1
|
Covarrubias M, Liang Q, Nguyen-Phuong L, Kennedy KJ, Alexander TD, Sam A. Structural insights into the function, dysfunction and modulation of Kv3 channels. Neuropharmacology 2025; 275:110483. [PMID: 40288604 DOI: 10.1016/j.neuropharm.2025.110483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 04/03/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
The third subfamily of voltage-gated K+ (Kv) channels includes four members, Kv3.1, Kv3.2, Kv3.3 and Kv3.4. Fast gating and activation at relatively depolarized membrane potentials allows Kv3 channels to be major drivers of fast action potential repolarization in the nervous system. Consequently, they help determine the fast-spiking phenotype of inhibitory interneurons and regulate fast synaptic transmission at glutamatergic synapses and the neuromuscular junction. Recent studies from our group and a team of collaborators have used cryo-EM to demonstrate the surprising gating role of the Kv3.1 cytoplasmic T1 domain, the structural basis of a developmental epileptic encephalopathy caused by the Kv3.2-C125Y variant and the mechanism of action of positive allosteric modulators involving unexpected interactions and conformational changes in Kv3.1 and Kv3.2. Furthermore, our recent work has shown that Kv3.4 regulates use-dependent spike broadening in a manner that depends on gating modulation by phosphorylation of the channel's N-terminal inactivation domain, which can impact activity-dependent synaptic facilitation. Here, we review and integrate these studies to provide a perspective on our current understanding of Kv3 channel function, dysfunction and pain modulation in the nervous system.
Collapse
Affiliation(s)
- Manuel Covarrubias
- Department of Neuroscience, Sidney Kimmel Medical College of Thomas Jefferson University, Bluemle Life Science Building, 233 South 10th Street, Room 231, Philadelphia, PA, 19107, USA; Vickie and Jack Farber Institute for Neuroscience, USA; Jefferson Synaptic Biology Center, USA.
| | - Qiansheng Liang
- Department of Neuroscience, Sidney Kimmel Medical College of Thomas Jefferson University, Bluemle Life Science Building, 233 South 10th Street, Room 231, Philadelphia, PA, 19107, USA; Vickie and Jack Farber Institute for Neuroscience, USA; Jefferson Synaptic Biology Center, USA
| | - Linh Nguyen-Phuong
- Department of Neuroscience, Sidney Kimmel Medical College of Thomas Jefferson University, Bluemle Life Science Building, 233 South 10th Street, Room 231, Philadelphia, PA, 19107, USA; Vickie and Jack Farber Institute for Neuroscience, USA; Jefferson Synaptic Biology Center, USA
| | - Kyle J Kennedy
- Department of Neuroscience, Sidney Kimmel Medical College of Thomas Jefferson University, Bluemle Life Science Building, 233 South 10th Street, Room 231, Philadelphia, PA, 19107, USA; Vickie and Jack Farber Institute for Neuroscience, USA; Jefferson Synaptic Biology Center, USA
| | - Tyler D Alexander
- Department of Neuroscience, Sidney Kimmel Medical College of Thomas Jefferson University, Bluemle Life Science Building, 233 South 10th Street, Room 231, Philadelphia, PA, 19107, USA; Vickie and Jack Farber Institute for Neuroscience, USA; Jefferson Synaptic Biology Center, USA
| | - Andrew Sam
- Department of Neuroscience, Sidney Kimmel Medical College of Thomas Jefferson University, Bluemle Life Science Building, 233 South 10th Street, Room 231, Philadelphia, PA, 19107, USA; Vickie and Jack Farber Institute for Neuroscience, USA; Jefferson Synaptic Biology Center, USA
| |
Collapse
|
2
|
Agarwal S, Kim ED, Lee S, Simon A, Accardi A, Nimigean CM. Ball-and-chain inactivation of a human large conductance calcium-activated potassium channel. Nat Commun 2025; 16:1769. [PMID: 39971906 PMCID: PMC11840039 DOI: 10.1038/s41467-025-56844-4] [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: 09/23/2024] [Accepted: 02/03/2025] [Indexed: 02/21/2025] Open
Abstract
BK channels are large-conductance calcium (Ca2+)-activated potassium channels crucial for neuronal excitability, muscle contraction, and neurotransmitter release. The pore-forming (α) subunits co-assemble with auxiliary (β and γ) subunits that modulate their function. Previous studies demonstrated that the N-termini of β2-subunits can inactivate BK channels, but with no structural correlate. Here, we investigate BK β2-subunit inactivation using cryo-electron microscopy, electrophysiology and molecular dynamics simulations. We find that the β2 N-terminus occludes the pore only in the Ca2+-bound open state, via a ball-and-chain mechanism. The first three hydrophobic residues of β2 are crucial for occlusion, while the remainder of the N-terminus remains flexible. Neither the closed channel conformation obtained in the absence of Ca2+ nor an intermediate conformation found in the presence of Ca2+ show density for the N-terminus of the β2 subunit in their pore, likely due to narrower side access portals preventing their entry into the channel pore.
Collapse
Affiliation(s)
- Shubhangi Agarwal
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Elizabeth D Kim
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Sangyun Lee
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Alexander Simon
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Alessio Accardi
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Crina M Nimigean
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA.
- Department of Physiology and Biophysics, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA.
| |
Collapse
|
3
|
Wu Y, Yan Y, Yang Y, Bian S, Rivetta A, Allen K, Sigworth FJ. CryoEM structures of Kv1.2 potassium channels, conducting and non-conducting. eLife 2025; 12:RP89459. [PMID: 39945513 PMCID: PMC11825129 DOI: 10.7554/elife.89459] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2025] Open
Abstract
We present near-atomic-resolution cryoEM structures of the mammalian voltage-gated potassium channel Kv1.2 in open, C-type inactivated, toxin-blocked and sodium-bound states at 3.2 Å, 2.5 Å, 3.2 Å, and 2.9 Å. These structures, all obtained at nominally zero membrane potential in detergent micelles, reveal distinct ion-occupancy patterns in the selectivity filter. The first two structures are very similar to those reported in the related Shaker channel and the much-studied Kv1.2-2.1 chimeric channel. On the other hand, two new structures show unexpected patterns of ion occupancy. First, the toxin α-Dendrotoxin, like Charybdotoxin, is seen to attach to the negatively-charged channel outer mouth, and a lysine residue penetrates into the selectivity filter, with the terminal amine coordinated by carbonyls, partially disrupting the outermost ion-binding site. In the remainder of the filter two densities of bound ions are observed, rather than three as observed with other toxin-blocked Kv channels. Second, a structure of Kv1.2 in Na+ solution does not show collapse or destabilization of the selectivity filter, but instead shows an intact selectivity filter with ion density in each binding site. We also attempted to image the C-type inactivated Kv1.2 W366F channel in Na+ solution, but the protein conformation was seen to be highly variable and only a low-resolution structure could be obtained. These findings present new insights into the stability of the selectivity filter and the mechanism of toxin block of this intensively studied, voltage-gated potassium channel.
Collapse
Affiliation(s)
- Yangyu Wu
- Department of Cellular and Molecular Physiology, Yale University School of MedicineNew HavenUnited States
| | - Yangyang Yan
- Department of Cellular and Molecular Physiology, Yale University School of MedicineNew HavenUnited States
| | - Youshan Yang
- Department of Cellular and Molecular Physiology, Yale University School of MedicineNew HavenUnited States
| | - Shumin Bian
- Department of Cellular and Molecular Physiology, Yale University School of MedicineNew HavenUnited States
| | - Alberto Rivetta
- Department of Cellular and Molecular Physiology, Yale University School of MedicineNew HavenUnited States
| | - Ken Allen
- Department of Cellular and Molecular Physiology, Yale University School of MedicineNew HavenUnited States
| | - Fred J Sigworth
- Department of Cellular and Molecular Physiology, Yale University School of MedicineNew HavenUnited States
| |
Collapse
|
4
|
Sykes A, Caruth L, Setia Verma S, Hoshi T, Deutsch C. Disease-associated Kv1.3 variants are energy compromised with impaired nascent chain folding. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.17.631970. [PMID: 39868087 PMCID: PMC11761497 DOI: 10.1101/2025.01.17.631970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Human Kv1.3, encoded by KCNA3 , is expressed in neuronal and immune cells. Its impaired expression or function produces chronic inflammatory disease and autoimmune disorders, the severity of which correlates with Kv1.3 protein expression. The intersubunit recognition domain, T1, at the cytosolic N-terminus of Kv1.3, acquires secondary, tertiary, and quaternary structures during early biogenesis while the nascent protein is attached to the ribosome and/or the ER membrane. In this study, we ask whether native KCNA3 gene variants in T1 are associated with human disease and whether they manifest early-stage folding defects, energetic instabilities, and conformational distortion of subunits. We use three approaches: first, the unbiased "genome-first" approach to determine phenotype associations of specific KCNA3 rare variants. Second, we use biochemical assays to assess early-stage tertiary and quaternary folding and membrane association of these variants during early biogenesis. Third, we use all-atom molecular dynamics simulations of the T1 tetramer to assess structural macroscopic and energetic stability differences between wildtype (WT) Kv1.3 and a single-point variant, R114G. Measured folding probabilities and membrane associations are dramatically reduced in several of the native variants compared to WT. Simulations strikingly show that the R114G variant produces more energetically unstable and dynamic T1 domains, concomitant with tertiary unwinding and impaired formation of symmetrical tetramers. Our findings identify molecular mechanisms by which rare variants influence channel assembly, potentially contributing to diverse clinical phenotypes underlying human disease.
Collapse
|
5
|
Das D, Lamothe SM, Wong AA, Baronas VA, Kurata HT. Competitive modulation of K V1.2 gating by LMAN2 and Slc7a5. FASEB J 2024; 38:e70243. [PMID: 39659243 PMCID: PMC11632407 DOI: 10.1096/fj.202401737rr] [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: 07/28/2024] [Revised: 11/12/2024] [Accepted: 11/27/2024] [Indexed: 12/12/2024]
Abstract
KV1.2 is a prominent ion channel in the CNS, where it regulates neuronal excitability. KV1.2 structure and function are well understood, but there is less consensus on mechanisms of regulation of KV1.2 and other potassium channels by auxiliary proteins. We previously identified novel regulators of KV1.2 by a mass spectrometry approach. The neutral amino acid transporter Slc7a5 causes a dramatic hyperpolarizing shift of channel activation. In contrast, the transmembrane lectin LMAN2 is a recently identified candidate regulator that has the opposite effect on gating: large depolarizing voltages are required to activate KV1.2 channels co-expressed with LMAN2. In this study, we characterized the functional interaction between LMAN2 and Slc7a5 on KV1.2 gating properties and identified key structural elements that underlie sensitivity to each regulator. When LMAN2 and Slc7a5 are expressed together, KV1.2 activation exhibits a bi-modal voltage-dependence, suggesting two distinct populations of channels regulated either by LMAN2 or Slc7a5, but not both. Using a KV1.2:1.5 chimeric approach, we identified specific regions between the S1 to S3 segments of the voltage sensing domain (VSD) that are distinct for either Slc7a5 or LMAN2 sensitivity. By replacing either segment with sequence from KV1.5, modulation by the corresponding regulator was selectively abolished. These results suggest that Slc7a5 and LMAN2 compete for interaction with the KV1.2 voltage sensor, leading to complex voltage-dependence of channel activity when both regulators are present in the cell.
Collapse
Affiliation(s)
- Damayantee Das
- Department of Pharmacology, Alberta Diabetes InstituteUniversity of AlbertaEdmontonAlbertaCanada
| | - Shawn M. Lamothe
- Department of Pharmacology, Alberta Diabetes InstituteUniversity of AlbertaEdmontonAlbertaCanada
| | - Anson A. Wong
- Department of Pharmacology, Alberta Diabetes InstituteUniversity of AlbertaEdmontonAlbertaCanada
| | - Victoria A. Baronas
- Department of Pharmacology, Alberta Diabetes InstituteUniversity of AlbertaEdmontonAlbertaCanada
| | - Harley T. Kurata
- Department of Pharmacology, Alberta Diabetes InstituteUniversity of AlbertaEdmontonAlbertaCanada
| |
Collapse
|
6
|
Iwamuro T, Itohara K, Furukawa Y. Stability of N-type inactivation and the coupling between N-type and C-type inactivation in the Aplysia Kv1 channel. Pflugers Arch 2024; 476:1493-1516. [PMID: 39008084 PMCID: PMC11639194 DOI: 10.1007/s00424-024-02982-5] [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: 02/27/2024] [Revised: 05/28/2024] [Accepted: 06/19/2024] [Indexed: 07/16/2024]
Abstract
The voltage-dependent potassium channels (Kv channels) show several different types of inactivation. N-type inactivation is a fast inactivating mechanism, which is essentially an open pore blockade by the amino-terminal structure of the channel itself or the auxiliary subunit. There are several functionally discriminatable slow inactivation (C-type, P-type, U-type), the mechanism of which is supposed to include rearrangement of the pore region. In some Kv1 channels, the actual inactivation is brought about by coupling of N-type and C-type inactivation (N-C coupling). In the present study, we focused on the N-C coupling of the Aplysia Kv1 channel (AKv1). AKv1 shows a robust N-type inactivation, but its recovery is almost thoroughly from C-type inactivated state owing to the efficient N-C coupling. In the I8Q mutant of AKv1, we found that the inactivation as well as its recovery showed two kinetic components apparently correspond to N-type and C-type inactivation. Also, the cumulative inactivation which depends on N-type mechanism in AKv1 was hindered in I8Q, suggesting that N-type inactivation of I8Q is less stable. We also found that Zn2 + specifically accelerates C-type inactivation of AKv1 and that H382 in the pore turret is involved in the Zn2 + binding. Because the region around Ile8 (I8) in AKv1 has been suggested to be involved in the pre-block binding of the amino-terminal structure, our results strengthen a hypothesis that the stability of the pre-block state is important for stable N-type inactivation as well as the N-C coupling in the Kv1 channel inactivation.
Collapse
Affiliation(s)
- Tokunari Iwamuro
- Laboratory of Neurobiology, Graduate School of Integrated Sciences of Life, Hiroshima University, Kagamiyama 1-7-1, 739-8521, Higashi-Hiroshima, Japan
| | - Kazuki Itohara
- Laboratory of Neurobiology, Graduate School of Integrated Sciences of Life, Hiroshima University, Kagamiyama 1-7-1, 739-8521, Higashi-Hiroshima, Japan
| | - Yasuo Furukawa
- Laboratory of Neurobiology, Graduate School of Integrated Sciences of Life, Hiroshima University, Kagamiyama 1-7-1, 739-8521, Higashi-Hiroshima, Japan.
| |
Collapse
|
7
|
Cheng K, Sun Y, Yu H, Hu Y, He Y, Shen Y. Staphylococcus aureus SOS response: Activation, impact, and drug targets. MLIFE 2024; 3:343-366. [PMID: 39359682 PMCID: PMC11442139 DOI: 10.1002/mlf2.12137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/17/2024] [Accepted: 04/10/2024] [Indexed: 10/04/2024]
Abstract
Staphylococcus aureus is a common cause of diverse infections, ranging from superficial to invasive, affecting both humans and animals. The widespread use of antibiotics in clinical treatments has led to the emergence of antibiotic-resistant strains and small colony variants. This surge presents a significant challenge in eliminating infections and undermines the efficacy of available treatments. The bacterial Save Our Souls (SOS) response, triggered by genotoxic stressors, encompasses host immune defenses and antibiotics, playing a crucial role in bacterial survival, invasiveness, virulence, and drug resistance. Accumulating evidence underscores the pivotal role of the SOS response system in the pathogenicity of S. aureus. Inhibiting this system offers a promising approach for effective bactericidal treatments and curbing the evolution of antimicrobial resistance. Here, we provide a comprehensive review of the activation, impact, and key proteins associated with the SOS response in S. aureus. Additionally, perspectives on therapeutic strategies targeting the SOS response for S. aureus, both individually and in combination with traditional antibiotics are proposed.
Collapse
Affiliation(s)
- Kaiying Cheng
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of MedicineZhejiang UniversityHangzhouChina
| | - Yukang Sun
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
| | - Huan Yu
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
| | - Yingxuan Hu
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
| | - Yini He
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
| | - Yuanyuan Shen
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
| |
Collapse
|
8
|
Reis MC, Mandler L, Kang J, Oliver D, Halaszovich C, Nolte D. A novel KCND3 variant in the N-terminus impairs the ionic current of Kv4.3 and is associated with SCA19/22. J Cell Mol Med 2024; 28:e70039. [PMID: 39180521 PMCID: PMC11344468 DOI: 10.1111/jcmm.70039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/05/2024] [Indexed: 08/26/2024] Open
Abstract
Spinocerebellar ataxias (SCAs) are a genetically heterogeneous group of autosomal dominant movement disorders. Among the SCAs associated with impaired ion channel function, SCA19/22 is caused by pathogenic variants in KCND3, which encodes the voltage-gated potassium channel Kv4.3. SCA19/22 is clinically characterized by ataxia, dysarthria and oculomotor dysfunction in combination with other signs and symptoms, including mild cognitive impairment, peripheral neuropathy and pyramidal signs. The known KCND3 pathogenic variants are localized either in the transmembrane segments, the connecting loops, or the C-terminal region of Kv4.3. We have identified a novel pathogenic variant, c.455A>G (p.D152G), localized in the N-terminus of Kv4.3. It is located in the immediate neighbourhood of the T1 domain, which is responsible for multimerization with the β-subunit KChIP2b and thus for the formation of functional heterooctamers. Electrophysiological studies showed that p.D152G does not affect channel gating, but reduces the ionic current in Kv4.3, even though the variant is not located in the transmembrane domains. Impaired channel trafficking to the plasma membrane may contribute to this effect. In a patient with a clinical picture corresponding to SCA19/22, p.D152G is the first pathogenic variant in the N-terminus of Kv4.3 to be described to date with an effect on ion channel activity.
Collapse
Affiliation(s)
| | - Laura Mandler
- Institute of Human GeneticsJustus‐Liebig‐University GiessenGiessenGermany
- Present address:
Department of NeurologyJustus‐Liebig‐University GiessenGiessenGermany
| | - Jun‐Suk Kang
- Department of NeurologyGoethe‐University FrankfurtFrankfurtGermany
- Present address:
NeuropraxisFrankfurtGermany
| | - Dominik Oliver
- Institute of PhysiologyPhilipps‐University MarburgMarburgGermany
| | | | - Dagmar Nolte
- Institute of Human GeneticsJustus‐Liebig‐University GiessenGiessenGermany
| |
Collapse
|
9
|
Raph SM, Calderin EP, Nong Y, Brittian K, Garrett L, Zhang D, Nystoriak MA. Kv beta complex facilitates exercise-induced augmentation of myocardial perfusion and cardiac growth. Front Cardiovasc Med 2024; 11:1411354. [PMID: 38978788 PMCID: PMC11228310 DOI: 10.3389/fcvm.2024.1411354] [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: 04/02/2024] [Accepted: 05/21/2024] [Indexed: 07/10/2024] Open
Abstract
The oxygen sensitivity of voltage-gated potassium (Kv) channels regulates cardiovascular physiology. Members of the Kv1 family interact with intracellular Kvβ proteins, which exhibit aldo-keto reductase (AKR) activity and confer redox sensitivity to Kv channel gating. The Kvβ proteins contribute to vasoregulation by controlling outward K+ currents in smooth muscle upon changes in tissue oxygen consumption and demand. Considering exercise as a primary physiological stimulus of heightened oxygen demand, the current study tested the role of Kvβ proteins in exercise performance, exercise-induced adaptations in myocardial perfusion, and physiological cardiac growth. Our findings reveal that genetic ablation of Kvβ2 proteins diminishes baseline exercise capacity in mice and attenuates the enhancement in exercise performance observed after long-term training. Moreover, we demonstrate that Kvβ2 proteins are critical for exercise-mediated enhancement in myocardial perfusion during cardiac stress as well as adaptive changes in cardiac structure. Our results underscore the importance of Kvβ proteins in metabolic vasoregulation, highlighting their role in modulating both exercise capacity and cardiovascular benefits associated with training. Furthermore, our study sheds light on a novel molecular target for enhancing exercise performance and improving the health benefits associated with exercise training in patients with limited capacity for physical activity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Matthew A. Nystoriak
- Center for Cardiometabolic Science, Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY, United States
| |
Collapse
|
10
|
Clatot J, Currin CB, Liang Q, Pipatpolkai T, Massey SL, Helbig I, Delemotte L, Vogels TP, Covarrubias M, Goldberg EM. A structurally precise mechanism links an epilepsy-associated KCNC2 potassium channel mutation to interneuron dysfunction. Proc Natl Acad Sci U S A 2024; 121:e2307776121. [PMID: 38194456 PMCID: PMC10801864 DOI: 10.1073/pnas.2307776121] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 11/17/2023] [Indexed: 01/11/2024] Open
Abstract
De novo heterozygous variants in KCNC2 encoding the voltage-gated potassium (K+) channel subunit Kv3.2 are a recently described cause of developmental and epileptic encephalopathy (DEE). A de novo variant in KCNC2 c.374G > A (p.Cys125Tyr) was identified via exome sequencing in a patient with DEE. Relative to wild-type Kv3.2, Kv3.2-p.Cys125Tyr induces K+ currents exhibiting a large hyperpolarizing shift in the voltage dependence of activation, accelerated activation, and delayed deactivation consistent with a relative stabilization of the open conformation, along with increased current density. Leveraging the cryogenic electron microscopy (cryo-EM) structure of Kv3.1, molecular dynamic simulations suggest that a strong π-π stacking interaction between the variant Tyr125 and Tyr156 in the α-6 helix of the T1 domain promotes a relative stabilization of the open conformation of the channel, which underlies the observed gain of function. A multicompartment computational model of a Kv3-expressing parvalbumin-positive cerebral cortex fast-spiking γ-aminobutyric acidergic (GABAergic) interneuron (PV-IN) demonstrates how the Kv3.2-Cys125Tyr variant impairs neuronal excitability and dysregulates inhibition in cerebral cortex circuits to explain the resulting epilepsy.
Collapse
Affiliation(s)
- Jerome Clatot
- Division of Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- The Epilepsy Neurogenetics Initiative, The Children’s Hospital of Philadelphia, Philadelphia,PA19104
| | | | - Qiansheng Liang
- Department of Neuroscience and Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA19107
| | - Tanadet Pipatpolkai
- Department of Applied Physics, Science for Life Laboratory, Royal Institute of Technology, SolnaSE-171 21, Sweden
| | - Shavonne L. Massey
- Division of Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- The Epilepsy Neurogenetics Initiative, The Children’s Hospital of Philadelphia, Philadelphia,PA19104
- The Department of Neurology, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
| | - Ingo Helbig
- Division of Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- The Epilepsy Neurogenetics Initiative, The Children’s Hospital of Philadelphia, Philadelphia,PA19104
- The Department of Neurology, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Department of Biomedical and Health Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Lucie Delemotte
- Department of Applied Physics, Science for Life Laboratory, Royal Institute of Technology, SolnaSE-171 21, Sweden
| | - Tim P. Vogels
- The Institute of Science and Technology Austria, Klosterneuburg3400, Austria
| | - Manuel Covarrubias
- Department of Neuroscience and Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA19107
| | - Ethan M. Goldberg
- Division of Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- The Epilepsy Neurogenetics Initiative, The Children’s Hospital of Philadelphia, Philadelphia,PA19104
- The Department of Neurology, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- The Department of Neuroscience, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
| |
Collapse
|
11
|
Zhang L, Zhou R, Fu X, Zhang G, Zhang L, Zhou SF, Jiang W. Specific coenzyme preference switching for an aldo-keto reductase that synthesizes the chiral intermediate of duloxetine. Enzyme Microb Technol 2023; 171:110326. [PMID: 37717530 DOI: 10.1016/j.enzmictec.2023.110326] [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: 07/02/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/19/2023]
Abstract
The synthesis of chiral intermediates for the traditional antidepressant duloxetine has gained significant attention as the number of depressed patients continues to grow. S-N, N-Dimethyl-3-hydroxy-3-(2-thienyl)-1-propanamide (S-DHTP) is a critical intermediate in the synthesis of duloxetine, and the chemical synthesis process is complex and environmentally unfriendly. Reduced nicotinamide adenine dinucleotide phosphate (NADPH) is a major cost driver in the biocatalytic production of S-DHTP from N, N-Dimethyl-3-keto-3-(2-thienyl)-1-propanamide (DKTP). Here, we successfully modified the coenzyme preference of an aldo-keto reductase (AKR7-2-1) to use the cheaper reduced nicotinamide adenine dinucleotide (NADH) through a coenzyme preference modification approach. We utilized protein engineering to create a superior mutant, Y53F, which increased the coenzyme specificity of AKR7-2-1 by 875-fold and improved its thermal stability, enhancing its potential for industrial applications. Molecular dynamics simulations were performed to demonstrate the effect of mutations at key sites on the protein, revealing the altered coenzyme preference and increased thermal stability from structural and energetic changes. This study validates the viability of the coenzyme preference modification strategy for aldo-keto reductase, offering valuable insights for fellow researchers and guiding future investigations.
Collapse
Affiliation(s)
- Lingzhi Zhang
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China
| | - Rui Zhou
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China
| | - Xiaoli Fu
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China
| | - Guangya Zhang
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China
| | - Lijuan Zhang
- State Key Laboratory of Microbial Technology, Shandong University, No. 72 Binhai Road, Qingdao 266237, PR China
| | - Shu-Feng Zhou
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China.
| | - Wei Jiang
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China.
| |
Collapse
|
12
|
Raph SM, Dwenger MM, Hu X, Nystoriak MA. Basal NAD(H) redox state permits hydrogen peroxide-induced mesenteric artery dilatation. J Physiol 2023; 601:2621-2634. [PMID: 37114864 PMCID: PMC11714382 DOI: 10.1113/jp284195] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 04/26/2023] [Indexed: 04/29/2023] Open
Abstract
Smooth muscle voltage-gated K+ (Kv) channels in resistance arteries control vascular tone and contribute to the coupling of blood flow with local metabolic activity. Members of the Kv1 family are expressed in vascular smooth muscle and are modulated upon physiological elevation of local metabolites, including the glycolytic end-product l-lactate and superoxide-derived hydrogen peroxide (H2 O2 ). Here, we show that l-lactate elicits vasodilatation of small-diameter mesenteric arteries in a mechanism that requires lactate dehydrogenase (LDH). Using the inside-out configuration of the patch clamp technique, we show that increases in NADH that reflect LDH-mediated conversion of l-lactate to pyruvate directly stimulate the activity of single Kv1 channels and significantly enhance the sensitivity of Kv1 activity to H2 O2 . Consistent with these findings, H2 O2 -evoked vasodilatation was significantly greater in the presence of 10 mM l-lactate relative to lactate-free conditions, yet was abolished in the presence of 10 mM pyruvate, which shifts the LDH reaction towards the generation of NAD+ . Moreover, the enhancement of H2 O2 -induced vasodilatation was abolished in arteries from double transgenic mice with selective overexpression of the intracellular Kvβ1.1 subunit in smooth muscle cells. Together, our results indicate that the Kvβ complex of native vascular Kv1 channels serves as a nodal effector for multiple redox signals to precisely control channel activity and vascular tone in the face of dynamic tissue-derived metabolic cues. KEY POINTS: Vasodilatation of mesenteric arteries by elevated external l-lactate requires its conversion by lactate dehydrogenase. Application of either NADH or H2 O2 potentiates single Kv channel currents in excised membrane patches from mesenteric artery smooth muscle cells. The binding of NADH enhances the stimulatory effects of H2 O2 on single Kv channel activity. The vasodilatory response to H2 O2 is differentially modified upon elevation of external l-lactate or pyruvate. The presence of l-lactate enhances the vasodilatory response to H2 O2 via the Kvβ subunit complex in smooth muscle.
Collapse
Affiliation(s)
- Sean M. Raph
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202
| | - Marc M. Dwenger
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202
| | - Xuemei Hu
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202
| | - Matthew A. Nystoriak
- Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| |
Collapse
|
13
|
Abbott GW. Kv Channel Ancillary Subunits: Where Do We Go from Here? Physiology (Bethesda) 2022; 37:0. [PMID: 35797055 PMCID: PMC9394777 DOI: 10.1152/physiol.00005.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/29/2022] [Accepted: 04/29/2022] [Indexed: 01/10/2023] Open
Abstract
Voltage-gated potassium (Kv) channels each comprise four pore-forming α-subunits that orchestrate essential duties such as voltage sensing and K+ selectivity and conductance. In vivo, however, Kv channels also incorporate regulatory subunits-some Kv channel specific, others more general modifiers of protein folding, trafficking, and function. Understanding all the above is essential for a complete picture of the role of Kv channels in physiology and disease.
Collapse
Affiliation(s)
- Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California
| |
Collapse
|
14
|
Diversification of Potassium Currents in Excitable Cells via Kvβ Proteins. Cells 2022; 11:cells11142230. [PMID: 35883673 PMCID: PMC9317154 DOI: 10.3390/cells11142230] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 12/10/2022] Open
Abstract
Excitable cells of the nervous and cardiovascular systems depend on an assortment of plasmalemmal potassium channels to control diverse cellular functions. Voltage-gated potassium (Kv) channels are central to the feedback control of membrane excitability in these processes due to their activation by depolarized membrane potentials permitting K+ efflux. Accordingly, Kv currents are differentially controlled not only by numerous cellular signaling paradigms that influence channel abundance and shape voltage sensitivity, but also by heteromeric configurations of channel complexes. In this context, we discuss the current knowledge related to how intracellular Kvβ proteins interacting with pore complexes of Shaker-related Kv1 channels may establish a modifiable link between excitability and metabolic state. Past studies in heterologous systems have indicated roles for Kvβ proteins in regulating channel stability, trafficking, subcellular targeting, and gating. More recent works identifying potential in vivo physiologic roles are considered in light of these earlier studies and key gaps in knowledge to be addressed by future research are described.
Collapse
|
15
|
Pyridine nucleotide redox potential in coronary smooth muscle couples myocardial blood flow to cardiac metabolism. Nat Commun 2022; 13:2051. [PMID: 35440632 PMCID: PMC9018695 DOI: 10.1038/s41467-022-29745-z] [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: 05/25/2021] [Accepted: 03/28/2022] [Indexed: 12/13/2022] Open
Abstract
Adequate oxygen delivery to the heart during stress is essential for sustaining cardiac function. Acute increases in myocardial oxygen demand evoke coronary vasodilation and enhance perfusion via functional upregulation of smooth muscle voltage-gated K+ (Kv) channels. Because this response is controlled by Kv1 accessory subunits (i.e., Kvβ), which are NAD(P)(H)-dependent aldo-keto reductases, we tested the hypothesis that oxygen demand modifies arterial [NAD(H)]i, and that resultant cytosolic pyridine nucleotide redox state influences Kv1 activity. High-resolution imaging mass spectrometry and live-cell imaging reveal cardiac workload-dependent increases in NADH:NAD+ in intramyocardial arterial myocytes. Intracellular NAD(P)(H) redox ratios reflecting elevated oxygen demand potentiate native coronary Kv1 activity in a Kvβ2-dependent manner. Ablation of Kvβ2 catalysis suppresses redox-dependent increases in Kv1 activity, vasodilation, and the relationship between cardiac workload and myocardial blood flow. Collectively, this work suggests that the pyridine nucleotide sensitivity and enzymatic activity of Kvβ2 controls coronary vasoreactivity and myocardial blood flow during metabolic stress. Physiological matching of blood flow to the demand for oxygen by the heart is required for sustained cardiac health, yet the underlying mechanisms are obscure. Here, the authors report a key role for acute modifications to the redox state of intracellular pyridine nucleotides in coronary smooth muscle and their impact on voltage-gated K + channels in metabolic vasodilation
Collapse
|
16
|
Ionic channels in nerve membranes, 50 years on. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 169-170:12-20. [PMID: 34856230 PMCID: PMC8977236 DOI: 10.1016/j.pbiomolbio.2021.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/25/2021] [Indexed: 12/19/2022]
Abstract
This retrospective traces the hypothesis of ion channels from an early statement in a 1970 essay in this journal (Hille, B., 1970, Prog. Biophys. Mol. Biol. 21, 1-32) to its realization today in biophysical, molecular, biochemical, and structural terms. The Na+ and K+ channels of the action potential have been isolated, reconstituted, cloned, mutated, and expressed. They are conformationally flexible, multi-pass glycosylated membrane proteins. Refined atomic structures of several conformational states are known. The discoveries over this half century history illustrate the growth of a field from initial ideas to a mature discipline of biology, physiology, and biomedical science.
Collapse
|
17
|
Lamothe SM, Kurata HT. Slc7a5 alters Kvβ-mediated regulation of Kv1.2. J Gen Physiol 2021; 152:151687. [PMID: 32311044 PMCID: PMC7335012 DOI: 10.1085/jgp.201912524] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/22/2020] [Accepted: 03/13/2020] [Indexed: 01/15/2023] Open
Abstract
The voltage-gated potassium channel Kv1.2 plays a pivotal role in neuronal excitability and is regulated by a variety of known and unknown extrinsic factors. The canonical accessory subunit of Kv1.2, Kvβ, promotes N-type inactivation and cell surface expression of the channel. We recently reported that a neutral amino acid transporter, Slc7a5, alters the function and expression of Kv1.2. In the current study, we investigated the effects of Slc7a5 on Kv1.2 in the presence of Kvβ1.2 subunits. We observed that Slc7a5-induced suppression of Kv1.2 current and protein expression was attenuated with cotransfection of Kvβ1.2. However, gating effects mediated by Slc7a5, including disinhibition and a hyperpolarizing shift in channel activation, were observed together with Kvβ-mediated inactivation, indicating convergent regulation of Kv1.2 by both regulatory proteins. Slc7a5 influenced several properties of Kvβ-induced inactivation of Kv1.2, including accelerated inactivation, a hyperpolarizing shift and greater extent of steady-state inactivation, and delayed recovery from inactivation. These modified inactivation properties were also apparent in altered deactivation of the Kv1.2/Kvβ/Slc7a5 channel complex. Taken together, these findings illustrate a functional interaction arising from simultaneous regulation of Kv1.2 by Kvβ and Slc7a5, leading to powerful effects on Kv1.2 expression, gating, and overall channel function.
Collapse
Affiliation(s)
- Shawn M Lamothe
- Department of Pharmacology, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Harley T Kurata
- Department of Pharmacology, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
18
|
Control of Biophysical and Pharmacological Properties of Potassium Channels by Ancillary Subunits. Handb Exp Pharmacol 2021; 267:445-480. [PMID: 34247280 DOI: 10.1007/164_2021_512] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Potassium channels facilitate and regulate physiological processes as diverse as electrical signaling, ion, solute and hormone secretion, fluid homeostasis, hearing, pain sensation, muscular contraction, and the heartbeat. Potassium channels are each formed by either a tetramer or dimer of pore-forming α subunits that co-assemble to create a multimer with a K+-selective pore that in most cases is capable of functioning as a discrete unit to pass K+ ions across the cell membrane. The reality in vivo, however, is that the potassium channel α subunit multimers co-assemble with ancillary subunits to serve specific physiological functions. The ancillary subunits impart specific physiological properties that are often required for a particular activity in vivo; in addition, ancillary subunit interaction often alters the pharmacology of the resultant complex. In this chapter the modes of action of ancillary subunits on K+ channel physiology and pharmacology are described and categorized into various mechanistic classes.
Collapse
|
19
|
Chen X, Zhang Y, Wang Q, Qin Y, Yang X, Xing Z, Shen Y, Wu H, Qi Y. The function of SUMOylation and its crucial roles in the development of neurological diseases. FASEB J 2021; 35:e21510. [PMID: 33710677 DOI: 10.1096/fj.202002702r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/02/2021] [Accepted: 02/22/2021] [Indexed: 11/11/2022]
Abstract
Neurological diseases are relatively complex diseases of a large system; however, the detailed mechanism of their pathogenesis has not been completely elucidated, and effective treatment methods are still lacking for some of the diseases. The SUMO (small ubiquitin-like modifier) modification is a dynamic and reversible process that is catalyzed by SUMO-specific E1, E2, and E3 ligases and reversed by a family of SENPs (SUMO/Sentrin-specific proteases). SUMOylation covalently conjugates numerous cellular proteins, and affects their cellular localization and biological activity in numerous cellular processes. A wide range of neuronal proteins have been identified as SUMO substrates, and the disruption of SUMOylation results in defects in synaptic plasticity, neuronal excitability, and neuronal stress responses. SUMOylation disorders cause many neurodegenerative diseases, such as Parkinson's disease, Alzheimer's disease, and Huntington's disease. By modulating the ion channel subunit, SUMOylation imbalance is responsible for the development of various channelopathies. The regulation of protein SUMOylation in neurons may provide a new strategy for the development of targeted therapeutic drugs for neurodegenerative diseases and channelopathies.
Collapse
Affiliation(s)
- Xu Chen
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yuhong Zhang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Qiqi Wang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yuanyuan Qin
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xinyi Yang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Zhengcao Xing
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yajie Shen
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Hongmei Wu
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yitao Qi
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
20
|
Lamothe SM, Sharmin N, Silver G, Satou M, Hao Y, Tateno T, Baronas VA, Kurata HT. Control of Slc7a5 sensitivity by the voltage-sensing domain of Kv1 channels. eLife 2020; 9:54916. [PMID: 33164746 PMCID: PMC7690953 DOI: 10.7554/elife.54916] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 11/06/2020] [Indexed: 01/13/2023] Open
Abstract
Many voltage-dependent ion channels are regulated by accessory proteins. We recently reported powerful regulation of Kv1.2 potassium channels by the amino acid transporter Slc7a5. In this study, we report that Kv1.1 channels are also regulated by Slc7a5, albeit with different functional outcomes. In heterologous expression systems, Kv1.1 exhibits prominent current enhancement ('disinhibition') with holding potentials more negative than −120 mV. Knockdown of endogenous Slc7a5 leads to larger Kv1.1 currents and strongly attenuates the disinhibition effect, suggesting that Slc7a5 regulation of Kv1.1 involves channel inhibition that can be reversed by supraphysiological hyperpolarizing voltages. We investigated chimeric combinations of Kv1.1 and Kv1.2, demonstrating that exchange of the voltage-sensing domain controls the sensitivity and response to Slc7a5, and localize a specific position in S1 with prominent effects on Slc7a5 sensitivity. Overall, our study highlights multiple Slc7a5-sensitive Kv1 subunits, and identifies the voltage-sensing domain as a determinant of Slc7a5 modulation of Kv1 channels.
Collapse
Affiliation(s)
- Shawn M Lamothe
- Department of Pharmacology, Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Nazlee Sharmin
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, School of Dentistry, Edmonton Clinic Health Academy (ECHA), Edmonton, Canada
| | - Grace Silver
- Department of Pharmacology, Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Motoyasu Satou
- Department of Biochemistry, Dokkyo Medical University School of Medicine, Tochigi, Japan.,Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Yubin Hao
- Department of Pharmacology, Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Toru Tateno
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Victoria A Baronas
- Department of Pharmacology, Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Harley T Kurata
- Department of Pharmacology, Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
21
|
Tian Y, Korn P, Tripathi P, Komnig D, Wiemuth D, Nikouee A, Classen A, Bolm C, Falkenburger BH, Lüscher B, Gründer S. The mono-ADP-ribosyltransferase ARTD10 regulates the voltage-gated K + channel Kv1.1 through protein kinase C delta. BMC Biol 2020; 18:143. [PMID: 33059680 PMCID: PMC7558731 DOI: 10.1186/s12915-020-00878-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 09/24/2020] [Indexed: 11/30/2022] Open
Abstract
Background ADP-ribosylation is a ubiquitous post-translational modification that involves both mono- and poly-ADP-ribosylation. ARTD10, also known as PARP10, mediates mono-ADP-ribosylation (MARylation) of substrate proteins. A previous screen identified protein kinase C delta (PKCδ) as a potential ARTD10 substrate, among several other kinases. The voltage-gated K+ channel Kv1.1 constitutes one of the dominant Kv channels in neurons of the central nervous system and the inactivation properties of Kv1.1 are modulated by PKC. In this study, we addressed the role of ARTD10-PKCδ as a regulator of Kv1.1. Results We found that ARTD10 inhibited PKCδ, which increased Kv1.1 current amplitude and the proportion of the inactivating current component in HeLa cells, indicating that ARTD10 regulates Kv1.1 in living cells. An inhibitor of ARTD10, OUL35, significantly decreased peak amplitude together with the proportion of the inactivating current component of Kv1.1-containing channels in primary hippocampal neurons, demonstrating that the ARTD10-PKCδ signaling cascade regulates native Kv1.1. Moreover, we show that the pharmacological blockade of ARTD10 increases excitability of hippocampal neurons. Conclusions Our results, for the first time, suggest that MARylation by ARTD10 controls neuronal excitability.
Collapse
Affiliation(s)
- Yuemin Tian
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Patricia Korn
- Institute of Biochemistry and Molecular Biology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Priyanka Tripathi
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.,Present address: Institute of Neuropathology, RWTH Aachen University Medical School, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Daniel Komnig
- Department of Neurology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.,JARA-Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich, Jülich, Germany
| | - Dominik Wiemuth
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Azadeh Nikouee
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Arno Classen
- Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52056, Aachen, Germany
| | - Carsten Bolm
- Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52056, Aachen, Germany
| | - Björn H Falkenburger
- Department of Neurology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.,JARA-Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich, Jülich, Germany.,Present address: Department of Neurology, Dresden University Medical Center, Fetscherstraße 74, 01307, Dresden, Germany
| | - Bernhard Lüscher
- Institute of Biochemistry and Molecular Biology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Stefan Gründer
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| |
Collapse
|
22
|
Accili E. An ion channel in the company of a transporter. J Gen Physiol 2020; 152:151884. [PMID: 32579683 PMCID: PMC7335010 DOI: 10.1085/jgp.202012590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Eric Accili
- Department of Cellular and Physiological Sciences, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
23
|
Wheeler R, Yu X, Hou C, Mitra P, Chen JM, Herkules F, Ivanov DN, Tsodikov OV, Rohr J. Discovery of a Cryptic Intermediate in Late Steps of Mithramycin Biosynthesis. Angew Chem Int Ed Engl 2020; 59:826-832. [PMID: 31702856 PMCID: PMC6940538 DOI: 10.1002/anie.201910241] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 10/14/2019] [Indexed: 12/23/2022]
Abstract
MtmOIV and MtmW catalyze the final two reactions in the mithramycin (MTM) biosynthetic pathway, the Baeyer-Villiger opening of the fourth ring of premithramycin B (PMB), creating the C3 pentyl side chain, strictly followed by reduction of the distal keto group on the new side chain. Unexpectedly this results in a C2 stereoisomer of mithramycin, iso-mithramycin (iso-MTM). Iso-MTM undergoes a non-enzymatic isomerization to MTM catalyzed by Mg2+ ions. Crystal structures of MtmW and its complexes with co-substrate NADPH and PEG, suggest a catalytic mechanism of MtmW. The structures also show that a tetrameric assembly of this enzyme strikingly resembles the ring-shaped β subunit of a vertebrate ion channel. We show that MtmW and MtmOIV form a complex in the presence of PMB and NADPH, presumably to hand over the unstable MtmOIV product to MtmW, yielding iso-MTM, as a potential self-resistance mechanism against MTM toxicity.
Collapse
Affiliation(s)
- Ryan Wheeler
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536-0596, USA
| | - Xia Yu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536-0596, USA
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Caixia Hou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536-0596, USA
| | - Prithiba Mitra
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536-0596, USA
| | - Jhong-Min Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536-0596, USA
| | - Frank Herkules
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Dmitri N Ivanov
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Oleg V Tsodikov
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536-0596, USA
| | - Jürgen Rohr
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536-0596, USA
| |
Collapse
|
24
|
Wheeler R, Yu X, Hou C, Mitra P, Chen J, Herkules F, Ivanov DN, Tsodikov OV, Rohr J. Discovery of a Cryptic Intermediate in Late Steps of Mithramycin Biosynthesis. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201910241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Ryan Wheeler
- Department of Pharmaceutical SciencesCollege of PharmacyUniversity of Kentucky 789 South Limestone Street Lexington KY 40536-0596 USA
| | - Xia Yu
- Department of Pharmaceutical SciencesCollege of PharmacyUniversity of Kentucky 789 South Limestone Street Lexington KY 40536-0596 USA
- Xiangya School of Pharmaceutical SciencesCentral South University Changsha Hunan 410013 P. R. China
| | - Caixia Hou
- Department of Pharmaceutical SciencesCollege of PharmacyUniversity of Kentucky 789 South Limestone Street Lexington KY 40536-0596 USA
| | - Prithiba Mitra
- Department of Pharmaceutical SciencesCollege of PharmacyUniversity of Kentucky 789 South Limestone Street Lexington KY 40536-0596 USA
| | - Jhong‐Min Chen
- Department of Pharmaceutical SciencesCollege of PharmacyUniversity of Kentucky 789 South Limestone Street Lexington KY 40536-0596 USA
| | - Frank Herkules
- Department of BiochemistryUniversity of Texas Health Science Center San Antonio TX 78229 USA
| | - Dmitri N. Ivanov
- Department of BiochemistryUniversity of Texas Health Science Center San Antonio TX 78229 USA
| | - Oleg V. Tsodikov
- Department of Pharmaceutical SciencesCollege of PharmacyUniversity of Kentucky 789 South Limestone Street Lexington KY 40536-0596 USA
| | - Jürgen Rohr
- Department of Pharmaceutical SciencesCollege of PharmacyUniversity of Kentucky 789 South Limestone Street Lexington KY 40536-0596 USA
| |
Collapse
|
25
|
Clusin WT, Wu TH, Shi LF, Kao PN. Further studies of ion channels in the electroreceptor of the skate through deep sequencing, cloning and cross species comparisons. Gene 2019; 718:143989. [PMID: 31326551 DOI: 10.1016/j.gene.2019.143989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 06/23/2019] [Accepted: 07/17/2019] [Indexed: 11/25/2022]
Abstract
Our comparative studies seek to understand the structure and function of ion channels in cartilaginous fish that can detect very low voltage gradients in seawater. The principal channels of the electroreceptor include a calcium activated K channel whose α subunit is Kcnma1, and a voltage-dependent calcium channel, Cacna1d. It has also been suggested based on physiological and pharmacological evidence that a voltage-gated K channel is present in the basal membranes of the receptor cells which modulates synaptic transmitter release. Large conductance calcium-activated K channels (BK) are comprised of four α subunits, encoded by Kcnma1 and modulatory β subunits of the Kcnmb class. We recently cloned and published the skate Kcnma1 gene and most of Kcnmb4 using purified mRNA of homogenized electroreceptors. Bellono et al. have recently performed RNA sequencing (RNA-seq) on purified mRNA from skate electroreceptors and found several ion channels including Kcnma1. We searched the Bellono et al. RNA-seq repository for additional channels and subunits. Our most significant findings are the presence of two Shaker type voltage dependent K channel sequences which are grouped together as isoforms in the data repository. The larger of these is a skate ortholog of the voltage dependent fast potassium channel Kv1.1, which is expressed at appreciable levels. The second ortholog is similar to Kv1.5 but has fewer N-terminal amino acids than other species. The sequence for Kv1.5 in the skate is very strongly aligned with the recently reported sequence for potassium channels in the electroreceptors of the cat shark, S. retifer, which also modulate synaptic transmission. The latter channel was designated as Kv1.3 in the initial report, but we suggest that these channels are actually orthologs of each other, and that Kv1.5 is the prevailing designation. We also found a beta subunit sequence (Kcnab2) which may co-assemble with one or both of the voltage gated channels. The new channels and subunits were verified by RT-PCR and the Kv1.1 sequence was confirmed by cloning. We also searched the RNA-seq repository for accessory subunits of Kcnma1, and found a computer-generated assembly that contained a complete sequence of its β subunit, Kcnmb2. Skate Kcnmb2 has a total of 279 amino acids, with 51 novel amino acids at the N-terminus which may play a specific physiological role. This sequence was confirmed by PCR and cloning. However, skate Kcnmb2 is expressed at low levels in the electroreceptor compared to Kcnma1 and skate Kcnmb1 is absent. The evolutionary origin of the newly described K channels and their subunits was studied by alignments with mammalian sequences, including human, and also those in related fish: the whale shark (R. typus), the ghost shark (C.milii), and (S. retifer). There are also orthologous K channels of the lamprey, which has electroreceptors. Tree building and bootstrap programs were used to confirm phylogenetic inferences. Further research should focus on the subcellular locations of these channels, their gating behavior, and the effects of accessory subunits on gating.
Collapse
Affiliation(s)
- William T Clusin
- Department of Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, United States of America.
| | - Ting-Hsuan Wu
- Department of Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, United States of America
| | - Ling-Fang Shi
- Department of Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, United States of America
| | - Peter N Kao
- Department of Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, United States of America
| |
Collapse
|
26
|
Linsdell P. Cystic fibrosis transmembrane conductance regulator (CFTR): Making an ion channel out of an active transporter structure. Channels (Austin) 2019; 12:284-290. [PMID: 30152709 PMCID: PMC6986785 DOI: 10.1080/19336950.2018.1502585] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cystic fibrosis is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR). CFTR is a member of the ATP-binding cassette (ABC) family of membrane transport proteins, most members of which function as ATP-dependent pumps. CFTR is unique among human ABC proteins in functioning not as a pump, but as an ion channel. Recent structural data has indicated that CFTR shares broadly similar overall architecture and ATP-dependent conformational changes as other ABC proteins. Functional investigations suggest that CFTR has a unique open portal connecting the cytoplasm to the transmembrane channel pore, that allows for a continuous pathway for Cl− ions to cross the membrane in one conformation. This lateral portal may be what allows CFTR to function as an ion channel rather than as a pump, suggesting a plausible mechanism by which channel function may have evolved in CFTR.
Collapse
Affiliation(s)
- Paul Linsdell
- a Department of Physiology & Biophysics , Dalhousie University , Halifax , Canada
| |
Collapse
|
27
|
Abraham MJ, Fleming KL, Raymond S, Wong AYC, Bergeron R. The sigma-1 receptor behaves as an atypical auxiliary subunit to modulate the functional characteristics of Kv1.2 channels expressed in HEK293 cells. Physiol Rep 2019; 7:e14147. [PMID: 31222975 PMCID: PMC6586770 DOI: 10.14814/phy2.14147] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/17/2019] [Accepted: 05/25/2019] [Indexed: 12/11/2022] Open
Abstract
Expression of Kv1.2 within Kv1.x potassium channel complexes is critical in maintaining appropriate neuronal excitability and determining the threshold for action potential firing. This is attributed to the interaction of Kv1.2 with a hitherto unidentified protein that confers bimodal channel activation gating, allowing neurons to adapt to repetitive trains of stimulation and protecting against hyperexcitability. One potential protein candidate is the sigma-1 receptor (Sig-1R), which regulates other members of the Kv1.x channel family; however, the biophysical nature of the interaction between Sig-1R and Kv1.2 has not been elucidated. We hypothesized that Sig-1R may regulate Kv1.2 and may further act as the unidentified modulator of Kv1.2 activation. In transiently transfected HEK293 cells, we found that ligand activation of the Sig-1R modulates Kv1.2 current amplitude. More importantly, Sig-1R interacts with Kv1.2 in baseline conditions to influence bimodal activation gating. These effects are abolished in the presence of the auxiliary subunit Kvβ2 and when the Sig-1R mutation underlying ALS16 (Sig-1R-E102Q), is expressed. These data suggest that Kvβ2 occludes the interaction of Sig-1R with Kv1.2, and that E102 may be a residue critical for Sig-1R modulation of Kv1.2. The results of this investigation describe an important new role for Sig-1R in the regulation of neuronal excitability and introduce a novel mechanism of pathophysiology in Sig-1R dysfunction.
Collapse
Affiliation(s)
- Madelyn J. Abraham
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
| | - Kayla L. Fleming
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
| | - Sophie Raymond
- NeuroscienceOttawa Hospital Research InstituteOttawaOntarioCanada
| | | | - Richard Bergeron
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
- NeuroscienceOttawa Hospital Research InstituteOttawaOntarioCanada
| |
Collapse
|
28
|
Haworth AS, Brackenbury WJ. Emerging roles for multifunctional ion channel auxiliary subunits in cancer. Cell Calcium 2019; 80:125-140. [PMID: 31071485 PMCID: PMC6553682 DOI: 10.1016/j.ceca.2019.04.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/16/2019] [Accepted: 04/16/2019] [Indexed: 02/07/2023]
Abstract
Several superfamilies of plasma membrane channels which regulate transmembrane ion flux have also been shown to regulate a multitude of cellular processes, including proliferation and migration. Ion channels are typically multimeric complexes consisting of conducting subunits and auxiliary, non-conducting subunits. Auxiliary subunits modulate the function of conducting subunits and have putative non-conducting roles, further expanding the repertoire of cellular processes governed by ion channel complexes to processes such as transcellular adhesion and gene transcription. Given this expansive influence of ion channels on cellular behaviour it is perhaps no surprise that aberrant ion channel expression is a common occurrence in cancer. This review will focus on the conducting and non-conducting roles of the auxiliary subunits of various Ca2+, K+, Na+ and Cl- channels and the burgeoning evidence linking such auxiliary subunits to cancer. Several subunits are upregulated (e.g. Cavβ, Cavγ) and downregulated (e.g. Kvβ) in cancer, while other subunits have been functionally implicated as oncogenes (e.g. Navβ1, Cavα2δ1) and tumour suppressor genes (e.g. CLCA2, KCNE2, BKγ1) based on in vivo studies. The strengthening link between ion channel auxiliary subunits and cancer has exposed these subunits as potential biomarkers and therapeutic targets. However further mechanistic understanding is required into how these subunits contribute to tumour progression before their therapeutic potential can be fully realised.
Collapse
Affiliation(s)
- Alexander S Haworth
- Department of Biology, University of York, Heslington, York, YO10 5DD, UK; York Biomedical Research Institute, University of York, Heslington, York, YO10 5DD, UK
| | - William J Brackenbury
- Department of Biology, University of York, Heslington, York, YO10 5DD, UK; York Biomedical Research Institute, University of York, Heslington, York, YO10 5DD, UK.
| |
Collapse
|
29
|
Carrillo-Reid L, Day M, Xie Z, Melendez AE, Kondapalli J, Plotkin JL, Wokosin DL, Chen Y, Kress GJ, Kaplitt M, Ilijic E, Guzman JN, Chan CS, Surmeier DJ. Mutant huntingtin enhances activation of dendritic Kv4 K + channels in striatal spiny projection neurons. eLife 2019; 8:e40818. [PMID: 31017573 PMCID: PMC6481990 DOI: 10.7554/elife.40818] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 04/02/2019] [Indexed: 01/09/2023] Open
Abstract
Huntington's disease (HD) is initially characterized by an inability to suppress unwanted movements, a deficit attributable to impaired synaptic activation of striatal indirect pathway spiny projection neurons (iSPNs). To better understand the mechanisms underlying this deficit, striatal neurons in ex vivo brain slices from mouse genetic models of HD were studied using electrophysiological, optical and biochemical approaches. Distal dendrites of iSPNs from symptomatic HD mice were hypoexcitable, a change that was attributable to increased association of dendritic Kv4 potassium channels with auxiliary KChIP subunits. This association was negatively modulated by TrkB receptor signaling. Dendritic excitability of HD iSPNs was rescued by knocking-down expression of Kv4 channels, by disrupting KChIP binding, by restoring TrkB receptor signaling or by lowering mutant-Htt (mHtt) levels with a zinc finger protein. Collectively, these studies demonstrate that mHtt induces reversible alterations in the dendritic excitability of iSPNs that could contribute to the motor symptoms of HD.
Collapse
Affiliation(s)
- Luis Carrillo-Reid
- Department of Physiology, Feinberg School of MedicineNorthwestern UniversityChicagoUnited States
- Department of Developmental Neurobiology and Neurophysiology, Neurobiology InstituteNational Autonomous University of MexicoQueretaroMexico
| | - Michelle Day
- Department of Physiology, Feinberg School of MedicineNorthwestern UniversityChicagoUnited States
| | - Zhong Xie
- Department of Physiology, Feinberg School of MedicineNorthwestern UniversityChicagoUnited States
| | - Alexandria E Melendez
- Department of Physiology, Feinberg School of MedicineNorthwestern UniversityChicagoUnited States
| | - Jyothisri Kondapalli
- Department of Physiology, Feinberg School of MedicineNorthwestern UniversityChicagoUnited States
| | - Joshua L Plotkin
- Department of Physiology, Feinberg School of MedicineNorthwestern UniversityChicagoUnited States
- Department of Neurobiology & BehaviorStony Brook University School of MedicineStony BrookUnited States
| | - David L Wokosin
- Department of Physiology, Feinberg School of MedicineNorthwestern UniversityChicagoUnited States
| | - Yu Chen
- Department of Physiology, Feinberg School of MedicineNorthwestern UniversityChicagoUnited States
| | - Geraldine J Kress
- Department of Physiology, Feinberg School of MedicineNorthwestern UniversityChicagoUnited States
- Department of NeurologyWashington University School of MedicineSt. LouisUnited States
| | - Michael Kaplitt
- Department of Neurological SurgeryWeill Cornell Medical CollegeNew YorkUnited States
| | - Ema Ilijic
- Department of Physiology, Feinberg School of MedicineNorthwestern UniversityChicagoUnited States
| | - Jaime N Guzman
- Department of Physiology, Feinberg School of MedicineNorthwestern UniversityChicagoUnited States
| | - C Savio Chan
- Department of Physiology, Feinberg School of MedicineNorthwestern UniversityChicagoUnited States
| | - D James Surmeier
- Department of Physiology, Feinberg School of MedicineNorthwestern UniversityChicagoUnited States
| |
Collapse
|
30
|
Raph SM, Bhatnagar A, Nystoriak MA. Biochemical and physiological properties of K + channel-associated AKR6A (Kvβ) proteins. Chem Biol Interact 2019; 305:21-27. [PMID: 30926318 DOI: 10.1016/j.cbi.2019.03.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/14/2019] [Accepted: 03/25/2019] [Indexed: 11/16/2022]
Abstract
Voltage-gated potassium (Kv) channels play an essential role in the regulation of membrane excitability and thereby control physiological processes such as cardiac excitability, neural communication, muscle contraction, and hormone secretion. Members of the Kv1 and Kv4 families are known to associate with auxiliary intracellular Kvβ subunits, which belong to the aldo-keto reductase superfamily. Electrophysiological studies have shown that these proteins regulate the gating properties of Kv channels. Although the three gene products encoding Kvβ proteins are functional enzymes in that they catalyze the nicotinamide adenine dinucleotide phosphate (NAD[P]H)-dependent reduction of a wide range of aldehyde and ketone substrates, the physiological role for these proteins and how each subtype may perform unique roles in coupling membrane excitability with cellular metabolic processes remains unclear. Here, we discuss current knowledge of the enzymatic properties of Kvβ proteins from biochemical studies with their described and purported physiological and pathophysiological influences.
Collapse
Affiliation(s)
- Sean M Raph
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA
| | - Aruni Bhatnagar
- Department of Medicine, Diabetes and Obesity Center, University of Louisville, Louisville, KY, 40202, USA
| | - Matthew A Nystoriak
- Department of Medicine, Diabetes and Obesity Center, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
31
|
Dwenger MM, Ohanyan V, Navedo MF, Nystoriak MA. Coronary microvascular Kv1 channels as regulatory sensors of intracellular pyridine nucleotide redox potential. Microcirculation 2018; 25. [PMID: 29110409 DOI: 10.1111/micc.12426] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/30/2017] [Indexed: 12/16/2022]
Abstract
Smooth muscle voltage-gated potassium (Kv) channels are important regulators of microvascular tone and tissue perfusion. Recent studies indicate that Kv1 channels represent a key component of the physiological coupling between coronary blood flow and myocardial oxygen demand. While the mechanisms by which metabolic changes in the heart are transduced to alter coronary Kv1 channel gating and promote vasodilation are unclear, a growing body of evidence underscores a pivotal role of Kv1 channels in sensing the cellular redox status. Here, we discuss current knowledge of mechanisms of Kv channel redox regulation with respect to pyridine nucleotide modulation of Kv1 function via ancillary Kvβ proteins as well as direct modulation of channel activity via reactive oxygen and nitrogen species. We identify areas of additional research to address the integration of regulatory processes under altered physiological and pathophysiological conditions that may reveal insights into novel treatment strategies for conditions in which the matching of coronary blood supply and myocardial oxygen demand is compromised.
Collapse
Affiliation(s)
- Marc M Dwenger
- Diabetes and Obesity Center, Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Vahagn Ohanyan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Manuel F Navedo
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Matthew A Nystoriak
- Diabetes and Obesity Center, Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY, USA
| |
Collapse
|
32
|
Lamothe SM, Hogan-Cann AE, Li W, Guo J, Yang T, Tschirhart JN, Zhang S. The N terminus and transmembrane segment S1 of Kv1.5 can coassemble with the rest of the channel independently of the S1-S2 linkage. J Biol Chem 2018; 293:15347-15358. [PMID: 30121572 DOI: 10.1074/jbc.ra118.004065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/14/2018] [Indexed: 11/06/2022] Open
Abstract
The voltage-gated potassium channel Kv1.5 belongs to the Shaker superfamily. Kv1.5 is composed of four subunits, each comprising 613 amino acids, which make up the N terminus, six transmembrane segments (S1-S6), and the C terminus. We recently demonstrated that, in HEK cells, extracellularly applied proteinase K (PK) cleaves Kv1.5 channels at a single site in the S1-S2 linker. This cleavage separates Kv1.5 into an N-fragment (N terminus to S1) and a C-fragment (S2 to C terminus). Interestingly, the cleavage does not impair channel function. Here, we investigated the role of the N terminus and S1 in Kv1.5 expression and function by creating plasmids encoding various fragments, including those that mimic PK-cleaved products. Our results disclosed that although expression of the pore-containing fragment (Frag(304-613)) alone could not produce current, coexpression with Frag(1-303) generated a functional channel. Immunofluorescence and biotinylation analyses uncovered that Frag(1-303) was required for Frag(304-613) to traffic to the plasma membrane. Biochemical analysis revealed that the two fragments interacted throughout channel trafficking and maturation. In Frag(1-303)+(304-613)-coassembled channels, which lack a covalent linkage between S1 and S2, amino acid residues 1-209 were important for association with Frag(304-613), and residues 210-303 were necessary for mediating trafficking of coassembled channels to the plasma membrane. We conclude that the N terminus and S1 of Kv1.5 can attract and coassemble with the rest of the channel (i.e. Frag(304-613)) to form a functional channel independently of the S1-S2 linkage.
Collapse
Affiliation(s)
- Shawn M Lamothe
- From the Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Aja E Hogan-Cann
- From the Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Wentao Li
- From the Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Jun Guo
- From the Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Tonghua Yang
- From the Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Jared N Tschirhart
- From the Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Shetuan Zhang
- From the Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario K7L 3N6, Canada
| |
Collapse
|
33
|
Niespodziany I, Mullier B, André VM, Ghisdal P, Jnoff E, Moreno-Delgado D, Swinnen D, Sands Z, Wood M, Wolff C. Discovery of a small molecule modulator of the Kv1.1/Kvβ1 channel complex that reduces neuronal excitability and in vitro epileptiform activity. CNS Neurosci Ther 2018; 25:442-451. [PMID: 30242974 DOI: 10.1111/cns.13060] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/13/2018] [Accepted: 08/15/2018] [Indexed: 11/27/2022] Open
Abstract
AIMS Kv1.1 (KCNA1) channels contribute to the control of neuronal excitability and have been associated with epilepsy. Kv1.1 channels can associate with the cytoplasmic Kvβ1 subunit resulting in rapid inactivating A-type currents. We hypothesized that removal of channel inactivation, by modulating Kv1.1/Kvβ1 interaction with a small molecule, would lead to decreased neuronal excitability and anticonvulsant activity. METHODS We applied high-throughput screening to identify ligands able to modulate the Kv1.1-T1 domain/Kvβ1 protein complex. We then selected a compound that was characterized on recombinant Kv1.1/Kvβ1 channels by electrophysiology and further evaluated on sustained neuronal firing and on in vitro epileptiform activity using a high K+ -low Ca2+ model in hippocampal slices. RESULTS We identified a novel compound able to modulate the interaction of the Kv1.1/Kvβ1 complex and that produced a functional inhibition of Kv1.1/Kvβ1 channel inactivation. We demonstrated that this compound reduced the sustained repetitive firing in hippocampal neurons and was able to abolish the development of in vitro epileptiform activity. CONCLUSIONS This study describes a rational drug discovery approach for the identification of novel ligands that inhibit Kv1.1 channel inactivation and provides pharmacological evidence that such a mechanism translates into physiological effects by reducing in vitro epileptiform activity.
Collapse
Affiliation(s)
| | - Brice Mullier
- Department of Neuroscience Research, UCB Pharma, Braine l'Alleud, Belgium
| | | | - Philippe Ghisdal
- Department of Neuroscience Research, UCB Pharma, Braine l'Alleud, Belgium
| | - Eric Jnoff
- Department of Neuroscience Research, UCB Pharma, Braine l'Alleud, Belgium
| | | | - Dominique Swinnen
- Department of Neuroscience Research, UCB Pharma, Braine l'Alleud, Belgium
| | - Zara Sands
- Department of Neuroscience Research, UCB Pharma, Braine l'Alleud, Belgium
| | - Martyn Wood
- Department of Neuroscience Research, UCB Pharma, Braine l'Alleud, Belgium
| | - Christian Wolff
- Department of Neuroscience Research, UCB Pharma, Braine l'Alleud, Belgium
| |
Collapse
|
34
|
Hong G, Pachter R, Ritz T. Coupling Drosophila melanogaster Cryptochrome Light Activation and Oxidation of the Kvβ Subunit Hyperkinetic NADPH Cofactor. J Phys Chem B 2018; 122:6503-6510. [PMID: 29847128 DOI: 10.1021/acs.jpcb.8b03493] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Motivated by the observations on the involvement of light-induced processes in the Drosophila melanogaster cryptochrome (DmCry) in regulation of the neuronal firing rate, which is achieved by a redox-state change of its voltage-dependent K+ channel Kvβ subunit hyperkinetic (Hk) reduced nicotinamide adenine dinucleotide phosphate (NADPH) cofactor, we propose in this work two hypothetical pathways that may potentially enable such coupling. In the first pathway, triggered by blue-light-induced formation of a radical pair [FAD•-TRP•+] in DmCry, the hole (TRP•+) may hop to Hk, for example, through a tryptophan chain and oxidize NADPH, possibly leading to inhibition of the N-terminus inactivation in the K+ channel. In a second possible pathway, DmCry's FAD•- is reoxidized by molecular oxygen, producing H2O2, which then diffuses to Hk and oxidizes NADPH. In this work, by applying a combination of quantum and empirical-based methods for free-energy calculations, we find that the oxidation of NADPH by TRP•+ or H2O2 and the reoxidation of FAD•- by O2 are thermodynamically feasible. Our results may have an implication in identifying a magnetic sensing signal transduction pathway, specifically upon Drosophila's Hk NADPH cofactor oxidation, with a subsequent inhibition of the K+ channel N-terminus inactivation gate, permitting K+ flux.
Collapse
Affiliation(s)
- Gongyi Hong
- Air Force Research Laboratory , Wright-Patterson Air Force Base , Dayton , Ohio 45433-7702 , United States
| | - Ruth Pachter
- Air Force Research Laboratory , Wright-Patterson Air Force Base , Dayton , Ohio 45433-7702 , United States
| | - Thorsten Ritz
- Department of Physics and Astronomy , University of California , Irvine , California 92697-4575 , United States
| |
Collapse
|
35
|
Baronas VA, Yang RY, Kurata HT. Extracellular redox sensitivity of Kv1.2 potassium channels. Sci Rep 2017; 7:9142. [PMID: 28831076 PMCID: PMC5567313 DOI: 10.1038/s41598-017-08718-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/17/2017] [Indexed: 12/03/2022] Open
Abstract
Kv1.2 is a prominent potassium channel subtype in the nervous system and serves as an important structural template for investigation of ion channel function. However, Kv1.2 voltage-dependence exhibits dramatic cell-to-cell variability due to a gating mode shift that is regulated by an unknown mechanism. We report that this variable behavior is regulated by the extracellular redox environment. Exposure to reducing agents promotes a shift in gating properties towards an 'inhibited' gating mode that resists opening, and causes channels to exhibit pronounced use-dependent activation during trains of repetitive depolarizations. This sensitivity to extracellular redox potential is absent in other Kv1 channels, but is apparent in heteromeric channels containing Kv1.2 subunits, and overlaps with the reported physiological range of extracellular redox couples. Mutagenesis of candidate cysteine residues fails to abolish redox sensitivity. Therefore, we suggest that an extrinsic, redox-sensitive binding partner imparts these properties.
Collapse
Affiliation(s)
- Victoria A Baronas
- Department of Pharmacology, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Runying Y Yang
- Department of Pharmacology, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Harley T Kurata
- Department of Pharmacology, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
36
|
Mutagenesis of the NaChBac sodium channel discloses a functional role for a conserved S6 asparagine. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2017; 46:665-674. [PMID: 28825121 PMCID: PMC5599482 DOI: 10.1007/s00249-017-1246-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 07/21/2017] [Accepted: 07/29/2017] [Indexed: 12/11/2022]
Abstract
Asparagine is conserved in the S6 transmembrane segments of all voltage-gated sodium, calcium, and TRP channels identified to date. A broad spectrum of channelopathies including cardiac arrhythmias, epilepsy, muscle diseases, and pain disorders is associated with its mutation. To investigate its effects on sodium channel functional properties, we mutated the simple prokaryotic sodium channel NaChBac. Electrophysiological characterization of the N225D mutant reveals that this conservative substitution shifts the voltage-dependence of inactivation by 25 mV to more hyperpolarized potentials. The mutant also displays greater thermostability, as determined by synchrotron radiation circular dichroism spectroscopy studies of purified channels. Based on our analyses of high-resolution structures of NaChBac homologues, we suggest that the side-chain amine group of asparagine 225 forms one or more hydrogen bonds with different channel elements and that these interactions are important for normal channel function. The N225D mutation eliminates these hydrogen bonds and the structural consequences involve an enhanced channel inactivation.
Collapse
|
37
|
Venom-derived peptide inhibitors of voltage-gated potassium channels. Neuropharmacology 2017; 127:124-138. [PMID: 28689025 DOI: 10.1016/j.neuropharm.2017.07.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/02/2017] [Accepted: 07/04/2017] [Indexed: 12/11/2022]
Abstract
Voltage-gated potassium channels play a key role in human physiology and pathology. Reflecting their importance, numerous channelopathies have been characterised that arise from mutations in these channels or from autoimmune attack on the channels. Voltage-gated potassium channels are also the target of a broad range of peptide toxins from venomous organisms, including sea anemones, scorpions, spiders, snakes and cone snails; many of these peptides bind to the channels with high potency and selectivity. In this review we describe the various classes of peptide toxins that block these channels and illustrate the broad range of three-dimensional structures that support channel blockade. The therapeutic opportunities afforded by these peptides are also highlighted. This article is part of the Special Issue entitled 'Venom-derived Peptides as Pharmacological Tools.'
Collapse
|
38
|
Heteromeric complexes of aldo-keto reductase auxiliary K Vβ subunits (AKR6A) regulate sarcolemmal localization of K V1.5 in coronary arterial myocytes. Chem Biol Interact 2017; 276:210-217. [PMID: 28342889 DOI: 10.1016/j.cbi.2017.03.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 03/02/2017] [Accepted: 03/21/2017] [Indexed: 01/20/2023]
Abstract
Redox-sensitive potassium channels consisting of the voltage-gated K+ (KV) channel pore subunit KV1.5 regulate resting membrane potential and thereby contractility of vascular smooth muscle cells. Members of the KV1 family associate with cytosolic auxiliary β subunits, which are members of the aldo-keto reductase (AKR) superfamily (AKR6A subfamily). The Kvβ subunits have been proposed to regulate Kv1 gating via pyridine nucleotide cofactor binding. However, the molecular identity of KVβ subunits that associate with native KV1.5 channels in the vasculature is unknown. Here, we examined mRNA and protein expression of KVβ subunits and tested whether KVβ isoforms interact with KV1.5 channels in murine coronary arteries. We detected KVβ1 (AKR6A3), KVβ2 (AKR6A5) and KVβ3 (AKR6A9) transcripts and KVβ1 and KVβ2 protein in left anterior descending coronary arteries by real time quantitative PCR and Western blot, respectively. In situ proximity ligation assays indicated abundant protein-protein interactions between KV1.5/KVβ1, KV1.5/KVβ2 and KVβ1/β2 in coronary arterial myocytes. Confocal microscopy and membrane fractionation analyses suggest that arterial myocytes from KVβ2-null mice have reduced abundance of sarcolemmal KV1.5. Together, data suggest that in coronary arterial myocytes, KV1.5 channels predominantly associate with KVβ1 and KVβ2 proteins and that KVβ2 performs a chaperone function for KV1.5 channels in arterial myocytes, thereby facilitating Kv1α trafficking and membrane localization.
Collapse
|
39
|
Unravelling the complexities of vascular smooth muscle ion channels: Fine tuning of activity by ancillary subunits. Pharmacol Ther 2017; 178:57-66. [PMID: 28336473 DOI: 10.1016/j.pharmthera.2017.03.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Which ion channel is the most important for regulating vascular tone? Which one is responsible for controlling the resting membrane potential or repolarization? Which channels are recruited by different intracellular signalling pathways or change in certain vascular diseases? Many different ion channels have been identified in the vasculature over the years and claimed as future therapeutic targets. Unfortunately, several of these ion channels are not just found in the vasculature, with many of them also found to have prominent functional roles in different organs of the body, which then leads to off-target effects. As cardiovascular diseases are expected to increase worldwide to epidemic proportions, ion channel research and the hunt for the next major therapeutic target to treat different vascular diseases has never been more important. However, I believe that the question we should now be asking is: which ancillary subunits are involved in regulating specific ion channels in the vasculature and do they have the potential to be new therapeutic targets?
Collapse
|
40
|
Moen MN, Fjær R, Hamdani EH, Laerdahl JK, Menchini RJ, Vigeland MD, Sheng Y, Undlien DE, Hassel B, Salih MA, El Khashab HY, Selmer KK, Chaudhry FA. Pathogenic variants in KCTD7 perturb neuronal K+ fluxes and glutamine transport. Brain 2016; 139:3109-3120. [PMID: 27742667 DOI: 10.1093/brain/aww244] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 06/11/2016] [Accepted: 08/17/2016] [Indexed: 12/11/2022] Open
Abstract
Progressive myoclonus epilepsy is a heterogeneous group of disorders characterized by myoclonic and tonic-clonic seizures, ataxia and cognitive decline. We here present two affected brothers. At 9 months of age the elder brother developed ataxia and myoclonic jerks. In his second year he lost the ability to walk and talk, and he developed drug-resistant progressive myoclonus epilepsy. The cerebrospinal fluid level of glutamate was decreased while glutamine was increased. His younger brother manifested similar symptoms from 6 months of age. By exome sequencing of the proband we identified a novel homozygous frameshift variant in the potassium channel tetramerization domain 7 (KCTD7) gene (NM_153033.1:c.696delT: p.F232fs), which results in a truncated protein. The identified F232fs variant is inherited in an autosomal recessive manner, and the healthy consanguineous parents carry the variant in a heterozygous state. Bioinformatic analyses and structure modelling showed that KCTD7 is a highly conserved protein, structurally similar to KCTD5 and several voltage-gated potassium channels, and that it may form homo- or heteromultimers. By heterologous expression in Xenopus laevis oocytes, we demonstrate that wild-type KCTD7 hyperpolarizes cells in a K+ dependent manner and regulates activity of the neuronal glutamine transporter SAT2 (Slc38a2), while the F232fs variant impairs K+ fluxes and obliterates SAT2-dependent glutamine transport. Characterization of four additional disease-causing variants (R94W, R184C, N273I, Y276C) bolster these results and reveal the molecular mechanisms involved in the pathophysiology of KCTD7-related progressive myoclonus epilepsy. Thus, our data demonstrate that KCTD7 has an impact on K+ fluxes, neurotransmitter synthesis and neuronal function, and that malfunction of the encoded protein may lead to progressive myoclonus epilepsy.
Collapse
Affiliation(s)
- Marivi Nabong Moen
- 1 The Institute of Basic Medical Sciences, Department of Molecular Medicine, University of Oslo, Oslo, Norway
| | - Roar Fjær
- 2 Department of Medical Genetics, Oslo University Hospital and University of Oslo, Norway
| | - El Hassan Hamdani
- 1 The Institute of Basic Medical Sciences, Department of Molecular Medicine, University of Oslo, Oslo, Norway.,3 Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Jon K Laerdahl
- 4 Department of Microbiology, Oslo University Hospital, Oslo, Norway.,5 Bioinformatics Core Facility, Department of Informatics, University of Oslo, Oslo, Norway
| | - Robin Johansen Menchini
- 1 The Institute of Basic Medical Sciences, Department of Molecular Medicine, University of Oslo, Oslo, Norway
| | - Magnus Dehli Vigeland
- 2 Department of Medical Genetics, Oslo University Hospital and University of Oslo, Norway
| | - Ying Sheng
- 2 Department of Medical Genetics, Oslo University Hospital and University of Oslo, Norway
| | - Dag Erik Undlien
- 2 Department of Medical Genetics, Oslo University Hospital and University of Oslo, Norway
| | - Bjørnar Hassel
- 6 Department of Complex Neurology and Neurohabilitation, Oslo University Hospital, Oslo, Norway
| | - Mustafa A Salih
- 7 Division of Paediatric Neurology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Heba Y El Khashab
- 7 Division of Paediatric Neurology, College of Medicine, King Saud University, Riyadh, Saudi Arabia.,8 Department of Paediatrics, Ain Shams University, Cairo, Egypt
| | - Kaja Kristine Selmer
- 2 Department of Medical Genetics, Oslo University Hospital and University of Oslo, Norway.,9 National Centre for Rare Epilepsy-related Disorders, Oslo University Hospital, Oslo, Norway
| | - Farrukh Abbas Chaudhry
- 1 The Institute of Basic Medical Sciences, Department of Molecular Medicine, University of Oslo, Oslo, Norway .,3 Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
41
|
Solé L, Roig SR, Vallejo-Gracia A, Serrano-Albarrás A, Martínez-Mármol R, Tamkun MM, Felipe A. The C-terminal domain of Kv1.3 regulates functional interactions with the KCNE4 subunit. J Cell Sci 2016; 129:4265-4277. [PMID: 27802162 DOI: 10.1242/jcs.191650] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 09/29/2016] [Indexed: 12/23/2022] Open
Abstract
The voltage-dependent K+ channel Kv1.3 (also known as KCNA3), which plays crucial roles in leukocytes, physically interacts with KCNE4. This interaction inhibits the K+ currents because the channel is retained within intracellular compartments. Thus, KCNE subunits are regulators of K+ channels in the immune system. Although the canonical interactions of KCNE subunits with Kv7 channels are under intensive investigation, the molecular determinants governing the important Kv1.3- KCNE4 association in the immune system are unknown. Our results suggest that the tertiary structure of the C-terminal domain of Kv1.3 is necessary and sufficient for such an interaction. However, this element is apparently not involved in modulating Kv1.3 gating. Furthermore, the KCNE4-dependent intracellular retention of the channel, which negatively affects the activity of Kv1.3, is mediated by two independent and additive mechanisms. First, KCNE4 masks the YMVIEE signature at the C-terminus of Kv1.3, which is crucial for the surface targeting of the channel. Second, we identify a potent endoplasmic reticulum retention motif in KCNE4 that further limits cell surface expression. Our results define specific molecular determinants that play crucial roles in the physiological function of Kv1.3 in leukocytes.
Collapse
Affiliation(s)
- Laura Solé
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, Barcelona 08028, Spain.,Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Sara R Roig
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, Barcelona 08028, Spain
| | - Albert Vallejo-Gracia
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, Barcelona 08028, Spain
| | - Antonio Serrano-Albarrás
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, Barcelona 08028, Spain
| | - Ramón Martínez-Mármol
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, Barcelona 08028, Spain.,Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Michael M Tamkun
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Antonio Felipe
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, Barcelona 08028, Spain
| |
Collapse
|
42
|
Basu S, Wallner B. DockQ: A Quality Measure for Protein-Protein Docking Models. PLoS One 2016; 11:e0161879. [PMID: 27560519 PMCID: PMC4999177 DOI: 10.1371/journal.pone.0161879] [Citation(s) in RCA: 223] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 08/12/2016] [Indexed: 01/26/2023] Open
Abstract
The state-of-the-art to assess the structural quality of docking models is currently based on three related yet independent quality measures: Fnat, LRMS, and iRMS as proposed and standardized by CAPRI. These quality measures quantify different aspects of the quality of a particular docking model and need to be viewed together to reveal the true quality, e.g. a model with relatively poor LRMS (>10Å) might still qualify as 'acceptable' with a descent Fnat (>0.50) and iRMS (<3.0Å). This is also the reason why the so called CAPRI criteria for assessing the quality of docking models is defined by applying various ad-hoc cutoffs on these measures to classify a docking model into the four classes: Incorrect, Acceptable, Medium, or High quality. This classification has been useful in CAPRI, but since models are grouped in only four bins it is also rather limiting, making it difficult to rank models, correlate with scoring functions or use it as target function in machine learning algorithms. Here, we present DockQ, a continuous protein-protein docking model quality measure derived by combining Fnat, LRMS, and iRMS to a single score in the range [0, 1] that can be used to assess the quality of protein docking models. By using DockQ on CAPRI models it is possible to almost completely reproduce the original CAPRI classification into Incorrect, Acceptable, Medium and High quality. An average PPV of 94% at 90% Recall demonstrating that there is no need to apply predefined ad-hoc cutoffs to classify docking models. Since DockQ recapitulates the CAPRI classification almost perfectly, it can be viewed as a higher resolution version of the CAPRI classification, making it possible to estimate model quality in a more quantitative way using Z-scores or sum of top ranked models, which has been so valuable for the CASP community. The possibility to directly correlate a quality measure to a scoring function has been crucial for the development of scoring functions for protein structure prediction, and DockQ should be useful in a similar development in the protein docking field. DockQ is available at http://github.com/bjornwallner/DockQ/.
Collapse
Affiliation(s)
- Sankar Basu
- Bioinformatics Division, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Björn Wallner
- Bioinformatics Division, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
- Swedish e-Science Research Center, Linköping University, Linköping, Sweden
| |
Collapse
|
43
|
Ca(2+)/calmodulin regulates Kvβ1.1-mediated inactivation of voltage-gated K(+) channels. Sci Rep 2015; 5:15509. [PMID: 26487174 PMCID: PMC4614385 DOI: 10.1038/srep15509] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 09/23/2015] [Indexed: 12/30/2022] Open
Abstract
A-type K+ channels open on membrane depolarization and undergo subsequent rapid inactivation such that they are ideally suited for fine-tuning the electrical signaling in neurons and muscle cells. Channel inactivation mostly follows the so-called ball-and-chain mechanism, in which the N-terminal structures of either the K+ channel’s α or β subunits occlude the channel pore entry facing the cytosol. Inactivation of Kv1.1 and Kv1.4 channels induced by Kvβ1.1 subunits is profoundly decelerated in response to a rise in the intracellular Ca2+ concentration, thus making the affected channel complexes negative feedback regulators to limit neuronal overexcitation. With electrophysiological and biochemical experiments we show that the Ca2+ dependence is gained by binding of calmodulin to the “chain” segment of Kvβ1.1 thereby compromising the mobility of the inactivation particle. Furthermore, inactivation regulation via Ca2+/calmodulin does not interfere with the β subunit’s enzymatic activity as an NADPH-dependent oxidoreductase, thus rendering the Kvβ1.1 subunit a multifunctional receptor that integrates cytosolic signals to be transduced to altered electrical cellular activity.
Collapse
|
44
|
The tetramerization domain potentiates Kv4 channel function by suppressing closed-state inactivation. Biophys J 2015; 107:1090-1104. [PMID: 25185545 DOI: 10.1016/j.bpj.2014.07.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 05/25/2014] [Accepted: 07/01/2014] [Indexed: 01/26/2023] Open
Abstract
A-type Kv4 potassium channels undergo a conformational change toward a nonconductive state at negative membrane potentials, a dynamic process known as pre-open closed states or closed-state inactivation (CSI). CSI causes inhibition of channel activity without the prerequisite of channel opening, thus providing a dynamic regulation of neuronal excitability, dendritic signal integration, and synaptic plasticity at resting. However, the structural determinants underlying Kv4 CSI remain largely unknown. We recently showed that the auxiliary KChIP4a subunit contains an N-terminal Kv4 inhibitory domain (KID) that directly interacts with Kv4.3 channels to enhance CSI. In this study, we utilized the KChIP4a KID to probe key structural elements underlying Kv4 CSI. Using fluorescence resonance energy transfer two-hybrid mapping and bimolecular fluorescence complementation-based screening combined with electrophysiology, we identified the intracellular tetramerization (T1) domain that functions to suppress CSI and serves as a receptor for the binding of KID. Disrupting the Kv4.3 T1-T1 interaction interface by mutating C110A within the C3H1 motif of T1 domain facilitated CSI and ablated the KID-mediated enhancement of CSI. Furthermore, replacing the Kv4.3 T1 domain with the T1 domain from Kv1.4 (without the C3H1 motif) or Kv2.1 (with the C3H1 motif) resulted in channels functioning with enhanced or suppressed CSI, respectively. Taken together, our findings reveal a novel (to our knowledge) role of the T1 domain in suppressing Kv4 CSI, and that KChIP4a KID directly interacts with the T1 domain to facilitate Kv4.3 CSI, thus leading to inhibition of channel function.
Collapse
|
45
|
El Hiani Y, Linsdell P. Functional Architecture of the Cytoplasmic Entrance to the Cystic Fibrosis Transmembrane Conductance Regulator Chloride Channel Pore. J Biol Chem 2015; 290:15855-15865. [PMID: 25944907 DOI: 10.1074/jbc.m115.656181] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Indexed: 12/19/2022] Open
Abstract
As an ion channel, the cystic fibrosis transmembrane conductance regulator must form a continuous pathway for the movement of Cl(-) and other anions between the cytoplasm and the extracellular solution. Both the structure and the function of the membrane-spanning part of this pathway are well defined. In contrast, the structure of the pathway that connects the cytoplasm to the membrane-spanning regions is unknown, and functional roles for different parts of the protein forming this pathway have not been described. We used patch clamp recording and substituted cysteine accessibility mutagenesis to identify positively charged amino acid side chains that attract cytoplasmic Cl(-) ions to the inner mouth of the pore. Our results indicate that the side chains of Lys-190, Arg-248, Arg-303, Lys-370, Lys-1041, and Arg-1048, located in different intracellular loops of the protein, play important roles in the electrostatic attraction of Cl(-) ions. Mutation and covalent modification of these residues have charge-dependent effects on the rate of Cl(-) permeation, demonstrating their functional role in maximization of Cl(-) flux. Other nearby positively charged side chains were not involved in electrostatic interactions with Cl(-). The location of these Cl(-)-attractive residues suggests that cytoplasmic Cl(-) ions enter the pore via a lateral portal located between the cytoplasmic extensions to the fourth and sixth transmembrane helices; a secondary, functionally less relevant portal might exist between the extensions to the 10th and 12th transmembrane helices. These results define the cytoplasmic mouth of the pore and show how it attracts Cl(-) ions from the cytoplasm.
Collapse
Affiliation(s)
- Yassine El Hiani
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Paul Linsdell
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada.
| |
Collapse
|
46
|
CRYPTOCHROME-mediated phototransduction by modulation of the potassium ion channel β-subunit redox sensor. Proc Natl Acad Sci U S A 2015; 112:2245-50. [PMID: 25646452 DOI: 10.1073/pnas.1416586112] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Blue light activation of the photoreceptor CRYPTOCHROME (CRY) evokes rapid depolarization and increased action potential firing in a subset of circadian and arousal neurons in Drosophila melanogaster. Here we show that acute arousal behavioral responses to blue light significantly differ in mutants lacking CRY, as well as mutants with disrupted opsin-based phototransduction. Light-activated CRY couples to membrane depolarization via a well conserved redox sensor of the voltage-gated potassium (K(+)) channel β-subunit (Kvβ) Hyperkinetic (Hk). The neuronal light response is almost completely absent in hk(-/-) mutants, but is functionally rescued by genetically targeted neuronal expression of WT Hk, but not by Hk point mutations that disable Hk redox sensor function. Multiple K(+) channel α-subunits that coassemble with Hk, including Shaker, Ether-a-go-go, and Ether-a-go-go-related gene, are ion conducting channels for CRY/Hk-coupled light response. Light activation of CRY is transduced to membrane depolarization, increased firing rate, and acute behavioral responses by the Kvβ subunit redox sensor.
Collapse
|
47
|
Domain Structure and Conformational Changes in rat KV2.1 ion Channel. J Neuroimmune Pharmacol 2014; 9:727-39. [DOI: 10.1007/s11481-014-9565-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 09/03/2014] [Indexed: 01/26/2023]
|
48
|
Abstract
Here we present the structure of the T1 domain derived from the voltage-dependent potassium channel K(v)1.3 of Homo sapiens sapiens at 1.2 Å resolution crystallized under near-physiological conditions. The crystals were grown without precipitant in 150 mM KP(i), pH 6.25. The crystals show I4 symmetry typical of the natural occurring tetrameric assembly of the single subunits. The obtained structural model is based on the highest resolution currently achieved for tetramerization domains of voltage-gated potassium channels. We identified an identical fold of the monomer but inside the tetramer the single monomers show a significant rotation which leads to a different orientation of the tetramer compared to other known structures. Such a rotational movement inside the tetrameric assembly might influence the gating properties of the channel. In addition we see two distinct side chain configurations for amino acids located in the top layer proximal to the membrane (Tyr109, Arg116, Ser129, Glu140, Met142, Arg146), and amino acids in the bottom layer of the T1-domain distal from the membrane (Val55, Ile56, Leu77, Arg86). The relative populations of these two states are ranging from 50:50 for Val55, Tyr109, Arg116, Ser129, Glu140, 60:40 for Met142, 65:35 for Arg86, 70:30 for Arg146, and 80:20 for Ile56 and Leu77. The data suggest that in solution these amino acids are involved in an equilibrium of conformational states that may be coupled to the functional states of the whole potassium channel.
Collapse
|
49
|
Jerng HH, Pfaffinger PJ. Modulatory mechanisms and multiple functions of somatodendritic A-type K (+) channel auxiliary subunits. Front Cell Neurosci 2014; 8:82. [PMID: 24723849 PMCID: PMC3973911 DOI: 10.3389/fncel.2014.00082] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 03/03/2014] [Indexed: 12/13/2022] Open
Abstract
Auxiliary subunits are non-conducting, modulatory components of the multi-protein ion channel complexes that underlie normal neuronal signaling. They interact with the pore-forming α-subunits to modulate surface distribution, ion conductance, and channel gating properties. For the somatodendritic subthreshold A-type potassium (ISA) channel based on Kv4 α-subunits, two types of auxiliary subunits have been extensively studied: Kv channel-interacting proteins (KChIPs) and dipeptidyl peptidase-like proteins (DPLPs). KChIPs are cytoplasmic calcium-binding proteins that interact with intracellular portions of the Kv4 subunits, whereas DPLPs are type II transmembrane proteins that associate with the Kv4 channel core. Both KChIPs and DPLPs genes contain multiple start sites that are used by various neuronal populations to drive the differential expression of functionally distinct N-terminal variants. In turn, these N-terminal variants generate tremendous functional diversity across the nervous system. Here, we focus our review on (1) the molecular mechanism underlying the unique properties of different N-terminal variants, (2) the shaping of native ISA properties by the concerted actions of KChIPs and DPLP variants, and (3) the surprising ways that KChIPs and DPLPs coordinate the activity of multiple channels to fine-tune neuronal excitability. Unlocking the unique contributions of different auxiliary subunit N-terminal variants may provide an important opportunity to develop novel targeted therapeutics to treat numerous neurological disorders.
Collapse
Affiliation(s)
- Henry H. Jerng
- Department of Neuroscience, Baylor College of MedicineHouston, TX, USA
| | | |
Collapse
|
50
|
Abstract
Many K(+) channels are oligomeric complexes with intrinsic structural symmetry arising from the homo-tetrameric core of their pore-forming subunits. Allosteric regulation of tetramerically symmetric proteins, whether by intrinsic sensing domains or associated auxiliary subunits, often mirrors the fourfold structural symmetry. Here, through patch-clamp recordings of channel population ensembles and also single channels, we examine regulation of the Ca(2+)- and voltage-activated large conductance Ca(2+)-activated K(+) (BK) channel by associated γ1-subunits. Through expression of differing ratios of γ1:α-subunits, the results reveal an all-or-none functional regulation of BK channels by γ-subunits: channels either exhibit a full gating shift or no shift at all. Furthermore, the γ1-induced shift exhibits a state-dependent labile behavior that recapitulates the fully shifted or unshifted behavior. The γ1-induced shift contrasts markedly to the incremental shifts in BK gating produced by 1-4 β-subunits and adds a new layer of complexity to the mechanisms by which BK channel functional diversity is generated.
Collapse
|