1
|
Kasuya G, Zempo B, Yamamoto Y, Ryu K, Ono F, Nakajo K. Identification of KCNE6, a new member of the KCNE family of potassium channel auxiliary subunits. Commun Biol 2024; 7:1662. [PMID: 39702752 DOI: 10.1038/s42003-024-07352-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024] Open
Abstract
The KCNE family (KCNE1-5) is a group of single transmembrane auxiliary subunits for the voltage-gated K+ channel KCNQ1. The KCNQ1-KCNE complexes are crucial for numerous physiological processes including ventricular repolarization and K+ recycling in epithelial cells. We identified a new member of the KCNE family, "KCNE6", from zebrafish. We found that KCNE6 is expressed in the zebrafish heart and is involved in cardiac excitability. When co-expressed with KCNQ1, KCNE6 produces a slowly activating current like the slow delayed-rectifier K+ current (IKs) induced by KCNE1, despite the fact that the KCNE6 amino acid sequence has the highest similarity to that of KCNE3, which forms a constitutively open channel with KCNQ1. The kcne6 nucleotide sequences exist throughout vertebrates, including humans, although only the KCNE6 proteins of lower vertebrates, up to marsupials, can modulate KCNQ1, and it has become a pseudogene in eutherians. Our findings will facilitate a better understanding of how the KCNE family has evolved to modulate KCNQ1.
Collapse
Affiliation(s)
- Go Kasuya
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| | - Buntaro Zempo
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Yasuhiro Yamamoto
- Department of Physiology, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki, 569-8686, Japan
| | - Kaei Ryu
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Fumihito Ono
- Department of Physiology, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki, 569-8686, Japan
| | - Koichi Nakajo
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| |
Collapse
|
2
|
Peters CH, Singh RK, Langley AA, Nichols WG, Ferris HR, Jeffrey DA, Proenza C, Bankston JR. LRMP inhibits cAMP potentiation of HCN4 channels by disrupting intramolecular signal transduction. eLife 2024; 12:RP92411. [PMID: 38652113 PMCID: PMC11037915 DOI: 10.7554/elife.92411] [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] [Indexed: 04/25/2024] Open
Abstract
Lymphoid restricted membrane protein (LRMP) is a specific regulator of the hyperpolarization-activated cyclic nucleotide-sensitive isoform 4 (HCN4) channel. LRMP prevents cAMP-dependent potentiation of HCN4, but the interaction domains, mechanisms of action, and basis for isoform-specificity remain unknown. Here, we identify the domains of LRMP essential for this regulation, show that LRMP acts by disrupting the intramolecular signal transduction between cyclic nucleotide binding and gating, and demonstrate that multiple unique regions in HCN4 are required for LRMP isoform-specificity. Using patch clamp electrophysiology and Förster resonance energy transfer (FRET), we identified the initial 227 residues of LRMP and the N-terminus of HCN4 as necessary for LRMP to associate with HCN4. We found that the HCN4 N-terminus and HCN4-specific residues in the C-linker are necessary for regulation of HCN4 by LRMP. Finally, we demonstrated that LRMP-regulation can be conferred to HCN2 by addition of the HCN4 N-terminus along with mutation of five residues in the S5 region and C-linker to the cognate HCN4 residues. Taken together, these results suggest that LRMP inhibits HCN4 through an isoform-specific interaction involving the N-terminals of both proteins that prevents the transduction of cAMP binding into a change in channel gating, most likely via an HCN4-specific orientation of the N-terminus, C-linker, and S4-S5 linker.
Collapse
Affiliation(s)
- Colin H Peters
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Rohit K Singh
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical CampusAuroraUnited States
- Skaggs School of Pharmacy, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Avery A Langley
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - William G Nichols
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Hannah R Ferris
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Danielle A Jeffrey
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Catherine Proenza
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical CampusAuroraUnited States
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - John R Bankston
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical CampusAuroraUnited States
| |
Collapse
|
3
|
Cámara-Checa A, Rubio-Alarcón M, Dago M, Crespo-García T, Rapún J, Marín M, Tamargo J, Gómez R, Caballero R, Delpón E. Reply to Benndorff and DiFrancesco: Reliable human HCN4 single-channel recordings using the cell-attached configuration in expression systems. Proc Natl Acad Sci U S A 2024; 121:e2402992121. [PMID: 38588416 PMCID: PMC11032473 DOI: 10.1073/pnas.2402992121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024] Open
Affiliation(s)
- Anabel Cámara-Checa
- Cardiac Cellular Electrophysiology Group at the Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Marcos Rubio-Alarcón
- Cardiac Cellular Electrophysiology Group at the Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - María Dago
- Cardiac Cellular Electrophysiology Group at the Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Teresa Crespo-García
- Cardiac Cellular Electrophysiology Group at the Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Josu Rapún
- Cardiac Cellular Electrophysiology Group at the Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - María Marín
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Juan Tamargo
- Cardiac Cellular Electrophysiology Group at the Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
| | - Ricardo Gómez
- Cardiac Cellular Electrophysiology Group at the Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Ricardo Caballero
- Cardiac Cellular Electrophysiology Group at the Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Eva Delpón
- Cardiac Cellular Electrophysiology Group at the Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| |
Collapse
|
4
|
Fu Q, Wang Y, Yan C, Xiang YK. Phosphodiesterase in heart and vessels: from physiology to diseases. Physiol Rev 2024; 104:765-834. [PMID: 37971403 PMCID: PMC11281825 DOI: 10.1152/physrev.00015.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/17/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
Phosphodiesterases (PDEs) are a superfamily of enzymes that hydrolyze cyclic nucleotides, including cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Both cyclic nucleotides are critical secondary messengers in the neurohormonal regulation in the cardiovascular system. PDEs precisely control spatiotemporal subcellular distribution of cyclic nucleotides in a cell- and tissue-specific manner, playing critical roles in physiological responses to hormone stimulation in the heart and vessels. Dysregulation of PDEs has been linked to the development of several cardiovascular diseases, such as hypertension, aneurysm, atherosclerosis, arrhythmia, and heart failure. Targeting these enzymes has been proven effective in treating cardiovascular diseases and is an attractive and promising strategy for the development of new drugs. In this review, we discuss the current understanding of the complex regulation of PDE isoforms in cardiovascular function, highlighting the divergent and even opposing roles of PDE isoforms in different pathogenesis.
Collapse
Affiliation(s)
- Qin Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Ying Wang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Chen Yan
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, New York, United States
| | - Yang K Xiang
- Department of Pharmacology, University of California at Davis, Davis, California, United States
- Department of Veterans Affairs Northern California Healthcare System, Mather, California, United States
| |
Collapse
|
5
|
Porro A, Saponaro A, Castelli R, Introini B, Hafez Alkotob A, Ranjbari G, Enke U, Kusch J, Benndorf K, Santoro B, DiFrancesco D, Thiel G, Moroni A. A high affinity switch for cAMP in the HCN pacemaker channels. Nat Commun 2024; 15:843. [PMID: 38287019 PMCID: PMC10825183 DOI: 10.1038/s41467-024-45136-y] [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: 05/25/2023] [Accepted: 01/16/2024] [Indexed: 01/31/2024] Open
Abstract
Binding of cAMP to Hyperpolarization activated cyclic nucleotide gated (HCN) channels facilitates pore opening. It is unclear why the isolated cyclic nucleotide binding domain (CNBD) displays in vitro lower affinity for cAMP than the full-length channel in patch experiments. Here we show that HCN are endowed with an affinity switch for cAMP. Alpha helices D and E, downstream of the cyclic nucleotide binding domain (CNBD), bind to and stabilize the holo CNBD in a high affinity state. These helices increase by 30-fold cAMP efficacy and affinity measured in patch clamp and ITC, respectively. We further show that helices D and E regulate affinity by interacting with helix C of the CNBD, similarly to the regulatory protein TRIP8b. Our results uncover an intramolecular mechanism whereby changes in binding affinity, rather than changes in cAMP concentration, can modulate HCN channels, adding another layer to the complex regulation of their activity.
Collapse
Affiliation(s)
| | - Andrea Saponaro
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milano, Italy
| | | | - Bianca Introini
- Department of Biosciences, University of Milan, Milano, Italy
| | | | - Golnaz Ranjbari
- Department of Biosciences, University of Milan, Milano, Italy
| | - Uta Enke
- Institut für Physiologie II, Universitätsklinikum Jena, Jena, Germany
| | - Jana Kusch
- Institut für Physiologie II, Universitätsklinikum Jena, Jena, Germany
| | - Klaus Benndorf
- Institut für Physiologie II, Universitätsklinikum Jena, Jena, Germany
| | - Bina Santoro
- Department of Neuroscience, Zuckerman Institute, Columbia University, New York, NY, USA
| | | | - Gerhard Thiel
- Department of Biology, TU-Darmstadt, Darmstadt, Germany
| | - Anna Moroni
- Department of Biosciences, University of Milan, Milano, Italy.
- Institute of Biophysics Milan, Consiglio Nazionale delle Ricerche, Milano, Italy.
| |
Collapse
|
6
|
Peters CH, Singh RK, Langley AA, Nichols WG, Ferris HR, Jeffrey DA, Proenza C, Bankston JR. LRMP inhibits cAMP potentiation of HCN4 channels by disrupting intramolecular signal transduction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.29.555242. [PMID: 37693562 PMCID: PMC10491135 DOI: 10.1101/2023.08.29.555242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Lymphoid restricted membrane protein (LRMP) is a specific regulator of the hyperpolarization-activated cyclic nucleotide-sensitive isoform 4 (HCN4) channel. LRMP prevents cAMP-dependent potentiation of HCN4 but the interaction domains, mechanisms of action, and basis for isoform-specificity remain unknown. Here we identify the domains of LRMP essential for regulation. We show that LRMP acts by disrupting the intramolecular signal transduction between cyclic nucleotide binding and gating. And we demonstrate that multiple unique regions in HCN4 are required for LRMP isoform-specificity. Using patch clamp electrophysiology and Förster resonance energy transfer (FRET), we showed that the initial 227 residues of LRMP and the N-terminus of HCN4 are necessary for LRMP to interact with HCN4. We found that the HCN4 N-terminus and HCN4-specific residues in the C-linker are necessary for regulation of HCN4 by LRMP. And we demonstrate that LRMP-regulation can be conferred to HCN2 by addition of the HCN4 N-terminus along with mutation of 5 residues in the S5 region and C-linker to the cognate HCN4 residues. Taken together, these results suggest that LRMP inhibits HCN4 through an isoform-specific interaction involving the N-terminals of both proteins that prevents the transduction of cAMP binding into a change in channel gating via an HCN4-specific orientation of the N-terminus, C-linker, and S4-S5 linker.
Collapse
Affiliation(s)
- Colin H Peters
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, 12800 E. 19 Avenue, Aurora, CO 80045
| | - Rohit K Singh
- Skaggs School of Pharmacy, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E. Montview Boulevard, Aurora, CO 80045
| | - Avery A Langley
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, 12800 E. 19 Avenue, Aurora, CO 80045
| | - William G Nichols
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, 12800 E. 19 Avenue, Aurora, CO 80045
| | - Hannah R Ferris
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, 12800 E. 19 Avenue, Aurora, CO 80045
| | - Danielle A Jeffrey
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, 12800 E. 19 Avenue, Aurora, CO 80045
| | - Catherine Proenza
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, 12800 E. 19 Avenue, Aurora, CO 80045
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, 12631 E. 17 Avenue, Aurora, CO 80045
| | - John R Bankston
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, 12800 E. 19 Avenue, Aurora, CO 80045
| |
Collapse
|
7
|
Cámara-Checa A, Perin F, Rubio-Alarcón M, Dago M, Crespo-García T, Rapún J, Marín M, Cebrián J, Gómez R, Bermúdez-Jiménez F, Monserrat L, Tamargo J, Caballero R, Jiménez-Jáimez J, Delpón E. A gain-of-function HCN4 mutant in the HCN domain is responsible for inappropriate sinus tachycardia in a Spanish family. Proc Natl Acad Sci U S A 2023; 120:e2305135120. [PMID: 38032931 PMCID: PMC10710060 DOI: 10.1073/pnas.2305135120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 10/12/2023] [Indexed: 12/02/2023] Open
Abstract
In a family with inappropriate sinus tachycardia (IST), we identified a mutation (p.V240M) of the hyperpolarization-activated cyclic nucleotide-gated type 4 (HCN4) channel, which contributes to the pacemaker current (If) in human sinoatrial node cells. Here, we clinically study fifteen family members and functionally analyze the p.V240M variant. Macroscopic (IHCN4) and single-channel currents were recorded using patch-clamp in cells expressing human native (WT) and/or p.V240M HCN4 channels. All p.V240M mutation carriers exhibited IST that was accompanied by cardiomyopathy in adults. IHCN4 generated by p.V240M channels either alone or in combination with WT was significantly greater than that generated by WT channels alone. The variant, which lies in the N-terminal HCN domain, increased the single-channel conductance and opening frequency and probability of HCN4 channels. Conversely, it did not modify the channel sensitivity for cAMP and ivabradine or the level of expression at the membrane. Treatment with ivabradine based on functional data reversed the IST and the cardiomyopathy of the carriers. In computer simulations, the p.V240M gain-of-function variant increases If and beating rate and thus explains the IST of the carriers. The results demonstrate the importance of the unique HCN domain in HCN4, which stabilizes the channels in the closed state.
Collapse
Affiliation(s)
- Anabel Cámara-Checa
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Francesca Perin
- Department of Pediatric Cardiology, Virgen de las Nieves University Hospital, Granada18014, Spain
- Instituto de Investigación Biosanitaria de Granada, Granada18014, Spain
| | - Marcos Rubio-Alarcón
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - María Dago
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Teresa Crespo-García
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Josu Rapún
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - María Marín
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Jorge Cebrián
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Ricardo Gómez
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Francisco Bermúdez-Jiménez
- Department of Pediatric Cardiology, Virgen de las Nieves University Hospital, Granada18014, Spain
- Instituto de Investigación Biosanitaria de Granada, Granada18014, Spain
- Centro Nacional de Investigaciones Cardiovasculares, Madrid28029, Spain
| | - Lorenzo Monserrat
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
- Health in Code Sociedad Limitada, A Coruña15008, Spain
| | - Juan Tamargo
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
| | - Ricardo Caballero
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Juan Jiménez-Jáimez
- Department of Pediatric Cardiology, Virgen de las Nieves University Hospital, Granada18014, Spain
- Instituto de Investigación Biosanitaria de Granada, Granada18014, Spain
| | - Eva Delpón
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| |
Collapse
|
8
|
Prüschenk S, Majer M, Schlossmann J. Novel Functional Features of cGMP Substrate Proteins IRAG1 and IRAG2. Int J Mol Sci 2023; 24:9837. [PMID: 37372987 DOI: 10.3390/ijms24129837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
The inositol triphosphate-associated proteins IRAG1 and IRAG2 are cGMP kinase substrate proteins that regulate intracellular Ca2+. Previously, IRAG1 was discovered as a 125 kDa membrane protein at the endoplasmic reticulum, which is associated with the intracellular Ca2+ channel IP3R-I and the PKGIβ and inhibits IP3R-I upon PKGIβ-mediated phosphorylation. IRAG2 is a 75 kDa membrane protein homolog of IRAG1 and was recently also determined as a PKGI substrate. Several (patho-)physiological functions of IRAG1 and IRAG2 were meanwhile elucidated in a variety of human and murine tissues, e.g., of IRAG1 in various smooth muscles, heart, platelets, and other blood cells, of IRAG2 in the pancreas, heart, platelets, and taste cells. Hence, lack of IRAG1 or IRAG2 leads to diverse phenotypes in these organs, e.g., smooth muscle and platelet disorders or secretory deficiency, respectively. This review aims to highlight the recent research regarding these two regulatory proteins to envision their molecular and (patho-)physiological tasks and to unravel their functional interplay as possible (patho-)physiological counterparts.
Collapse
Affiliation(s)
- Sally Prüschenk
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Regensburg, 93040 Regensburg, Germany
| | - Michael Majer
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Regensburg, 93040 Regensburg, Germany
| | - Jens Schlossmann
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Regensburg, 93040 Regensburg, Germany
| |
Collapse
|
9
|
Peters CH, Singh RK, Bankston JR, Proenza C. Regulation of HCN Channels by Protein Interactions. Front Physiol 2022; 13:928507. [PMID: 35795651 PMCID: PMC9251338 DOI: 10.3389/fphys.2022.928507] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/01/2022] [Indexed: 11/30/2022] Open
Abstract
Hyperpolarization-activated, cyclic nucleotide-sensitive (HCN) channels are key regulators of subthreshold membrane potentials in excitable cells. The four mammalian HCN channel isoforms, HCN1-HCN4, are expressed throughout the body, where they contribute to diverse physiological processes including cardiac pacemaking, sleep-wakefulness cycles, memory, and somatic sensation. While all HCN channel isoforms produce currents when expressed by themselves, an emerging list of interacting proteins shape HCN channel excitability to influence the physiologically relevant output. The best studied of these regulatory proteins is the auxiliary subunit, TRIP8b, which binds to multiple sites in the C-terminus of the HCN channels to regulate expression and disrupt cAMP binding to fine-tune neuronal HCN channel excitability. Less is known about the mechanisms of action of other HCN channel interaction partners like filamin A, Src tyrosine kinase, and MinK-related peptides, which have a range of effects on HCN channel gating and expression. More recently, the inositol trisphosphate receptor-associated cGMP-kinase substrates IRAG1 and LRMP (also known as IRAG2), were discovered as specific regulators of the HCN4 isoform. This review summarizes the known protein interaction partners of HCN channels and their mechanisms of action and identifies gaps in our knowledge.
Collapse
Affiliation(s)
- Colin H. Peters
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Rohit K. Singh
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - John R. Bankston
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Catherine Proenza
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- *Correspondence: Catherine Proenza,
| |
Collapse
|
10
|
Prüschenk S, Schlossmann J. Function of IRAG2 Is Modulated by NO/cGMP in Murine Platelets. Int J Mol Sci 2022; 23:ijms23126695. [PMID: 35743138 PMCID: PMC9223716 DOI: 10.3390/ijms23126695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/08/2022] [Accepted: 06/14/2022] [Indexed: 01/27/2023] Open
Abstract
Inositol 1,4,5-triphosphate receptor-associated 2 (IRAG2) is a type II membrane protein located at the endoplasmic reticulum. It is a homologue of inositol 1,4,5-triphosphate receptor-associated cGMP kinase substrate 1 (IRAG1), a substrate protein of cGMP-dependent protein kinase I (PKGI), and is among others expressed in platelets. Here, we studied if IRAG2 is also located in platelets and might be a substrate protein of PKGI. IRAG2 was detected in platelets of IRAG2-WT animals but not in those of IRAG2-KO animals. Next, we validated by co-immunoprecipitation studies that IRAG2 is associated with IP3R1-3. No direct stable interaction with PKGIβ or with IRAG1 was observed. Phosphorylation of IRAG2 in murine platelets using a Ser/Thr-specific phospho-antibody was found in vitro and ex vivo upon cGMP stimulation. To gain insight into the function of IRAG2, platelet aggregation studies were performed using thrombin and collagen as agonists for treatment of isolated IRAG2-WT or IRAG2-KO platelets. Interestingly, platelet aggregation was reduced in the absence of IRAG2. Pretreatment of wild type or IRAG2-KO platelets with sodium nitroprusside (SNP) or 8-pCPT-cGMP revealed a further reduction in platelet aggregation in the absence of IRAG2. These results show that IRAG2 is a substrate of PKGI in murine platelets. Furthermore, our results indicate that IRAG2 is involved in the induction of thrombin- or collagen-induced platelet aggregation and that this effect is enhanced by cGMP-dependent phosphorylation of IRAG2. As IRAG1 was previously shown to inhibit platelet aggregation in a cGMP-dependent manner, it can be speculated that IRAG2 exerts an opposing function and might be an IRAG1 counterpart in murine platelets.
Collapse
|
11
|
Okumura W, Kozono T, Sato H, Matsui H, Takagi T, Tonozuka T, Nishikawa A. Jaw1/LRMP increases Ca 2+ influx upon GPCR stimulation with heterogeneous effect on the activity of each ITPR subtype. Sci Rep 2022; 12:9476. [PMID: 35676525 PMCID: PMC9177832 DOI: 10.1038/s41598-022-13620-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/11/2022] [Indexed: 11/24/2022] Open
Abstract
Ca2+ influx upon G protein-coupled receptor (GPCR) stimulation is observed as a cytosolic Ca2+ concentration oscillation crucial to initiating downstream responses including cell proliferation, differentiation, and cell–cell communication. Although Jaw1 is known to interact with inositol 1,4,5-triphosphate receptor (ITPRs), Ca2+ channels on the endoplasmic reticulum, the function of Jaw1 in the Ca2+ dynamics with physiological stimulation remains unclear. In this study, using inducible Jaw1-expressing HEK293 cells, we showed that Jaw1 increases Ca2+ influx by GPCR stimulation via changing the Ca2+ influx oscillation pattern. Furthermore, we showed that Jaw1 increases the Ca2+ release activity of all ITPR subtypes in a subtly different manner. It is well known that the Ca2+ influx oscillation pattern varies from cell type to cell type, therefore these findings provide an insight into the relationship between the heterogeneous Ca2+ dynamics and the specific ITPR and Jaw1 expression patterns.
Collapse
Affiliation(s)
- Wataru Okumura
- Department of Food and Energy Systems Science, Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, Tokyo, 184-8588, Japan
| | - Takuma Kozono
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, Tokyo, 183-8509, Japan
| | - Hiroyuki Sato
- Cooperative Major in Advanced Health Science, Tokyo University of Agriculture and Technology, Tokyo, 184-8588, Japan
| | - Hitomi Matsui
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, 183-8509, Japan
| | - Tsubasa Takagi
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, 183-8509, Japan
| | - Takashi Tonozuka
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, 183-8509, Japan
| | - Atsushi Nishikawa
- Department of Food and Energy Systems Science, Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, Tokyo, 184-8588, Japan. .,Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, Tokyo, 183-8509, Japan. .,Cooperative Major in Advanced Health Science, Tokyo University of Agriculture and Technology, Tokyo, 184-8588, Japan. .,Department of Applied Biological Chemistry, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, 183-8509, Japan.
| |
Collapse
|
12
|
Ng LCT, Li YX, Van Petegem F, Accili EA. Altered cyclic nucleotide-binding and pore opening in a diseased human HCN4 channel. Biophys J 2022; 121:1166-1183. [PMID: 35219649 PMCID: PMC9034293 DOI: 10.1016/j.bpj.2022.02.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/20/2021] [Accepted: 02/22/2022] [Indexed: 11/16/2022] Open
Abstract
A growing number of nonsynonymous mutations in the human HCN4 channel gene, the major component of the funny channel of the sinoatrial node, are associated with disease but how they impact channel structure and function, and, thus, how they result in disease, is not clear for any of them. Here, we study the S672R mutation, in the cyclic nucleotide-binding domain of the channel, which has been associated with an inherited bradycardia in an Italian family. This may be the best studied of all known mutations, yet the underlying molecular and atomistic mechanisms remain unclear and controversial. We combine measurements of binding by isothermal titration calorimetry to a naturally occurring tetramer of the HCN4 C-terminal region with a mathematical model to show that weaker binding of cAMP to the mutant channel contributes to a lower level of facilitation of channel opening at submicromolar ligand concentrations but that, in general, facilitation occurs over a range that is similar between the mutant and wild-type because of enhanced opening of the mutant channel when liganded. We also show that the binding affinity for cGMP, which produces the same maximum facilitation of HCN4 opening as cAMP, is weaker in the mutant HCN4 channel but that, for both wild-type and mutant, high-affinity binding of cGMP occurs in a range of concentrations below 1 μM. Thus, binding of cGMP to the HCN4 channel may be relevant normally in vivo and reduced binding of cGMP, as well as cAMP, to the mutant channel may contribute to the reduced resting heart rate observed in the affected family.
Collapse
Affiliation(s)
- Leo C T Ng
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Yue Xian Li
- Department of Mathematics, University of British Columbia, Vancouver, Canada
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Eric A Accili
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
13
|
Prüschenk S, Majer M, Schreiber R, Schlossmann J. IRAG2 Interacts with IP 3-Receptor Types 1, 2, and 3 and Regulates Intracellular Ca 2+ in Murine Pancreatic Acinar Cells. Int J Mol Sci 2021; 22:ijms222413409. [PMID: 34948204 PMCID: PMC8707672 DOI: 10.3390/ijms222413409] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 01/18/2023] Open
Abstract
The inositol 1,4,5-triphosphate receptor-associated 2 (IRAG2) is also known as Jaw1 or lymphoid-restricted membrane protein (LRMP) and shares homology with the inositol 1,4,5-triphosphate receptor-associated cGMP kinase substrate 1 (IRAG1). IRAG1 interacts with inositol trisphosphate receptors (IP3 receptors /IP3R) via its coiled-coil domain and modulates Ca2+ release from intracellular stores. Due to the homology of IRAG1 and IRAG2, especially in its coiled-coil domain, it is possible that IRAG2 has similar interaction partners like IRAG1 and that IRAG2 also modulates intracellular Ca2+ signaling. In our study, we localized IRAG2 in pancreatic acinar cells of the exocrine pancreas, and we investigated the interaction of IRAG2 with IP3 receptors and its impact on intracellular Ca2+ signaling and exocrine pancreatic function, like amylase secretion. We detected the interaction of IRAG2 with different subtypes of IP3R and altered Ca2+ release in pancreatic acinar cells from mice lacking IRAG2. IRAG2 deficiency decreased basal levels of intracellular Ca2+, suggesting that IRAG2 leads to activation of IP3R under unstimulated basal conditions. Moreover, we observed that loss of IRAG2 impacts the secretion of amylase. Our data, therefore, suggest that IRAG2 modulates intracellular Ca2+ signaling, which regulates exocrine pancreatic function.
Collapse
Affiliation(s)
- Sally Prüschenk
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Regensburg, 93040 Regensburg, Germany; (S.P.); (M.M.)
| | - Michael Majer
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Regensburg, 93040 Regensburg, Germany; (S.P.); (M.M.)
| | - Rainer Schreiber
- Institute of Physiology, University of Regensburg, 93040 Regensburg, Germany;
| | - Jens Schlossmann
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Regensburg, 93040 Regensburg, Germany; (S.P.); (M.M.)
- Correspondence: ; Tel.: +49-941-943-4770
| |
Collapse
|
14
|
Wu X, Kong C, Wu Y. Long intergenic non-protein coding RNA 1094 (LINC01094) promotes the progression of breast cancer (BC) by regulating the microRNA-340-5p (miR-340-5p)/E2F transcription factor 3 (E2F3) axis. Bioengineered 2021; 12:9046-9057. [PMID: 34657558 PMCID: PMC8806954 DOI: 10.1080/21655979.2021.1993715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The present study was targeted at investigating the effects of long intergenic non-protein coding RNA 1094 on breast cancer (BC) cell proliferation, apoptosis, and cell cycle and its related mechanism. In this study, Western blot and quantitative real-time polymerase chain reaction (qRT-PCR) were conducted to detect the expressions of LINC01094, microRNA (miRNA, miR)-340-5p, and E2F transcription factor 3 (E2F3) in BC tissues and cells. With transfection, LINC01094 and miR-340-5p expressions were selectively up-regulated or down-regulated in BC cell lines, and then cell proliferation, cell cycle, and apoptosis were examined by cell counting kit-8 (CCK-8), 5-bromo-2ʹ-deoxyuridine (BrdU), and flow cytometry assays. Bioinformatics was utilized to predict the targeted relationships between miR-340-5p and LINC01094, as well as miR-340-5p and E2F3 mRNA 3ʹ-untranslated region (3ʹUTR), and RNA immunoprecipitation (RIP) assay and dual-luciferase reporter gene assay were employed to validate them. It was revealed that, LINC01094 expression was enhanced in BC cells and tissues, and LINC01094 overexpression promoted BC cell proliferation, accelerated cell cycle progression, and inhibited apoptosis while knocking down LINC01094 worked oppositely. LINC01094 directly targeted miR-340-5p and negatively regulated its expression in BC cells. Besides, E2F3 was substantiated to be the target gene of miR-340-5p, and E2F3 expression could be indirectly and positively modulated by LINC01094. All in all, LINC01094 promotes BC cell proliferation and cell cycle progression and inhibits apoptosis via modulating miR-340-5p/E2F3 molecular axis.
Collapse
Affiliation(s)
- Xia Wu
- Department of Oncology, The Third People's Hospital of Linyi, Linyi, Shandong, China
| | - Cui Kong
- Department of Personnel, The Third People's Hospital of Linyi, Linyi, Shandong, China
| | - Yilei Wu
- Department of Both Glandular and Hemangioma Families, Shandong Provincial Third Hospital, Jinan, Shandong, China
| |
Collapse
|
15
|
Peters CH, Liu PW, Morotti S, Gantz SC, Grandi E, Bean BP, Proenza C. Bidirectional flow of the funny current (I f) during the pacemaking cycle in murine sinoatrial node myocytes. Proc Natl Acad Sci U S A 2021; 118:e2104668118. [PMID: 34260402 PMCID: PMC8285948 DOI: 10.1073/pnas.2104668118] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Sinoatrial node myocytes (SAMs) act as cardiac pacemaker cells by firing spontaneous action potentials (APs) that initiate each heartbeat. The funny current (If) is critical for the generation of these spontaneous APs; however, its precise role during the pacemaking cycle remains unresolved. Here, we used the AP-clamp technique to quantify If during the cardiac cycle in mouse SAMs. We found that If is persistently active throughout the sinoatrial AP, with surprisingly little voltage-dependent gating. As a consequence, it carries both inward and outward current around its reversal potential of -30 mV. Despite operating at only 2 to 5% of its maximal conductance, If carries a substantial fraction of both depolarizing and repolarizing net charge movement during the firing cycle. We also show that β-adrenergic receptor stimulation increases the percentage of net depolarizing charge moved by If, consistent with a contribution of If to the fight-or-flight increase in heart rate. These properties were confirmed by heterologously expressed HCN4 channels and by mathematical models of If Modeling further suggested that the slow rates of activation and deactivation of the HCN4 isoform underlie the persistent activity of If during the sinoatrial AP. These results establish a new conceptual framework for the role of If in pacemaking, in which it operates at a very small fraction of maximal activation but nevertheless drives membrane potential oscillations in SAMs by providing substantial driving force in both inward and outward directions.
Collapse
Affiliation(s)
- Colin H Peters
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Pin W Liu
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115
| | - Stefano Morotti
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Stephanie C Gantz
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Eleonora Grandi
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Bruce P Bean
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115
| | - Catherine Proenza
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045;
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|