylation of nuclear and cytosolic proteins maintains homeostasis in eukaryotic cells and orchestrates programmed responses to changes in metabolic demand or extracellular stimuli. In excitable cells, SUMOylation tunes the biophysical properties and trafficking of ion channels. Ion channel SUMOylation status is determined by the opposing enzyme activities of SUMO ligases and deconjugases. Phosphorylation also plays a permissive role in SUMOylation. SUMO deconjugases have been identified for several ion channels, but their corresponding E3 ligases remain unknown. This study shows PIAS3, a.k.a. KChAP, is a bona fide SUMO E3 ligase for Kv4.2 and HCN2 channels in HEK cells, and endogenous Kv4.2 and Kv4.3 channels in cardiomyocytes. PIAS3-mediated SUMOylation at Kv4.2-K579 increases channel surface expression through a rab11a-dependent recycling mechanism. PKA phosphorylation at Kv4.2-S552 reduces the current mediated by Kv4 channels in HEK293 cells, cardiomyocytes, and neurons. This study shows PKA mediated phosphorylation blocks Kv4.2-K579 SUMOylation in HEK cells and cardiomyocytes. Together, these data identify PIAS3 as a key downstream mediator in signaling cascades that control ion channel surface expression.
Collapse
Affiliation(s)
| | - Meghyn A Welch
- Department of Biology, Georgia State University, Atlanta, GA, USA
- Present Address: Section on Molecular Neurophysiology and Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Leigh D Plant
- Department of Pharmaceutical Sciences, Center for Drug Discovery, Northeastern University, Boston, MA, USA
| | - Deborah J Baro
- Department of Biology, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
2
|
Anees P, Saminathan A, Rozmus ER, Di A, Malik AB, Delisle BP, Krishnan Y. Detecting organelle-specific activity of potassium channels with a DNA nanodevice. Nat Biotechnol 2024; 42:1065-1074. [PMID: 37735264 PMCID: PMC11021130 DOI: 10.1038/s41587-023-01928-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 07/31/2023] [Indexed: 09/23/2023]
Abstract
Cell surface potassium ion (K+) channels regulate nutrient transport, cell migration and intercellular communication by controlling K+ permeability and are thought to be active only at the plasma membrane. Although these channels transit the trans-Golgi network, early and recycling endosomes, whether they are active in these organelles is unknown. Here we describe a pH-correctable, ratiometric reporter for K+ called pHlicKer, use it to probe the compartment-specific activity of a prototypical voltage-gated K+ channel, Kv11.1, and show that this cell surface channel is active in organelles. Lumenal K+ in organelles increased in cells expressing wild-type Kv11.1 channels but not after treatment with current blockers. Mutant Kv11.1 channels, with impaired transport function, failed to increase K+ levels in recycling endosomes, an effect rescued by pharmacological correction. By providing a way to map the organelle-specific activity of K+ channels, pHlicKer technology could help identify new organellar K+ channels or channel modulators with nuanced functions.
Collapse
Affiliation(s)
- Palapuravan Anees
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
- Grossman Center for Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, USA
- Institute of Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
| | - Anand Saminathan
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
- Grossman Center for Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, USA
| | - Ezekiel R Rozmus
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Anke Di
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of Medicine, Chicago, IL, USA
| | - Asrar B Malik
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of Medicine, Chicago, IL, USA
| | - Brian P Delisle
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA.
| | - Yamuna Krishnan
- Department of Chemistry, The University of Chicago, Chicago, IL, USA.
- Grossman Center for Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, USA.
- Institute of Biophysical Dynamics, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
3
|
Maraslioglu-Sperber A, Pizzi E, Fisch JO, Kattler K, Ritter T, Friauf E. Molecular and functional profiling of cell diversity and identity in the lateral superior olive, an auditory brainstem center with ascending and descending projections. Front Cell Neurosci 2024; 18:1354520. [PMID: 38846638 PMCID: PMC11153811 DOI: 10.3389/fncel.2024.1354520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/15/2024] [Indexed: 06/09/2024] Open
Abstract
The lateral superior olive (LSO), a prominent integration center in the auditory brainstem, contains a remarkably heterogeneous population of neurons. Ascending neurons, predominantly principal neurons (pLSOs), process interaural level differences for sound localization. Descending neurons (lateral olivocochlear neurons, LOCs) provide feedback into the cochlea and are thought to protect against acoustic overload. The molecular determinants of the neuronal diversity in the LSO are largely unknown. Here, we used patch-seq analysis in mice at postnatal days P10-12 to classify developing LSO neurons according to their functional and molecular profiles. Across the entire sample (n = 86 neurons), genes involved in ATP synthesis were particularly highly expressed, confirming the energy expenditure of auditory neurons. Two clusters were identified, pLSOs and LOCs. They were distinguished by 353 differentially expressed genes (DEGs), most of which were novel for the LSO. Electrophysiological analysis confirmed the transcriptomic clustering. We focused on genes affecting neuronal input-output properties and validated some of them by immunohistochemistry, electrophysiology, and pharmacology. These genes encode proteins such as osteopontin, Kv11.3, and Kvβ3 (pLSO-specific), calcitonin-gene-related peptide (LOC-specific), or Kv7.2 and Kv7.3 (no DEGs). We identified 12 "Super DEGs" and 12 genes showing "Cluster similarity." Collectively, we provide fundamental and comprehensive insights into the molecular composition of individual ascending and descending neurons in the juvenile auditory brainstem and how this may relate to their specific functions, including developmental aspects.
Collapse
Affiliation(s)
- Ayse Maraslioglu-Sperber
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Erika Pizzi
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Jonas O. Fisch
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Kathrin Kattler
- Genetics/Epigenetics Group, Department of Biological Sciences, Saarland University, Saarbrücken, Germany
| | - Tamara Ritter
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Eckhard Friauf
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| |
Collapse
|
4
|
Huang LS, Anas M, Xu J, Zhou B, Toth PT, Krishnan Y, Di A, Malik AB. Endosomal trafficking of two-pore K + efflux channel TWIK2 to plasmalemma mediates NLRP3 inflammasome activation and inflammatory injury. eLife 2023; 12:e83842. [PMID: 37158595 PMCID: PMC10202452 DOI: 10.7554/elife.83842] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 05/07/2023] [Indexed: 05/10/2023] Open
Abstract
Potassium efflux via the two-pore K+ channel TWIK2 is a requisite step for the activation of NLRP3 inflammasome, however, it remains unclear how K+ efflux is activated in response to select cues. Here, we report that during homeostasis, TWIK2 resides in endosomal compartments. TWIK2 is transported by endosomal fusion to the plasmalemma in response to increased extracellular ATP resulting in the extrusion of K+. We showed that ATP-induced endosomal TWIK2 plasmalemma translocation is regulated by Rab11a. Deleting Rab11a or ATP-ligated purinergic receptor P2X7 each prevented endosomal fusion with the plasmalemma and K+ efflux as well as NLRP3 inflammasome activation in macrophages. Adoptive transfer of Rab11a-depleted macrophages into mouse lungs prevented NLRP3 inflammasome activation and inflammatory lung injury. We conclude that Rab11a-mediated endosomal trafficking in macrophages thus regulates TWIK2 localization and activity at the cell surface and the downstream activation of the NLRP3 inflammasome. Results show that endosomal trafficking of TWIK2 to the plasmalemma is a potential therapeutic target in acute or chronic inflammatory states.
Collapse
Affiliation(s)
- Long Shuang Huang
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of MedicineChicagoUnited States
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong UniversityShanghaiChina
| | - Mohammad Anas
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of MedicineChicagoUnited States
| | - Jingsong Xu
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of MedicineChicagoUnited States
| | - Bisheng Zhou
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of MedicineChicagoUnited States
| | - Peter T Toth
- Fluorescence Imaging Core, The University of Illinois College of MedicineChicagoUnited States
| | - Yamuna Krishnan
- Department of Chemistry, University of ChicagoChicagoUnited States
| | - Anke Di
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of MedicineChicagoUnited States
| | - Asrar B Malik
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of MedicineChicagoUnited States
| |
Collapse
|
5
|
Zhang H, Graham V, Nepliouev I, Stiber JA, Rosenberg P. STIM1 interacts with HCN4 channels to coordinate diastolic depolarization in the mouse Sinoatrial node. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.03.539287. [PMID: 37205552 PMCID: PMC10187156 DOI: 10.1101/2023.05.03.539287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Cardiomyocytes in the sinoatrial node (SAN) are specialized to undergo spontaneous diastolic depolarization (DD) to create action potentials (AP) that serve as the origin of the heartbeat. Two cellular clocks govern DD: the membrane clock where ion channels contribute ionic conductance to create DD and the Ca 2+ clock where rhythmic Ca 2+ release from sarcoplasmic reticulum (SR) during diastole contributes pacemaking. How the membrane and Ca 2+ clocks interact to synchronize and drive DD is not well understood. Here, we identified stromal interaction molecule 1 (STIM1), the activator of store operated Ca 2+ entry (SOCE), in the P-cell cardiomyocytes of the SAN. Functional studies from STIM1 KO mice reveal dramatic changes in properties of AP and DD. Mechanistically, we show that STIM1 regulates the funny currents and HCN4 channels that are required to initiate DD and maintain sinus rhythm in mice. Taken together, our studies suggest that STIM1 acts as a sensor for both the Ca 2+ and membrane clocks for mouse SAN for cardiac pacemaking.
Collapse
|
6
|
Genetics and Molecular Basis of Congenital Heart Defects in Down Syndrome: Role of Extracellular Matrix Regulation. Int J Mol Sci 2023; 24:ijms24032918. [PMID: 36769235 PMCID: PMC9918028 DOI: 10.3390/ijms24032918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Down syndrome (DS), a complex disorder that is caused by the trisomy of chromosome 21 (Hsa21), is a major cause of congenital heart defects (CHD). Interestingly, only about 50% of individuals with Hsa21 trisomy manifest CHD. Here we review the genetic basis of CHD in DS, focusing on genes that regulate extracellular matrix (ECM) organization. The overexpression of Hsa21 genes likely underlies the molecular mechanisms that contribute to CHD, even though the genes responsible for CHD could only be located in a critical region of Hsa21. A role in causing CHD has been attributed not only to protein-coding Hsa21 genes, but also to genes on other chromosomes, as well as miRNAs and lncRNAs. It is likely that the contribution of more than one gene is required, and that the overexpression of Hsa21 genes acts in combination with other genetic events, such as specific mutations or polymorphisms, amplifying their effect. Moreover, a key function in determining alterations in cardiac morphogenesis might be played by ECM. A large number of genes encoding ECM proteins are overexpressed in trisomic human fetal hearts, and many of them appear to be under the control of a Hsa21 gene, the RUNX1 transcription factor.
Collapse
|
7
|
Yang HQ, Echeverry FA, ElSheikh A, Gando I, Anez Arredondo S, Samper N, Cardozo T, Delmar M, Shyng SL, Coetzee WA. Subcellular trafficking and endocytic recycling of K ATP channels. Am J Physiol Cell Physiol 2022; 322:C1230-C1247. [PMID: 35508187 PMCID: PMC9169827 DOI: 10.1152/ajpcell.00099.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/27/2022] [Accepted: 04/30/2022] [Indexed: 11/22/2022]
Abstract
Sarcolemmal/plasmalemmal ATP-sensitive K+ (KATP) channels have key roles in many cell types and tissues. Hundreds of studies have described how the KATP channel activity and ATP sensitivity can be regulated by changes in the cellular metabolic state, by receptor signaling pathways and by pharmacological interventions. These alterations in channel activity directly translate to alterations in cell or tissue function, that can range from modulating secretory responses, such as insulin release from pancreatic β-cells or neurotransmitters from neurons, to modulating contractile behavior of smooth muscle or cardiac cells to elicit alterations in blood flow or cardiac contractility. It is increasingly becoming apparent, however, that KATP channels are regulated beyond changes in their activity. Recent studies have highlighted that KATP channel surface expression is a tightly regulated process with similar implications in health and disease. The surface expression of KATP channels is finely balanced by several trafficking steps including synthesis, assembly, anterograde trafficking, membrane anchoring, endocytosis, endocytic recycling, and degradation. This review aims to summarize the physiological and pathophysiological implications of KATP channel trafficking and mechanisms that regulate KATP channel trafficking. A better understanding of this topic has potential to identify new approaches to develop therapeutically useful drugs to treat KATP channel-related diseases.
Collapse
Affiliation(s)
- Hua-Qian Yang
- Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China
| | | | - Assmaa ElSheikh
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon
- Department of Medical Biochemistry, Tanta University, Tanta, Egypt
| | - Ivan Gando
- Department of Pathology, NYU School of Medicine, New York, New York
| | | | - Natalie Samper
- Department of Pathology, NYU School of Medicine, New York, New York
| | - Timothy Cardozo
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
| | - Mario Delmar
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
- Department of Medicine, NYU School of Medicine, New York, New York
| | - Show-Ling Shyng
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon
| | - William A Coetzee
- Department of Pathology, NYU School of Medicine, New York, New York
- Department of Neuroscience & Physiology, NYU School of Medicine, New York, New York
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
| |
Collapse
|
8
|
Disease-associated HCN4 V759I variant is not sufficient to impair cardiac pacemaking. Pflugers Arch 2020; 472:1733-1742. [PMID: 33095298 PMCID: PMC7691308 DOI: 10.1007/s00424-020-02481-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/11/2020] [Accepted: 10/16/2020] [Indexed: 11/16/2022]
Abstract
The hyperpolarization-activated cation current If is a key determinant for cardiac pacemaker activity. It is conducted by subunits of the hyperpolarization-activated cyclic nucleotide–gated (HCN) channel family, of which HCN4 is predominant in mammalian heart. Both loss-of-function and gain-of-function mutations of the HCN4 gene are associated with sinus node dysfunction in humans; however, their functional impact is not fully understood yet. Here, we sought to characterize a HCN4 V759I variant detected in a patient with a family history of sick sinus syndrome. The genomic analysis yielded a mono-allelic HCN4 V759I variant in a 49-year-old woman presenting with a family history of sick sinus syndrome. This HCN4 variant was previously classified as putatively pathogenic because genetically linked to sudden infant death syndrome and malignant epilepsy. However, detailed electrophysiological and cell biological characterization of HCN4 V759I in Xenopus laevis oocytes and embryonic rat cardiomyocytes, respectively, did not reveal any obvious abnormality. Voltage dependence and kinetics of mutant channel activation, modulation of cAMP-gating by the neuronal HCN channel auxiliary subunit PEX5R, and cell surface expression were indistinguishable from wild-type HCN4. In good agreement, the clinically likewise affected mother of the patient does not exhibit the reported HCN4 variance. HCN4 V759I resembles an innocuous genetic HCN channel variant, which is not sufficient to disturb cardiac pacemaking. Once more, our work emphasizes the importance of careful functional interpretation of genetic findings not only in the context of hereditary cardiac arrhythmias.
Collapse
|
9
|
Semmler J, Kormann J, Srinivasan SP, Köster A, Sälzer D, Reppel M, Hescheler J, Plomann M, Nguemo F. Pacsin 2 is required for the maintenance of a normal cardiac function in the developing mouse heart. Pharmacol Res 2017; 128:200-210. [PMID: 29107716 DOI: 10.1016/j.phrs.2017.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 06/26/2017] [Accepted: 10/15/2017] [Indexed: 11/27/2022]
Abstract
The Pacsin proteins (Pacsin 1, 2 and 3) play an important role in intracellular trafficking and thereby signal transduction in many cells types. This study was designed to examine the role of Pacsin 2 in cardiac development and function. We investigated the development and electrophysiological properties of Pacsin 2 knockout (P2KO) hearts and single cardiomyocytes isolated from 11.5 and 15.5days old fetal mice. Immunofluorescence experiments confirmed the lack of Pacsin 2 protein expression in P2KO cardiac myocytes in comparison to wildtype (WT). Western blotting demonstrates low expression levels of connexin 43 and T-box 3 proteins in P2KO compared to wildtype (WT). Electrophysiology measurements including online Multi-Electrode Array (MEA) based field potential (FP) recordings on isolated whole heart of P2KO mice showed a prolonged AV-conduction time. Patch clamp measurements of P2KO cardiomyocytes revealed differences in action potential (AP) parameters and decreased pacemaker funny channel (If), as well as L-type Ca2+ channel (ICaL), and sodium channel (INa). These findings demonstrate that Pacsin 2 is necessary for cardiac development and function in mouse embryos, which will enhance our knowledge to better understand the genesis of cardiovascular diseases.
Collapse
Affiliation(s)
- Judith Semmler
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany
| | - Jan Kormann
- Institute of Biochemistry, University of Cologne, 50931 Cologne, Germany
| | | | - Annette Köster
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany
| | - Daniel Sälzer
- Institute of Biochemistry, University of Cologne, 50931 Cologne, Germany
| | - Michael Reppel
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany; Department of Cardiology, University of Lübeck, Lübeck, Germany
| | - Jürgen Hescheler
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany
| | - Markus Plomann
- Institute of Biochemistry, University of Cologne, 50931 Cologne, Germany
| | - Filomain Nguemo
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
10
|
Hassinen M, Haverinen J, Vornanen M. Small functional If current in sinoatrial pacemaker cells of the brown trout ( Salmo trutta fario) heart despite strong expression of HCN channel transcripts. Am J Physiol Regul Integr Comp Physiol 2017; 313:R711-R722. [PMID: 28855177 DOI: 10.1152/ajpregu.00227.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 08/21/2017] [Accepted: 08/24/2017] [Indexed: 01/26/2023]
Abstract
Funny current (If), formed by hyperpolarization-activated cyclic nucleotide-gated channels (HCN channels), is supposed to be crucial for the membrane clock regulating the cardiac pacemaker mechanism. We examined the presence and activity of HCN channels in the brown trout (Salmo trutta fario) sinoatrial (SA) pacemaker cells and their putative role in heart rate (fH) regulation. Six HCN transcripts (HCN1, HCN2a, HCN2ba, HCN2bb, HCN3, and HCN4) were expressed in the brown trout heart. The total HCN transcript abundance was 4.0 and 4.9 times higher in SA pacemaker tissue than in atrium and ventricle, respectively. In the SA pacemaker, HCN3 and HCN4 were the main isoforms representing 35.8 ± 2.7 and 25.0 ± 1.5%, respectively, of the total HCN transcripts. Only a small If with a mean current density of -1.2 ± 0.37 pA/pF at -140 mV was found in 4 pacemaker cells out of 16 spontaneously beating cells examined, despite the optimization of recording conditions for If activity. If was not found in any of the 24 atrial myocytes and 21 ventricular myocytes examined. HCN4 coexpressed with the MinK-related peptide 1 (MiRP1) β-subunit in CHO cells generated large If currents. In contrast, HCN3 (+MiRP1) failed to produce If in the same expression system. Cs+ (2 mM), which blocked 84 ± 12% of the native If, reversibly reduced fH 19.2 ± 3.6% of the excised multicellular pacemaker tissue from 53 ± 5 to 44 ± 5 beats/min (P < 0.05). However, this effect was probably due to the reduction of IKr, which was also inhibited (63.5 ± 4.6%) by Cs+ These results strongly suggest that fH regulation in the brown trout heart is largely independent on If.
Collapse
Affiliation(s)
- Minna Hassinen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| | - Jaakko Haverinen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| | - Matti Vornanen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| |
Collapse
|
11
|
Parker AR, Welch MA, Forster LA, Tasneem SM, Dubhashi JA, Baro DJ. SUMOylation of the Hyperpolarization-Activated Cyclic Nucleotide-Gated Channel 2 Increases Surface Expression and the Maximal Conductance of the Hyperpolarization-Activated Current. Front Mol Neurosci 2017; 9:168. [PMID: 28127275 PMCID: PMC5226956 DOI: 10.3389/fnmol.2016.00168] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 12/26/2016] [Indexed: 11/13/2022] Open
Abstract
Small Ubiquitin-like Modifier (SUMO) is a ∼10 kDa peptide that can be post-translationally added to a lysine (K) on a target protein to facilitate protein–protein interactions. Recent studies have found that SUMOylation can be regulated in an activity-dependent manner and that ion channel SUMOylation can alter the biophysical properties and surface expression of the channel. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channel surface expression can be regulated in an activity-dependent manner through unknown processes. We hypothesized that SUMOylation might influence the surface expression of HCN2 channels. In this manuscript, we show that HCN2 channels are SUMOylated in the mouse brain. Baseline levels of SUMOylation were also observed for a GFP-tagged HCN2 channel stably expressed in Human embryonic kidney (Hek) cells. Elevating GFP-HCN2 channel SUMOylation above baseline in Hek cells led to an increase in surface expression that augmented the hyperpolarization-activated current (Ih) mediated by these channels. Increased SUMOylation did not alter Ih voltage-dependence or kinetics of activation. There are five predicted intracellular SUMOylation sites on HCN2. Site-directed mutagenesis indicated that more than one K on the GFP-HCN2 channel was SUMOylated. Enhancing SUMOylation at one of the five predicted sites, K669, led to the increase in surface expression and IhGmax. The role of SUMOylation at additional sites is currently unknown. The SUMOylation site at K669 is also conserved in HCN1 channels. Aberrant SUMOylation has been linked to neurological diseases that also display alterations in HCN1 and HCN2 channel expression, such as seizures and Parkinson’s disease. This work is the first report that HCN channels can be SUMOylated and that this can regulate surface expression and Ih.
Collapse
Affiliation(s)
- Anna R Parker
- Department of Biology, Georgia State University Atlanta, GA, USA
| | - Meghyn A Welch
- Department of Biology, Georgia State University Atlanta, GA, USA
| | - Lori A Forster
- Neuroscience Institute, Georgia State University Atlanta, GA, USA
| | - Sarah M Tasneem
- Department of Biology, Georgia State University Atlanta, GA, USA
| | | | - Deborah J Baro
- Department of Biology, Georgia State UniversityAtlanta, GA, USA; Neuroscience Institute, Georgia State UniversityAtlanta, GA, USA
| |
Collapse
|
12
|
Christensen AH, Chatelain FC, Huttner IG, Olesen MS, Soka M, Feliciangeli S, Horvat C, Santiago CF, Vandenberg JI, Schmitt N, Olesen SP, Lesage F, Fatkin D. The two-pore domain potassium channel, TWIK-1, has a role in the regulation of heart rate and atrial size. J Mol Cell Cardiol 2016; 97:24-35. [PMID: 27103460 DOI: 10.1016/j.yjmcc.2016.04.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 04/11/2016] [Accepted: 04/14/2016] [Indexed: 12/24/2022]
Abstract
The two-pore domain potassium (K(+)) channel TWIK-1 (or K2P1.1) contributes to background K(+) conductance in diverse cell types. TWIK-1, encoded by the KCNK1 gene, is present in the human heart with robust expression in the atria, however its physiological significance is unknown. To evaluate the cardiac effects of TWIK-1 deficiency, we studied zebrafish embryos after knockdown of the two KCNK1 orthologues, kcnk1a and kcnk1b. Knockdown of kcnk1a or kcnk1b individually caused bradycardia and atrial dilation (p<0.001 vs. controls), while ventricular stroke volume was preserved. Combined knockdown of both kcnk1a and kcnk1b resulted in a more severe phenotype, which was partially reversed by co-injection of wild-type human KCNK1 mRNA, but not by a dominant negative variant of human KCNK1 mRNA. To determine whether genetic variants in KCNK1 might cause atrial fibrillation (AF), we sequenced protein-coding regions in two independent cohorts of patients (373 subjects) and identified three non-synonymous variants, p.R171H, p.I198M and p.G236S, that were all located in highly conserved amino acid residues. In transfected mammalian cells, zebrafish and wild-type human TWIK-1 channels had a similar cellular distribution with predominant localization in the endosomal compartment. Two-electrode voltage-clamp experiments using Xenopus oocytes showed that both zebrafish and wild-type human TWIK-1 channels produced K(+) currents that are sensitive to external K(+) concentration as well as acidic pH. There were no effects of the three KCNK1 variants on cellular localization, current amplitude or reversal potential at pH7.4 or pH6. Our data indicate that TWIK-1 has a highly conserved role in cardiac function and is required for normal heart rate and atrial morphology. Despite the functional importance of TWIK-1 in the atrium, genetic variation in KCNK1 is not a common primary cause of human AF.
Collapse
Affiliation(s)
- Alex Hørby Christensen
- Molecular Cardiology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Franck C Chatelain
- LabEx ICST, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, and Université de Nice Sophia-Antipolis, Sophia-Antipolis, Valbonne, France
| | - Inken G Huttner
- Molecular Cardiology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia
| | - Morten Salling Olesen
- Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark
| | - Magdalena Soka
- Molecular Cardiology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Sylvain Feliciangeli
- LabEx ICST, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, and Université de Nice Sophia-Antipolis, Sophia-Antipolis, Valbonne, France
| | - Claire Horvat
- Molecular Cardiology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Celine F Santiago
- Molecular Cardiology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Jamie I Vandenberg
- Molecular Cardiology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia
| | - Nicole Schmitt
- Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark
| | - Søren-Peter Olesen
- Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark
| | - Florian Lesage
- LabEx ICST, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, and Université de Nice Sophia-Antipolis, Sophia-Antipolis, Valbonne, France
| | - Diane Fatkin
- Molecular Cardiology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia; Cardiology Department, St Vincent's Hospital, Darlinghurst, New South Wales, Australia.
| |
Collapse
|
13
|
Smith T, Al Otaibi M, Sathish J, Djouhri L. Increased expression of HCN2 channel protein in L4 dorsal root ganglion neurons following axotomy of L5- and inflammation of L4-spinal nerves in rats. Neuroscience 2015; 295:90-102. [DOI: 10.1016/j.neuroscience.2015.03.041] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 02/28/2015] [Accepted: 03/19/2015] [Indexed: 12/31/2022]
|
14
|
Abstract
One of the main strategies for cancer therapy is to use tyrosine kinase inhibitors for inhibiting tumor proliferation. Increasing evidence has demonstrated the potential risks of cardiac arrhythmias (such as prolonged QT interval) of these drugs. We report here that a widely used selective inhibitor of Src tyrosine kinases, 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP2), can inhibit and prevent β-adrenergic stimulation of cardiac pacemaker activity. First, in dissected rat sinus node, PP2 inhibited and prevented the isoproterenol-induced increase of spontaneous beating rate. Second, in isolated rat sinus node myocytes, PP2 suppressed the hyperpolarization-activated "funny" current (traditionally called cardiac pacemaker current, I(f)) by negatively shifting the activation curve and decelerating activation kinetics. Third, in isolated rat sinus node myocytes, PP2 decreased the Src kinase activity, the cell surface expression, and tyrosine phosphorylation of hyperpolarization-activated, cyclic nucleotide-modulated channel 4 (HCN4) channel proteins. Finally, in human embryonic kidney 293 cells overexpressing recombinant human HCN4 channels, PP2 reversed the enhancement of HCN4 channels by isoproterenol and inhibited 573x, a cyclic adenosine momophosphate-insensitive human HCN4 mutant. These results demonstrated that inhibition of Src kinase activity in heart by PP2 decreased and prevented β-adrenergic stimulation of cardiac pacemaker activity. These effects are mediated, at least partially, by a cAMP-independent attenuation of channel activity and cell surface expression of HCN4, the main channel protein that controls the heart rate.
Collapse
|
15
|
Hyperpolarization-activated cyclic nucleotide-gated channels and cAMP-dependent modulation of exocytosis in cultured rat lactotrophs. J Neurosci 2015; 34:15638-47. [PMID: 25411492 DOI: 10.1523/jneurosci.5290-13.2014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hormone and neurotransmitter release from vesicles is mediated by regulated exocytosis, where an aqueous channel-like structure, termed a fusion pore, is formed. It was recently shown that second messenger cAMP modulates the fusion pore, but the detailed mechanisms remain elusive. In this study, we asked whether the hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, which are activated by cAMP, are involved in the regulation of unitary exocytic events. By using the Western blot technique, a real-time PCR, immunocytochemistry in combination with confocal microscopy, and voltage-clamp measurements of hyperpolarizing currents, we show that HCN channels are present in the plasma membrane and in the membrane of secretory vesicles of isolated rat lactotrophs. Single vesicle membrane capacitance measurements of lactotrophs, where HCN channels were either augmented by transfection or blocked with an HCN channel blocker (ZD7288), show modulated fusion pore properties. We suggest that the changes in local cation concentration, mediated through HCN channels, which are located on or near secretory vesicles, have an important role in modulating exocytosis.
Collapse
|
16
|
Pan Y, Laird JG, Yamaguchi DM, Baker SA. A di-arginine ER retention signal regulates trafficking of HCN1 channels from the early secretory pathway to the plasma membrane. Cell Mol Life Sci 2014; 72:833-43. [PMID: 25142030 PMCID: PMC4309907 DOI: 10.1007/s00018-014-1705-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 07/14/2014] [Accepted: 08/12/2014] [Indexed: 12/25/2022]
Abstract
Hyperpolarization-activated cyclic nucleotide-gated 1 (HCN1) channels carry Ih, which contributes to neuronal excitability and signal transmission in the nervous system. Controlling the trafficking of HCN1 is an important aspect of its regulation, yet the details of this process are poorly understood. Here, we investigated how the C-terminus of HCN1 regulates trafficking by testing for its ability to redirect the localization of a non-targeted reporter in transgenic Xenopus laevis photoreceptors. We found that HCN1 contains an ER localization signal and through a series of deletion constructs, identified the responsible di-arginine ER retention signal. This signal is located in the intrinsically disordered region of the C-terminus of HCN1. To test the function of the ER retention signal in intact channels, we expressed wild type and mutant HCN1 in HEK293 cells and found this signal negatively regulates surface expression of HCN1. In summary, we report a new mode of regulating HCN1 trafficking: through the use of a di-arginine ER retention signal that monitors processing of the channel in the early secretory pathway.
Collapse
Affiliation(s)
- Yuan Pan
- Department of Biochemistry, Carver College of Medicine, University of Iowa, 51 Newton Road, Biochemistry, 4-712 BSB, Iowa City, IA, 52242, USA
| | | | | | | |
Collapse
|
17
|
Dufour MA, Woodhouse A, Goaillard JM. Somatodendritic ion channel expression in substantia nigra pars compacta dopaminergic neurons across postnatal development. J Neurosci Res 2014; 92:981-99. [PMID: 24723263 DOI: 10.1002/jnr.23382] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 02/14/2014] [Accepted: 02/14/2014] [Indexed: 01/12/2023]
Abstract
Dopaminergic neurons of the substantia nigra pars compacta (SNc) are involved in the control of movement, sleep, reward, learning, and nervous system disorders and disease. To date, a thorough characterization of the ion channel phenotype of this important neuronal population is lacking. Using immunohistochemistry, we analyzed the somatodendritic expression of voltage-gated ion channel subunits that are involved in pacemaking activity in SNc dopaminergic neurons in 6-, 21-, and 40-day-old rats. Our results demonstrate that the same complement of somatodendritic ion channels is present in SNc dopaminergic neurons from P6 to P40. The major developmental changes were an increase in the dendritic range of the immunolabeling for the HCN, T-type calcium, Kv4.3, delayed rectifier, and SK channels. Our study sheds light on the ion channel subunits that contribute to the somatodendritic delayed rectifier (Kv1.3, Kv2.1, Kv3.2, Kv3.3), A-type (Kv4.3) and calcium-activated SK (SK1, SK2, SK3) potassium currents, IH (mainly HCN2, HCN4), and the L- (Cav1.2, Cav1.3) and T-type (mainly Cav3.1, Cav3.3) calcium currents in SNc dopaminergic neurons. Finally, no robust differences in voltage-gated ion channel immunolabeling were observed across the population of SNc dopaminergic neurons for each age examined, suggesting that differing levels of individual ion channels are unlikely to distinguish between specific subpopulations of SNc dopaminergic neurons. This is significant in light of previous studies suggesting that age- or region-associated variations in the expression profile of voltage-gated ion channels in SNc dopaminergic neurons may underlie their vulnerability to dysfunction and disease.
Collapse
Affiliation(s)
- Martial A Dufour
- INSERM, UMR_S 1072, 13015, Marseille, France; Aix-Marseille Université, UNIS, 13015, Marseille, France
| | | | | |
Collapse
|
18
|
Wilkars W, Wollberg J, Mohr E, Han M, Chetkovich DM, Bähring R, Bender RA. Nedd4‐2 regulates surface expression and may affect
N
‐glycosylation of hyperpolarization‐activated cyclic nucleotide‐gated (HCN)‐1 channels. FASEB J 2014; 28:2177-90. [DOI: 10.1096/fj.13-242032] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Wiebke Wilkars
- Institute of NeuroanatomyUniversity of Hamburg Medical CenterHamburgGermany
| | - Jessica Wollberg
- Institute of Cellular and Integrative PhysiologyUniversity of Hamburg Medical CenterHamburgGermany
| | - Evita Mohr
- Institute of NeuroanatomyUniversity of Hamburg Medical CenterHamburgGermany
| | - Mieri Han
- Institute of NeuroanatomyUniversity of Hamburg Medical CenterHamburgGermany
| | - Dane M. Chetkovich
- Davee Department of Neurology and Clinical NeurosciencesNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Department of PhysiologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Robert Bähring
- Institute of Cellular and Integrative PhysiologyUniversity of Hamburg Medical CenterHamburgGermany
| | - Roland A. Bender
- Institute of NeuroanatomyUniversity of Hamburg Medical CenterHamburgGermany
| |
Collapse
|
19
|
Zhao ZG, Niu CY, Qiu JF, Chen XD, Li JC. Effect of mesenteric lymph duct ligation on gene expression profiles of renal tissue in hemorrhagic shock rats with fluid resuscitation. Ren Fail 2013; 36:271-7. [DOI: 10.3109/0886022x.2013.844623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
20
|
Rapetti-Mauss R, O'Mahony F, Sepulveda FV, Urbach V, Harvey BJ. Oestrogen promotes KCNQ1 potassium channel endocytosis and postendocytic trafficking in colonic epithelium. J Physiol 2013; 591:2813-31. [PMID: 23529131 PMCID: PMC3690688 DOI: 10.1113/jphysiol.2013.251678] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 03/19/2013] [Indexed: 12/23/2022] Open
Abstract
The cAMP-regulated potassium channel KCNQ1:KCNE3 plays an essential role in transepithelial Cl(-) secretion. Recycling of K(+) across the basolateral membrane provides the driving force necessary to maintain apical Cl(-) secretion. The steroid hormone oestrogen (17β-oestradiol; E2), produces a female-specific antisecretory response in rat distal colon through the inhibition of the KCNQ1:KCNE3 channel. It has previously been shown that rapid inhibition of the channel conductance results from E2-induced uncoupling of the KCNE3 regulatory subunit from the KCNQ1 channel pore complex. The purpose of this study was to determine the mechanism required for sustained inhibition of the channel function. We found that E2 plays a role in regulation of KCNQ1 cell membrane abundance by endocytosis. Ussing chamber experiments have shown that E2 inhibits both Cl(-) secretion and KCNQ1 current in a colonic cell line, HT29cl.19A, when cultured as a confluent epithelium. Following E2 treatment, KCNQ1 was retrieved from the plasma membrane by a clathrin-mediated endocytosis, which involved the association between KCNQ1 and the clathrin adaptor, AP-2. Following endocytosis, KCNQ1 was accumulated in early endosomes. Following E2-induced endocytosis, rather than being degraded, KCNQ1 was recycled by a biphasic mechanism involving Rab4 and Rab11. Protein kinase Cδ and AMP-dependent kinase were rapidly phosphorylated in response to E2 on their activating phosphorylation sites, Ser643 and Thr172, respectively (as previously shown). Both kinases are necessary for the E2-induced endocytosis, because E2 failed to induce KCNQ1 internalization following pretreatment with specific inhibitors of both protein kinase Cδ and AMP-dependent kinase. The ubiquitin ligase Nedd4.2 binds KCNQ1 in response to E2 to induce channel internalization. This study has provided the first demonstration of hormonal regulation of KCNQ1 trafficking. In conclusion, we propose that internalization of KCNQ1 is a key event in the sustained antisecretory response to oestrogen.
Collapse
Affiliation(s)
- Raphael Rapetti-Mauss
- Department of Molecular Medicine, RCSI-ERC, Beaumont Hospital, PO Box 9063, Dublin 9, Ireland
| | | | | | | | | |
Collapse
|
21
|
Paspalas CD, Wang M, Arnsten AFT. Constellation of HCN channels and cAMP regulating proteins in dendritic spines of the primate prefrontal cortex: potential substrate for working memory deficits in schizophrenia. ACTA ACUST UNITED AC 2012; 23:1643-54. [PMID: 22693343 DOI: 10.1093/cercor/bhs152] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Schizophrenia associates with impaired prefrontal cortical (PFC) function and alterations in cyclic AMP (cAMP) signaling pathways. These include genetic insults to disrupted-in-schizophrenia (DISC1) and phosphodiesterases (PDE4s) regulating cAMP hydrolysis, and increased dopamine D1 receptor (D1R) expression that elevates cAMP. We used immunoelectron microscopy to localize DISC1, PDE4A, PDE4B, and D1R in monkey PFC and to view spatial interactions with hyperpolarization-activated cyclic nucleotide-gated (HCN) channels that gate network inputs when opened by cAMP. Physiological interactions between PDE4s and HCN channels were tested in recordings of PFC neurons in monkeys performing a spatial working memory task. The study reveals a constellation of cAMP-related proteins (DISC1, PDE4A, and D1R) and HCN channels next to excitatory synapses and the spine neck in thin spines of superficial PFC, where working memory microcircuits interconnect and spine loss is most evident in schizophrenia. In contrast, channels in dendrites were distant from synapses and cAMP-related proteins, and were associated with endosomal trafficking. The data suggest that a cAMP signalplex is selectively positioned in the spines to gate PFC pyramidal cell microcircuits. Single-unit recordings confirmed physiological interactions between cAMP and HCN channels, consistent with gating actions. These data may explain why PFC networks are especially vulnerable to genetic insults that dysregulate cAMP signaling.
Collapse
|
22
|
Regulation of axonal HCN1 trafficking in perforant path involves expression of specific TRIP8b isoforms. PLoS One 2012; 7:e32181. [PMID: 22363812 PMCID: PMC3283722 DOI: 10.1371/journal.pone.0032181] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 01/23/2012] [Indexed: 12/22/2022] Open
Abstract
The functions of HCN channels in neurons depend critically on their subcellular localization, requiring fine-tuned machinery that regulates subcellular channel trafficking. Here we provide evidence that regulatory mechanisms governing axonal HCN channel trafficking involve association of the channels with specific isoforms of the auxiliary subunit TRIP8b. In the medial perforant path, which normally contains HCN1 channels in axon terminals in immature but not in adult rodents, we found axonal HCN1 significantly increased in adult mice lacking TRIP8b (TRIP8b−/−). Interestingly, adult mice harboring a mutation that results in expression of only the two most abundant TRIP8b isoforms (TRIP8b[1b/2]−/−) exhibited an HCN1 expression pattern similar to wildtype mice, suggesting that presence of one or both of these isoforms (TRIP8b(1a), TRIP8b(1a-4)) prevents HCN1 from being transported to medial perforant path axons in adult mice. Concordantly, expression analyses demonstrated a strong increase of expression of both TRIP8b isoforms in rat entorhinal cortex with age. However, when overexpressed in cultured entorhinal neurons of rats, TRIP8b(1a), but not TRIP8b(1a-4), altered substantially the subcellular distribution of HCN1 by promoting somatodendritic and reducing axonal expression of the channels. Taken together, we conclude that TRIP8b isoforms are important regulators of HCN1 trafficking in entorhinal neurons and that the alternatively-spliced isoform TRIP8b(1a) could be responsible for the age-dependent redistribution of HCN channels out of perforant path axon terminals.
Collapse
|
23
|
Tonic nanomolar dopamine enables an activity-dependent phase recovery mechanism that persistently alters the maximal conductance of the hyperpolarization-activated current in a rhythmically active neuron. J Neurosci 2012; 31:16387-97. [PMID: 22072689 DOI: 10.1523/jneurosci.3770-11.2011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The phases at which network neurons fire in rhythmic motor outputs are critically important for the proper generation of motor behaviors. The pyloric network in the crustacean stomatogastric ganglion generates a rhythmic motor output wherein neuronal phase relationships are remarkably invariant across individuals and throughout lifetimes. The mechanisms for maintaining these robust phase relationships over the long-term are not well described. Here we show that tonic nanomolar dopamine (DA) acts at type 1 DA receptors (D1Rs) to enable an activity-dependent mechanism that can contribute to phase maintenance in the lateral pyloric (LP) neuron. The LP displays continuous rhythmic bursting. The activity-dependent mechanism was triggered by a prolonged decrease in LP burst duration, and it generated a persistent increase in the maximal conductance (G(max)) of the LP hyperpolarization-activated current (I(h)), but only in the presence of steady-state DA. Interestingly, micromolar DA produces an LP phase advance accompanied by a decrease in LP burst duration that abolishes normal LP network function. During a 1 h application of micromolar DA, LP phase recovered over tens of minutes because, the activity-dependent mechanism enabled by steady-state DA was triggered by the micromolar DA-induced decrease in LP burst duration. Presumably, this mechanism restored normal LP network function. These data suggest steady-state DA may enable homeostatic mechanisms that maintain motor network output during protracted neuromodulation. This DA-enabled, activity-dependent mechanism to preserve phase may be broadly relevant, as diminished dopaminergic tone has recently been shown to reduce I(h) in rhythmically active neurons in the mammalian brain.
Collapse
|
24
|
Netter MF, Zuzarte M, Schlichthörl G, Klöcker N, Decher N. The HCN4 Channel Mutation D553N Associated With Bradycardia Has a C-linker Mediated Gating Defect. Cell Physiol Biochem 2012; 30:1227-40. [DOI: 10.1159/000343314] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2012] [Indexed: 11/19/2022] Open
|
25
|
Schmidt N, Akaaboune M, Gajendran N, Martinez-Pena y Valenzuela I, Wakefield S, Thurnheer R, Brenner HR. Neuregulin/ErbB regulate neuromuscular junction development by phosphorylation of α-dystrobrevin. ACTA ACUST UNITED AC 2011; 195:1171-84. [PMID: 22184199 PMCID: PMC3246897 DOI: 10.1083/jcb.201107083] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neuregulin/ErbB signaling maintains high efficacy of synaptic transmission by stabilizing the postsynaptic apparatus via phosphorylation of α-dystrobrevin1. Neuregulin (NRG)/ErbB signaling is involved in numerous developmental processes in the nervous system, including synapse formation and function in the central nervous system. Although intensively investigated, its role at the neuromuscular synapse has remained elusive. Here, we demonstrate that loss of neuromuscular NRG/ErbB signaling destabilized anchoring of acetylcholine receptors (AChRs) in the postsynaptic muscle membrane and that this effect was caused by dephosphorylation of α-dystrobrevin1, a component of the postsynaptic scaffold. Specifically, in mice in which NRG signaling to muscle was genetically or pharmacologically abolished, postsynaptic AChRs moved rapidly from the synaptic to the perisynaptic membrane, and the subsynaptic scaffold that anchors the AChRs was impaired. These defects combined compromised synaptic transmission. We further show that blockade of NRG/ErbB signaling abolished tyrosine phosphorylation of α-dystrobrevin1, which reduced the stability of receptors in agrin-induced AChR clusters in cultured myotubes. Our data indicate that NRG/ErbB signaling maintains high efficacy of synaptic transmission by stabilizing the postsynaptic apparatus via phosphorylation of α-dystrobrevin1.
Collapse
Affiliation(s)
- Nadine Schmidt
- Institute of Physiology, Department of Biomedicine, University of Basel, CH-4056, Basel, Switzerland
| | | | | | | | | | | | | |
Collapse
|
26
|
Li X, DiFiglia M. The recycling endosome and its role in neurological disorders. Prog Neurobiol 2011; 97:127-41. [PMID: 22037413 DOI: 10.1016/j.pneurobio.2011.10.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 10/14/2011] [Accepted: 10/17/2011] [Indexed: 02/08/2023]
Abstract
The recycling endosome (RE) is an organelle in the endocytic pathway where plasma membranes (proteins and lipids) internalized by endocytosis are processed back to the cell surface for reuse. Endocytic recycling is the primary way for the cell to maintain constituents of the plasma membrane (Griffiths et al., 1989), i.e., to maintain the abundance of receptors and transporters on cell surfaces. Membrane traffic through the RE is crucial for several key cellular processes including cytokinesis and cell migration. In polarized cells, including neurons, the RE is vital for the generation and maintenance of the polarity of the plasma membrane. Many RE dependent cargo molecules are known to be important for neuronal function and there is evidence that improper function of key proteins in RE-associated pathways may contribute to the pathogenesis of neurological disorders, including Huntington's disease. The function of the RE in neurons is poorly understood. Therefore, there is need to understand how membrane dynamics in RE-associated pathways are affected or participate in the development or progression of neurological diseases. This review summarizes advances in understanding endocytic recycling associated with the RE, challenges in elucidating molecular mechanisms underlying RE function, and evidence for RE dysfunction in neurological disorders.
Collapse
Affiliation(s)
- Xueyi Li
- Laboratory of Cellular Neurobiology and Department of Neurology, Massachusetts General Hospital, 114 16th Street, Charlestown, MA 02129, USA
| | | |
Collapse
|
27
|
Different subcellular populations of L-type Ca2+ channels exhibit unique regulation and functional roles in cardiomyocytes. J Mol Cell Cardiol 2011; 52:376-87. [PMID: 21888911 DOI: 10.1016/j.yjmcc.2011.08.014] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 07/11/2011] [Accepted: 08/17/2011] [Indexed: 11/23/2022]
Abstract
Influx of Ca(2+) through L-type Ca(2+) channels (LTCCs) contributes to numerous cellular processes in cardiomyocytes including excitation-contraction (EC) coupling, membrane excitability, and transcriptional regulation. Distinct subpopulations of LTCCs have been identified in cardiac myocytes, including those at dyadic junctions and within different plasma membrane microdomains such as lipid rafts and caveolae. These subpopulations of LTCCs exhibit regionally distinct functional properties and regulation, affording precise spatiotemporal modulation of L-type Ca(2+) current (I(Ca,L)). Different subcellular LTCC populations demonstrate variable rates of Ca(2+)-dependent inactivation and sometimes coupled gating of neighboring channels, which can lead to focal, persistent I(Ca,L). In addition, the assembly of spatially defined macromolecular signaling complexes permits compartmentalized regulation of I(Ca,L) by a variety of neurohormonal pathways. For example, β-adrenergic receptor subtypes signal to different LTCC subpopulations, with β(2)-adrenergic activation leading to enhanced I(Ca,L) through caveolar LTCCs and β(1)-adrenergic stimulation modulating LTCCs outside of caveolae. Disruptions in the normal subcellular targeting of LTCCs and associated signaling proteins may contribute to the pathophysiology of a variety of cardiac diseases including heart failure and certain arrhythmias. Further identifying the characteristic functional properties and array of regulatory molecules associated with specific LTCC subpopulations will provide a mechanistic framework to understand how LTCCs contribute to diverse cellular processes in normal and diseased myocardium. This article is part of a Special Issue entitled "Local Signaling in Myocytes".
Collapse
|
28
|
Towards an integrated view of HCN channel role in epilepsy. Curr Opin Neurobiol 2011; 21:873-9. [PMID: 21782415 DOI: 10.1016/j.conb.2011.06.013] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2011] [Revised: 06/11/2011] [Accepted: 06/30/2011] [Indexed: 12/17/2022]
Abstract
Epilepsy is the third most common brain disorder and affects millions of people. Epilepsy is characterized by the occurrence of spontaneous seizures, that is, bursts of synchronous firing of large populations of neurons. These are believed to result from abnormal regulation of neuronal excitability that favors hypersynchrony. Among the intrinsic conductances that govern neuronal excitability, the hyperpolarization-activated current (I(h)) plays complex and important roles in the fine-tuning of both cellular and network activity. Not surprisingly, dysregulation of I(h) and/or of its conducting ion-channels (HCN) has been strongly implicated in various experimental models of epilepsy, as well as in human epilepsy. Here we provide an overview of recent findings on the distinct physiological roles played by I(h) in specific contexts, and the cellular mechanisms that underlie these functions, including the subunit make-up of the channels. We further discuss current knowledge of dysregulation of I(h) and HCN channels in epilepsy in light of the multifaceted functions of I(h) in the brain.
Collapse
|
29
|
Lewis AS, Vaidya SP, Blaiss CA, Liu Z, Stoub TR, Brager DH, Chen X, Bender RA, Estep CM, Popov AB, Kang CE, Van Veldhoven PP, Bayliss DA, Nicholson DA, Powell CM, Johnston D, Chetkovich DM. Deletion of the hyperpolarization-activated cyclic nucleotide-gated channel auxiliary subunit TRIP8b impairs hippocampal Ih localization and function and promotes antidepressant behavior in mice. J Neurosci 2011; 31:7424-40. [PMID: 21593326 PMCID: PMC3169171 DOI: 10.1523/jneurosci.0936-11.2011] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Revised: 03/29/2011] [Accepted: 03/31/2011] [Indexed: 12/15/2022] Open
Abstract
Output properties of neurons are greatly shaped by voltage-gated ion channels, whose biophysical properties and localization within axodendritic compartments serve to significantly transform the original input. The hyperpolarization-activated current, I(h), is mediated by hyperpolarization-activated cyclic nucleotide-gated (HCN) channels and plays a fundamental role in influencing neuronal excitability by regulating both membrane potential and input resistance. In neurons such as cortical and hippocampal pyramidal neurons, the subcellular localization of HCN channels plays a critical functional role, yet mechanisms controlling HCN channel trafficking are not fully understood. Because ion channel function and localization are often influenced by interacting proteins, we generated a knock-out mouse lacking the HCN channel auxiliary subunit, tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b). Eliminating expression of TRIP8b dramatically reduced I(h) expression in hippocampal pyramidal neurons. Loss of I(h)-dependent membrane voltage properties was attributable to reduction of HCN channels on the neuronal surface, and there was a striking disruption of the normal expression pattern of HCN channels in pyramidal neuron dendrites. In heterologous cells and neurons, absence of TRIP8b increased HCN subunit targeting to and degradation by lysosomes. Mice lacking TRIP8b demonstrated motor learning deficits and enhanced resistance to multiple tasks of behavioral despair with high predictive validity for antidepressant efficacy. We observed similar resistance to behavioral despair in distinct mutant mice lacking HCN1 or HCN2. These data demonstrate that interaction with the auxiliary subunit TRIP8b is a major mechanism underlying proper expression of HCN channels and I(h) in vivo, and suggest that targeting I(h) may provide a novel approach to treatment of depression.
Collapse
Affiliation(s)
- Alan S. Lewis
- Davee Department of Neurology and Clinical Neurosciences
| | - Sachin P. Vaidya
- Center for Learning and Memory, University of Texas at Austin, Austin, Texas 78712
| | | | - Zhiqiang Liu
- Davee Department of Neurology and Clinical Neurosciences
| | - Travis R. Stoub
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois 60612
| | - Darrin H. Brager
- Center for Learning and Memory, University of Texas at Austin, Austin, Texas 78712
| | | | - Roland A. Bender
- Institute of Anatomy I, University of Hamburg Medical Center, D-20246 Hamburg, Germany, and
| | - Chad M. Estep
- Davee Department of Neurology and Clinical Neurosciences
| | | | | | - Paul P. Van Veldhoven
- Laboratorium voor Lipidenbiochemie en Proteïnen-Interactie, Departement Moleculaire Celbiologie, Katholieke Universiteit Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium
| | - Douglas A. Bayliss
- Departments of Pharmacology and
- Anesthesiology, University of Virginia, Charlottesville, Virginia 22908
| | - Daniel A. Nicholson
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois 60612
| | - Craig M. Powell
- Departments of Neurology and
- Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8813
| | - Daniel Johnston
- Center for Learning and Memory, University of Texas at Austin, Austin, Texas 78712
| | - Dane M. Chetkovich
- Davee Department of Neurology and Clinical Neurosciences
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| |
Collapse
|
30
|
Gopalakrishnan K, Morgan EE, Yerga-Woolwine S, Farms P, Kumarasamy S, Kalinoski A, Liu X, Wu J, Liu L, Joe B. Augmented rififylin is a risk factor linked to aberrant cardiomyocyte function, short-QT interval and hypertension. Hypertension 2011; 57:764-71. [PMID: 21357277 PMCID: PMC3060303 DOI: 10.1161/hypertensionaha.110.165803] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Accepted: 01/27/2011] [Indexed: 01/14/2023]
Abstract
Using congenic strains of the Dahl salt-sensitive (S) rat introgressed with genomic segments from the normotensive Lewis rat, a blood pressure quantitative trait locus was previously mapped within 104 kb on chromosome 10. The goal of the current study was to conduct extensive phenotypic studies and to further fine-map this locus. At 14 weeks of age, the blood pressure of the congenic rats fed a low-salt diet was significantly higher by 47 mm Hg (P<0.001) compared with that of the S rat. A time-course study showed that the blood pressure effect was significant from very young ages of 50 to 52 days (13 mm Hg; P<0.01). The congenic strain implanted with electrocardiography transmitters demonstrated shorter-QT intervals and increased heart rate compared with S rats (P<0.01). The average survival of the congenic strain was shorter (134 days) compared with the S rat (175 days; P<0.0007). The critical region was narrowed to <42.5 kb containing 171 variants and a single gene, rififylin. Both the mRNA and protein levels of rififylin were significantly higher in the hearts of the congenic strain. Overexpression of rififylin is known to delay endocytic recycling. Endocytic recycling of fluorescently labeled holotransferrin from cardiomyocytes of the congenic strain was slower than that of S rats (P<0.01). Frequency of cardiomyocyte beats in the congenic strain (62±9 bpm) was significantly higher than that of the S rat (24±6 bpm; P<0.001). Taken together, our study provides evidence to suggest that early perturbations in endocytic recycling caused by the overexpression of Rffl is a novel physiological mechanism potentially underlying the development of hypertension.
Collapse
Affiliation(s)
- Kathirvel Gopalakrishnan
- Physiological Genomics Laboratory, University of Toledo College of Medicine, Toledo, OH, 43614, USA
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, 43614, USA
| | - Eric E. Morgan
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, 43614, USA
| | - Shane Yerga-Woolwine
- Physiological Genomics Laboratory, University of Toledo College of Medicine, Toledo, OH, 43614, USA
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, 43614, USA
| | - Phyllis Farms
- Physiological Genomics Laboratory, University of Toledo College of Medicine, Toledo, OH, 43614, USA
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, 43614, USA
| | - Sivarajan Kumarasamy
- Physiological Genomics Laboratory, University of Toledo College of Medicine, Toledo, OH, 43614, USA
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, 43614, USA
| | - Andrea Kalinoski
- Department of Surgery, University of Toledo College of Medicine, Toledo, OH, 43614, USA
- University of Toledo Advanced Microscopy Imaging Center, University of Toledo College of Medicine, Toledo, OH, 43614, USA
| | - Xiaochen Liu
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, 43614, USA
| | - Jian Wu
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, 43614, USA
| | - Lijun Liu
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, 43614, USA
| | - Bina Joe
- Physiological Genomics Laboratory, University of Toledo College of Medicine, Toledo, OH, 43614, USA
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, 43614, USA
| |
Collapse
|
31
|
Zaarour N, Defontaine N, Demaretz S, Azroyan A, Cheval L, Laghmani K. Secretory carrier membrane protein 2 regulates exocytic insertion of NKCC2 into the cell membrane. J Biol Chem 2011; 286:9489-9502. [PMID: 21205824 PMCID: PMC3059028 DOI: 10.1074/jbc.m110.166546] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 12/23/2010] [Indexed: 12/20/2022] Open
Abstract
The renal-specific Na-K-2Cl co-transporter, NKCC2, plays a pivotal role in regulating body salt levels and blood pressure. NKCC2 mutations lead to type I Bartter syndrome, a life-threatening kidney disease. Regulation of NKCC2 trafficking behavior serves as a major mechanism in controlling NKCC2 activity across the plasma membrane. However, the identities of the protein partners involved in cell surface targeting of NKCC2 are largely unknown. To gain insight into these processes, we used a yeast two-hybrid system to screen a kidney cDNA library for proteins that interact with the NKCC2 C terminus. One binding partner we identified was SCAMP2 (secretory carrier membrane protein 2). Microscopic confocal imaging and co-immunoprecipitation assays confirmed NKCC2-SCAMP2 interaction in renal cells. SCAMP2 associated also with the structurally related co-transporter NCC, suggesting that the interaction with SCAMP2 is a common feature of sodium-dependent chloride co-transporters. Heterologous expression of SCAMP2 specifically decreased cell surface abundance as well as transport activity of NKCC2 across the plasma membrane. Co-immunolocalization experiments revealed that intracellularly retained NKCC2 co-localizes with SCAMP2 in recycling endosomes. The rate of NKCC2 endocytic retrieval, assessed by the sodium 2-mercaptoethane sulfonate cleavage assay, was not affected by SCAMP2. The surface-biotinylatable fraction of newly inserted NKCC2 in the plasma membrane was reduced by SCAMP2, demonstrating that SCAMP2-induced decrease in surface NKCC2 is due to decreased exocytotic trafficking. Finally, a single amino acid mutation, cysteine 201 to alanine, within the conserved cytoplasmic E peptide of SCAMP2, which is believed to regulate exocytosis, abolished SCAMP2-mediated down-regulation of the co-transporter. Taken together, these data are consistent with a model whereby SCAMP2 regulates NKCC2 transit through recycling endosomes and limits the cell surface targeting of the co-transporter by interfering with its exocytotic trafficking.
Collapse
Affiliation(s)
- Nancy Zaarour
- From INSERM, Centre de Recherche des Cordeliers, UMRS 872, CNRS, ERL7226, 75006 Paris, France
- Université Pierre et Marie Curie, 75006 Paris, France, and
- the Faculté de Médecine, Université Paris-Descartes, 75005 Paris, France
| | - Nadia Defontaine
- From INSERM, Centre de Recherche des Cordeliers, UMRS 872, CNRS, ERL7226, 75006 Paris, France
- Université Pierre et Marie Curie, 75006 Paris, France, and
- the Faculté de Médecine, Université Paris-Descartes, 75005 Paris, France
| | - Sylvie Demaretz
- From INSERM, Centre de Recherche des Cordeliers, UMRS 872, CNRS, ERL7226, 75006 Paris, France
- Université Pierre et Marie Curie, 75006 Paris, France, and
- the Faculté de Médecine, Université Paris-Descartes, 75005 Paris, France
| | - Anie Azroyan
- From INSERM, Centre de Recherche des Cordeliers, UMRS 872, CNRS, ERL7226, 75006 Paris, France
- Université Pierre et Marie Curie, 75006 Paris, France, and
- the Faculté de Médecine, Université Paris-Descartes, 75005 Paris, France
| | - Lydie Cheval
- From INSERM, Centre de Recherche des Cordeliers, UMRS 872, CNRS, ERL7226, 75006 Paris, France
- Université Pierre et Marie Curie, 75006 Paris, France, and
- the Faculté de Médecine, Université Paris-Descartes, 75005 Paris, France
| | - Kamel Laghmani
- From INSERM, Centre de Recherche des Cordeliers, UMRS 872, CNRS, ERL7226, 75006 Paris, France
- Université Pierre et Marie Curie, 75006 Paris, France, and
- the Faculté de Médecine, Université Paris-Descartes, 75005 Paris, France
| |
Collapse
|
32
|
Naslavsky N, Caplan S. EHD proteins: key conductors of endocytic transport. Trends Cell Biol 2011; 21:122-31. [PMID: 21067929 PMCID: PMC3052690 DOI: 10.1016/j.tcb.2010.10.003] [Citation(s) in RCA: 196] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Revised: 10/07/2010] [Accepted: 10/07/2010] [Indexed: 12/12/2022]
Abstract
Regulation of endocytic transport is controlled by an elaborate network of proteins. Rab GTP-binding proteins and their effectors have well-defined roles in mediating specific endocytic transport steps, but until recently less was known about the four mammalian dynamin-like C-terminal Eps15 homology domain (EHD) proteins that also regulate endocytic events. In recent years, however, great strides have been made in understanding the structure and function of these unique proteins. Indeed, a growing body of literature addresses EHD protein structure, interactions with binding partners, functions in mammalian cells, and the generation of various new model systems. Accordingly, this is now an opportune time to pause and review the function and mechanisms of action of EHD proteins, and to highlight some of the challenges and future directions for the field.
Collapse
Affiliation(s)
- Naava Naslavsky
- Department of Biochemistry and Molecular Biology and Eppley Cancer Center, University of Nebraska Medical Center, Omaha, NE 68130, USA
| | | |
Collapse
|
33
|
Best JM, Foell JD, Buss CR, Delisle BP, Balijepalli RC, January CT, Kamp TJ. Small GTPase Rab11b regulates degradation of surface membrane L-type Cav1.2 channels. Am J Physiol Cell Physiol 2011; 300:C1023-33. [PMID: 21248079 DOI: 10.1152/ajpcell.00288.2010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
L-type Ca(2+) channels (LTCCs) play a critical role in Ca(2+)-dependent signaling processes in a variety of cell types. The number of functional LTCCs at the plasma membrane strongly influences the strength and duration of Ca(2+) signals. Recent studies demonstrated that endosomal trafficking provides a mechanism for dynamic changes in LTCC surface membrane density. The purpose of the current study was to determine whether the small GTPase Rab11b, a known regulator of endosomal recycling, impacts plasmalemmal expression of Ca(v)1.2 LTCCs. Disruption of endogenous Rab11b function with a dominant negative Rab11b S25N mutant led to a significant 64% increase in peak L-type Ba(2+) current (I(Ba,L)) in human embryonic kidney (HEK)293 cells. Short-hairpin RNA (shRNA)-mediated knockdown of Rab11b also significantly increased peak I(Ba,L) by 66% compared when with cells transfected with control shRNA, whereas knockdown of Rab11a did not impact I(Ba,L). Rab11b S25N led to a 1.7-fold increase in plasma membrane density of hemagglutinin epitope-tagged Ca(v)1.2 expressed in HEK293 cells. Cell surface biotinylation experiments demonstrated that Rab11b S25N does not significantly impact anterograde trafficking of LTCCs to the surface membrane but rather slows degradation of plasmalemmal Ca(v)1.2 channels. We further demonstrated Rab11b expression in ventricular myocardium and showed that Rab11b S25N significantly increases peak I(Ba,L) by 98% in neonatal mouse cardiac myocytes. These findings reveal a novel role for Rab11b in limiting, rather than promoting, the plasma membrane expression of Ca(v)1.2 LTCCs in contrast to its effects on other ion channels including human ether-a-go-go-related gene (hERG) K(+) channels and cystic fibrosis transmembrane conductance regulator. This suggests Rab11b differentially regulates the trafficking of distinct cargo and extends our understanding of how endosomal transport impacts the functional expression of LTCCs.
Collapse
Affiliation(s)
- Jabe M Best
- University of Wisconsin School of Medicine and Public Health, H6/370 Clinical Science Center, 600 Highland Ave., Madison, WI 53792-3248, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Guetg N, Aziz SA, Holbro N, Turecek R, Rose T, Seddik R, Gassmann M, Moes S, Jenoe P, Oertner TG, Casanova E, Bettler B. NMDA receptor-dependent GABAB receptor internalization via CaMKII phosphorylation of serine 867 in GABAB1. Proc Natl Acad Sci U S A 2010; 107:13924-9. [PMID: 20643921 PMCID: PMC2922270 DOI: 10.1073/pnas.1000909107] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
GABAB receptors are the G-protein-coupled receptors for GABA, the main inhibitory neurotransmitter in the brain. GABAB receptors are abundant on dendritic spines, where they dampen postsynaptic excitability and inhibit Ca2+ influx through NMDA receptors when activated by spillover of GABA from neighboring GABAergic terminals. Here, we show that an excitatory signaling cascade enables spines to counteract this GABAB-mediated inhibition. We found that NMDA application to cultured hippocampal neurons promotes dynamin-dependent endocytosis of GABAB receptors. NMDA-dependent internalization of GABAB receptors requires activation of Ca2+/Calmodulin-dependent protein kinase II (CaMKII), which associates with GABAB receptors in vivo and phosphorylates serine 867 (S867) in the intracellular C terminus of the GABAB1 subunit. Blockade of either CaMKII or phosphorylation of S867 renders GABAB receptors refractory to NMDA-mediated internalization. Time-lapse two-photon imaging of organotypic hippocampal slices reveals that activation of NMDA receptors removes GABAB receptors within minutes from the surface of dendritic spines and shafts. NMDA-dependent S867 phosphorylation and internalization is predominantly detectable with the GABAB1b subunit isoform, which is the isoform that clusters with inhibitory effector K+ channels in the spines. Consistent with this, NMDA receptor activation in neurons impairs the ability of GABAB receptors to activate K+ channels. Thus, our data support that NMDA receptor activity endocytoses postsynaptic GABAB receptors through CaMKII-mediated phosphorylation of S867. This provides a means to spare NMDA receptors at individual glutamatergic synapses from reciprocal inhibition through GABAB receptors.
Collapse
Affiliation(s)
- Nicole Guetg
- Department of Biomedicine, Institute of Physiology, Pharmazentrum, University of Basel, 4056 Basel, Switzerland
| | - Said Abdel Aziz
- Department of Biomedicine, Institute of Physiology, Pharmazentrum, University of Basel, 4056 Basel, Switzerland
| | - Niklaus Holbro
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Rostislav Turecek
- Department of Biomedicine, Institute of Physiology, Pharmazentrum, University of Basel, 4056 Basel, Switzerland
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 142 20 Prague, Czech Republic; and
| | - Tobias Rose
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Riad Seddik
- Department of Biomedicine, Institute of Physiology, Pharmazentrum, University of Basel, 4056 Basel, Switzerland
| | - Martin Gassmann
- Department of Biomedicine, Institute of Physiology, Pharmazentrum, University of Basel, 4056 Basel, Switzerland
| | - Suzette Moes
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Paul Jenoe
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Thomas G. Oertner
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Emilio Casanova
- Department of Biomedicine, Institute of Physiology, Pharmazentrum, University of Basel, 4056 Basel, Switzerland
| | - Bernhard Bettler
- Department of Biomedicine, Institute of Physiology, Pharmazentrum, University of Basel, 4056 Basel, Switzerland
| |
Collapse
|
35
|
Feliciangeli S, Tardy MP, Sandoz G, Chatelain FC, Warth R, Barhanin J, Bendahhou S, Lesage F. Potassium channel silencing by constitutive endocytosis and intracellular sequestration. J Biol Chem 2009; 285:4798-805. [PMID: 19959478 DOI: 10.1074/jbc.m109.078535] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Tandem of P domains in a weak inwardly rectifying K(+) channel 1 (TWIK1) is a K(+) channel that produces unusually low levels of current. Replacement of lysine 274 by a glutamic acid (K274E) is associated with stronger currents. This mutation would prevent conjugation of a small ubiquitin modifier peptide to Lys-274, a mechanism proposed to be responsible for channel silencing. However, we found no biochemical evidence of TWIK1 sumoylation, and we showed that the conservative change K274R did not increase current, suggesting that K274E modifies TWIK1 gating through a charge effect. Now we rule out an eventual effect of K274E on TWIK1 trafficking, and we provide convincing evidence that TWIK1 silencing results from its rapid retrieval from the cell surface. TWIK1 is internalized via a dynamin-dependent mechanism and addressed to the recycling endosomal compartment. Mutation of a diisoleucine repeat located in its cytoplasmic C terminus (I293A,I294A) stabilizes TWIK1 at the plasma membrane, resulting in robust currents. The effects of I293A,I294A on channel trafficking and of K274E on channel activity are cumulative, promoting even more currents. Activation of serotoninergic receptor 5-HT(1)R or adrenoreceptor alpha2A-AR stimulates TWIK1 but has no effect on TWIK1I293A,I294A, suggesting that G(i) protein activation is a physiological signal for increasing the number of active channels at the plasma membrane.
Collapse
Affiliation(s)
- Sylvain Feliciangeli
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, and Université de Nice Sophia-Antipolis, Sophia-Antipolis, 06560 Valbonne, France
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Biel M, Wahl-Schott C, Michalakis S, Zong X. Hyperpolarization-activated cation channels: from genes to function. Physiol Rev 2009; 89:847-85. [PMID: 19584315 DOI: 10.1152/physrev.00029.2008] [Citation(s) in RCA: 760] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels comprise a small subfamily of proteins within the superfamily of pore-loop cation channels. In mammals, the HCN channel family comprises four members (HCN1-4) that are expressed in heart and nervous system. The current produced by HCN channels has been known as I(h) (or I(f) or I(q)). I(h) has also been designated as pacemaker current, because it plays a key role in controlling rhythmic activity of cardiac pacemaker cells and spontaneously firing neurons. Extensive studies over the last decade have provided convincing evidence that I(h) is also involved in a number of basic physiological processes that are not directly associated with rhythmicity. Examples for these non-pacemaking functions of I(h) are the determination of the resting membrane potential, dendritic integration, synaptic transmission, and learning. In this review we summarize recent insights into the structure, function, and cellular regulation of HCN channels. We also discuss in detail the different aspects of HCN channel physiology in the heart and nervous system. To this end, evidence on the role of individual HCN channel types arising from the analysis of HCN knockout mouse models is discussed. Finally, we provide an overview of the impact of HCN channels on the pathogenesis of several diseases and discuss recent attempts to establish HCN channels as drug targets.
Collapse
Affiliation(s)
- Martin Biel
- Center for Integrated Protein Science CIPS-M and Zentrum für Pharmaforschung, Department Pharmazie, Pharmakologie für Naturwissenschaften, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, Munich D-81377, Germany.
| | | | | | | |
Collapse
|
37
|
Cholesterol modulates the recruitment of Kv1.5 channels from Rab11-associated recycling endosome in native atrial myocytes. Proc Natl Acad Sci U S A 2009; 106:14681-6. [PMID: 19706553 DOI: 10.1073/pnas.0902809106] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cholesterol is an important determinant of cardiac electrical properties. However, underlying mechanisms are still poorly understood. Here, we examine the hypothesis that cholesterol modulates the turnover of voltage-gated potassium channels based on previous observations showing that depletion of membrane cholesterol increases the atrial repolarizing current I(Kur). Whole-cell currents and single-channel activity were recorded in rat adult atrial myocytes (AAM) or after transduction with hKv1.5-EGFP. Channel mobility and expression were studied using fluorescence recovery after photobleaching (FRAP) and 3-dimensional microscopy. In both native and transduced-AAMs, the cholesterol-depleting agent MbetaCD induced a delayed ( approximately 7 min) increase in I(Kur); the cholesterol donor LDL had an opposite effect. Single-channel recordings revealed an increased number of active Kv1.5 channels upon MbetaCD application. Whole-cell recordings indicated that this increase was not dependent on new synthesis but on trafficking of existing pools of intracellular channels whose exocytosis could be blocked by both N-ethylmaleimide and nonhydrolyzable GTP analogues. Rab11 was found to coimmunoprecipitate with hKv1.5-EGFP channels and transfection with Rab11 dominant negative (DN) but not Rab4 DN prevented the MbetaCD-induced I(Kur) increase. Three-dimensional microscopy showed a decrease in colocalization of Kv1.5 and Rab11 in MbetaCD-treated AAM. These results suggest that cholesterol regulates Kv1.5 channel expression by modulating its trafficking through the Rab11-associated recycling endosome. Therefore, this compartment provides a submembrane pool of channels readily available for recruitment into the sarcolemma of myocytes. This process could be a major mechanism for the tuning of cardiac electrical properties and might contribute to the understanding of cardiac effects of lipid-lowering drugs.
Collapse
|
38
|
Zolles G, Wenzel D, Bildl W, Schulte U, Hofmann A, Müller CS, Thumfart JO, Vlachos A, Deller T, Pfeifer A, Fleischmann BK, Roeper J, Fakler B, Klöcker N. Association with the auxiliary subunit PEX5R/Trip8b controls responsiveness of HCN channels to cAMP and adrenergic stimulation. Neuron 2009; 62:814-25. [PMID: 19555650 DOI: 10.1016/j.neuron.2009.05.008] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Revised: 03/30/2009] [Accepted: 05/07/2009] [Indexed: 11/18/2022]
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are key modulators of neuronal activity by providing the depolarizing cation current I(h) involved in rhythmogenesis, dendritic integration, and synaptic transmission. These tasks critically depend on the availability of HCN channels, which is dynamically regulated by intracellular cAMP; the range of this regulation, however, largely differs among neurons in the mammalian brain. Using affinity purification and high-resolution mass spectrometry, we identify the PEX5R/Trip8b protein as the beta subunit of HCN channels in the mammalian brain. Coassembly of PEX5R/Trip8b affects HCN channel gating in a subtype-dependent and mode-specific way: activation of HCN2 and HCN4 by cAMP is largely impaired, while gating by phosphoinositides and basal voltage-dependence remain unaffected. De novo expression of PEX5R/Trip8b in cardiomyocytes abolishes beta-adrenergic stimulation of HCN channels. These results demonstrate that PEX5R/Trip8b is an intrinsic auxiliary subunit of brain HCN channels and establish HCN-PEX5R/Trip8b coassembly as a mechanism to control the channels' responsiveness to cyclic nucleotide signaling.
Collapse
Affiliation(s)
- Gerd Zolles
- Institute of Physiology, University of Freiburg, Engesserstrasse 4, 79108 Freiburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|