1
|
Thi Hong Van N, Hyun Nam J. Intermediate conductance calcium-activated potassium channel (KCa3.1) in cancer: Emerging roles and therapeutic potentials. Biochem Pharmacol 2024; 230:116573. [PMID: 39396649 DOI: 10.1016/j.bcp.2024.116573] [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: 07/20/2024] [Revised: 10/02/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
The KCa3.1 channel (also known as the KCNN4, IK1, or SK4 channel) is an intermediate-conductance calcium-activated potassium channel that regulates the membrane potential and maintains calcium homeostasis. Recently, KCa3.1 channels have attracted increasing attention because of their diverse roles in various types of cancers. In cancer cells, KCa3.1 channels regulate key processes, including cell proliferation, cell cycle, migration, invasion, tumor microenvironments, and therapy resistance. In addition, abnormal KCa3.1 expression in cancers is utilized to distinguish between tumor and normal tissues, classify cancer stages, and predict patient survival outcomes. This review comprehensively examines the current understanding of the contribution of KCa3.1 channels to tumor formation, metastasis, and its mechanisms. We evaluated the potential of KCa3.1 as a biomarker for cancer diagnosis and prognosis. Finally, we discuss the advances and challenges of applying KCa3.1 modulators in cancer treatment and propose approaches to overcome these obstacles. In summary, this review highlights the importance of this ion channel as a potent therapeutic target and prognostic biomarker of cancer.
Collapse
Affiliation(s)
- Nhung Thi Hong Van
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea; Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea; Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea.
| |
Collapse
|
2
|
Hirata K, Kudo K, Amano T, Kawaguchi M. Effects of the novel acaricide acynonapyr on the calcium-activated potassium channel. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 204:106074. [PMID: 39277387 DOI: 10.1016/j.pestbp.2024.106074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/30/2024] [Accepted: 08/03/2024] [Indexed: 09/17/2024]
Abstract
Resistance to insecticides and acaricides is a major impediment to effectively controlling insect pests worldwide. These pests include the two-spotted spider mite Tetranychus urticae (T. urticae), which exists globally. This polyphagous herbivore causes major agricultural problems and can develop resistance to the agents above. Therefore, the continuous development of acaricides with new modes of action is important to circumvent the resistance of insects to pesticides. Acynonapyr is a novel class of acaricides containing an azabicyclo ring. In this study, we determined the activity of acynonapyr and its analogs on calcium-activated potassium (KCa2) channels in two-spotted spider mites using electrophysiological techniques (patch-clamp). We also examined their acaricidal efficacy against mites in the laboratory. The acynonapyr and analogs blocked T. urticae KCa2 (TurKCa2) channels in a concentration-dependent manner. A comparison of acaricidal activity against T. urticae with inhibitory activity against TurKCa2 revealed that TurKCa2 channels are the primary toxicological targets. Finally, we examined the effect of acynonapyr on Homo sapiens KCa2 (HsaKCa2.2) channels and demonstrated that the compound at 10 μM had a limited effect on the activity of this channel.
Collapse
Affiliation(s)
- Koichi Hirata
- Marketing Section, Product Promotion Department, Agrochemical Division, Nippon Soda Co., Ltd., 7-2 Marunouchi 2-Chome, Chiyodaku, Tokyo 100-7010, Japan; Business Strategy and Administration Department, Chemical Business Division, Nippon Soda Co., Ltd.,.
| | - Kaori Kudo
- Insecticide Group, Biological Research Department, Research and Innovation Center, Nippon Soda Co., Ltd., 345 Takada, Odawara, Kanagawa 250-0216, Japan
| | - Tomohiro Amano
- Insecticide Group, Biological Research Department, Research and Innovation Center, Nippon Soda Co., Ltd., 345 Takada, Odawara, Kanagawa 250-0216, Japan
| | - Masahiro Kawaguchi
- Insecticide Group, Biological Research Department, Research and Innovation Center, Nippon Soda Co., Ltd., 345 Takada, Odawara, Kanagawa 250-0216, Japan
| |
Collapse
|
3
|
Zhang Y, Shaabani S, Vowinkel K, Trombetta-Lima M, Sabogal-Guáqueta AM, Chen T, Hoekstra J, Lembeck J, Schmidt M, Decher N, Dömling A, Dolga AM. Novel SK channel positive modulators prevent ferroptosis and excitotoxicity in neuronal cells. Biomed Pharmacother 2024; 171:116163. [PMID: 38242037 DOI: 10.1016/j.biopha.2024.116163] [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: 10/23/2023] [Revised: 01/07/2024] [Accepted: 01/11/2024] [Indexed: 01/21/2024] Open
Abstract
Small conductance calcium-activated potassium (SK) channel activity has been proposed to play a role in the pathology of several neurological diseases. Besides regulating plasma membrane excitability, SK channel activation provides neuroprotection against ferroptotic cell death by reducing mitochondrial Ca2+ uptake and reactive oxygen species (ROS). In this study, we employed a multifaceted approach, integrating structure-based and computational techniques, to strategically design and synthesize an innovative class of potent small-molecule SK2 channel modifiers through highly efficient multicomponent reactions (MCRs). The compounds' neuroprotective activity was compared with the well-studied SK positive modulator, CyPPA. Pharmacological SK channel activation by selected compounds confers neuroprotection against ferroptosis at low nanomolar ranges compared to CyPPA, that mediates protection at micromolar concentrations, as shown by an MTT assay, real-time cell impedance measurements and propidium iodide staining (PI). These novel compounds suppress increased mitochondrial ROS and Ca2+ level induced by ferroptosis inducer RSL3. Moreover, axonal degeneration was rescued by these novel SK channel activators in primary mouse neurons and they attenuated glutamate-induced neuronal excitability, as shown via microelectrode array. Meanwhile, functional afterhyperpolarization of the novel SK2 channel modulators was validated by electrophysiological measurements showing more current change induced by the novel modulators than the reference compound, CyPPA. These data support the notion that SK2 channel activation can represent a therapeutic target for brain diseases in which ferroptosis and excitotoxicity contribute to the pathology.
Collapse
Affiliation(s)
- Yuequ Zhang
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Shabnam Shaabani
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Kirsty Vowinkel
- Institute of Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, 35037 Marburg, Germany
| | - Marina Trombetta-Lima
- Department of Pharmaceutical Technologies and Biopharmacy, Research Institute of Pharmacy, University of Groningen, the Netherlands
| | | | - Tingting Chen
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Jan Hoekstra
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Jan Lembeck
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Martina Schmidt
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Niels Decher
- Institute of Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, 35037 Marburg, Germany
| | - Alexander Dömling
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands.
| | - Amalia M Dolga
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands.
| |
Collapse
|
4
|
Dewey JA, Delalande C, Azizi SA, Lu V, Antonopoulos D, Babnigg G. Molecular Glue Discovery: Current and Future Approaches. J Med Chem 2023; 66:9278-9296. [PMID: 37437222 PMCID: PMC10805529 DOI: 10.1021/acs.jmedchem.3c00449] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
The intracellular interactions of biomolecules can be maneuvered to redirect signaling, reprogram the cell cycle, or decrease infectivity using only a few dozen atoms. Such "molecular glues," which can drive both novel and known interactions between protein partners, represent an enticing therapeutic strategy. Here, we review the methods and approaches that have led to the identification of small-molecule molecular glues. We first classify current FDA-approved molecular glues to facilitate the selection of discovery methods. We then survey two broad discovery method strategies, where we highlight the importance of factors such as experimental conditions, software packages, and genetic tools for success. We hope that this curation of methodologies for directed discovery will inspire diverse research efforts targeting a multitude of human diseases.
Collapse
Affiliation(s)
- Jeffrey A Dewey
- Biosciences Division, Argonne National Laboratory, Lemont, Illinois 60439, United States
| | - Clémence Delalande
- Department of Chemistry, University of Chicago, Chicago, Illinois 60637, United States
| | - Saara-Anne Azizi
- Pritzker School of Medicine, University of Chicago, Chicago, Illinois 60637, United States
| | - Vivian Lu
- Department of Chemistry, University of Chicago, Chicago, Illinois 60637, United States
| | - Dionysios Antonopoulos
- Biosciences Division, Argonne National Laboratory, Lemont, Illinois 60439, United States
| | - Gyorgy Babnigg
- Biosciences Division, Argonne National Laboratory, Lemont, Illinois 60439, United States
| |
Collapse
|
5
|
Wubulikasimu A, He Z, Long Y, Yuan F, Hou W, Liao Q, Chen H, Rong M. Molecular mechanism of HNTX-I activates the intermediate-conductance Ca 2+-activated K + (IK) channels. Int J Biol Macromol 2023:125197. [PMID: 37285887 DOI: 10.1016/j.ijbiomac.2023.125197] [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: 05/01/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/09/2023]
Abstract
The IK channel, a potassium ion channel regulated by calcium ions and voltages in a bidirectional manner, has been implicated in a range of diseases. However, there are currently few compounds available that can target the IK channel with high potency and specificity. Hainantoxin-I (HNTX-I) is the first peptide activator of IK channel discovered so far, but its activity is not ideal, and the underlying mechanism interaction between HNTX-I toxin and IK channel remains unclear. Thus, our study aimed to enhance the potency of IK channel activating peptides derived from HNTX-I and elucidate the molecular mechanism underlying the interaction between HNTX-I and the IK channel. By employing virtual alanine scanning mutagenesis, we generated 11 HNTX-I mutants using site-directed mutagenesis to pinpoint specific residues crucial for the HNTX-I and IK channel interaction. Subsequently, we identified key residues on the IK channel that are involved in the interaction with HNTX-I. Additionally, molecular docking was employed to guide the molecular engineering process and clarify the binding interface between HNTX-I and the IK channel. Our results demonstrate that HNTX-I primarily acts on the IK channel via the N-terminal amino acid, and its interaction with the IK channel is mediated by electrostatic and hydrophobic interactions, specifically the amino acid residues at positions 1, 3, 5, and 7 on HNTX-I. This study provides valuable insights into the peptide toxins that may serve as potential templates for the development of activators with enhanced potency and selectivity for the IK channel.
Collapse
Affiliation(s)
- Atikan Wubulikasimu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, People's Republic of China
| | - Ziyan He
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, People's Republic of China
| | - Yanping Long
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, People's Republic of China
| | - Fuchu Yuan
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, People's Republic of China
| | - Wenqian Hou
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, People's Republic of China
| | - Qiong Liao
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, People's Republic of China
| | - Haiyan Chen
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, People's Republic of China.
| | - Mingqiang Rong
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, People's Republic of China.
| |
Collapse
|
6
|
Rui H, Ashton KS, Min J, Wang C, Potts PR. Protein-protein interfaces in molecular glue-induced ternary complexes: classification, characterization, and prediction. RSC Chem Biol 2023; 4:192-215. [PMID: 36908699 PMCID: PMC9994104 DOI: 10.1039/d2cb00207h] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/02/2023] [Indexed: 01/04/2023] Open
Abstract
Molecular glues are a class of small molecules that stabilize the interactions between proteins. Naturally occurring molecular glues are present in many areas of biology where they serve as central regulators of signaling pathways. Importantly, several clinical compounds act as molecular glue degraders that stabilize interactions between E3 ubiquitin ligases and target proteins, leading to their degradation. Molecular glues hold promise as a new generation of therapeutic agents, including those molecular glue degraders that can redirect the protein degradation machinery in a precise way. However, rational discovery of molecular glues is difficult in part due to the lack of understanding of the protein-protein interactions they stabilize. In this review, we summarize the structures of known molecular glue-induced ternary complexes and the interface properties. Detailed analysis shows different mechanisms of ternary structure formation. Additionally, we also review computational approaches for predicting protein-protein interfaces and highlight the promises and challenges. This information will ultimately help inform future approaches for rational molecular glue discovery.
Collapse
Affiliation(s)
- Huan Rui
- Center for Research Acceleration by Digital Innovation, Amgen Research Thousand Oaks CA 91320 USA
| | - Kate S Ashton
- Medicinal Chemistry, Amgen Research Thousand Oaks CA 91320 USA
| | - Jaeki Min
- Induced Proximity Platform, Amgen Research Thousand Oaks CA 91320 USA
| | - Connie Wang
- Digital, Technology & Innovation, Amgen Thousand Oaks CA 91320 USA
| | | |
Collapse
|
7
|
Halling DB, Philpo AE, Aldrich RW. Calcium dependence of both lobes of calmodulin is involved in binding to a cytoplasmic domain of SK channels. eLife 2022; 11:e81303. [PMID: 36583726 PMCID: PMC9803350 DOI: 10.7554/elife.81303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 12/15/2022] [Indexed: 12/31/2022] Open
Abstract
KCa2.1-3 Ca2+-activated K+-channels (SK) require calmodulin to gate in response to cellular Ca2+. A model for SK gating proposes that the N-terminal domain (N-lobe) of calmodulin is required for activation, but an immobile C-terminal domain (C-lobe) has constitutive, Ca2+-independent binding. Although structures support a domain-driven hypothesis of SK gate activation by calmodulin, only a partial understanding is possible without measuring both channel activity and protein binding. We measured SK2 (KCa2.2) activity using inside-out patch recordings. Currents from calmodulin-disrupted SK2 channels can be restored with exogenously applied calmodulin. We find that SK2 activity only approaches full activation with full-length calmodulin with both an N- and a C-lobe. We measured calmodulin binding to a C-terminal SK peptide (SKp) using both composition-gradient multi-angle light-scattering and tryptophan emission spectra. Isolated lobes bind to SKp with high affinity, but isolated lobes do not rescue SK2 activity. Consistent with earlier models, N-lobe binding to SKp is stronger in Ca2+, and C-lobe-binding affinity is strong independent of Ca2+. However, a native tryptophan in SKp is sensitive to Ca2+ binding to both the N- and C-lobes of calmodulin at Ca2+ concentrations that activate SK2, demonstrating that the C-lobe interaction with SKp changes with Ca2+. Our peptide-binding data and electrophysiology show that SK gating models need deeper scrutiny. We suggest that the Ca2+-dependent associations of both lobes of calmodulin to SKp are crucial events during gating. Additional investigations are necessary to complete a mechanistic gating model consistent with binding, physiology, and structure.
Collapse
Affiliation(s)
- David B Halling
- Department of Neuroscience, The University of Texas at AustinAustinUnited States
| | - Ashley E Philpo
- Department of Neuroscience, The University of Texas at AustinAustinUnited States
| | - Richard W Aldrich
- Department of Neuroscience, The University of Texas at AustinAustinUnited States
| |
Collapse
|
8
|
Allosteric inhibitors targeting the calmodulin-PIP2 interface of SK4 K + channels for atrial fibrillation treatment. Proc Natl Acad Sci U S A 2022; 119:e2202926119. [PMID: 35969786 PMCID: PMC9407317 DOI: 10.1073/pnas.2202926119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Ca2+-activated SK4 K+ channel is gated by Ca2+-calmodulin (CaM) and is expressed in immune cells, brain, and heart. A cryoelectron microscopy (cryo-EM) structure of the human SK4 K+ channel recently revealed four CaM molecules per channel tetramer, where the apo CaM C-lobe and the holo CaM N-lobe interact with the proximal carboxyl terminus and the linker S4-S5, respectively, to gate the channel. Here, we show that phosphatidylinositol 4-5 bisphosphate (PIP2) potently activates SK4 channels by docking to the boundary of the CaM-binding domain. An allosteric blocker, BA6b9, was designed to act to the CaM-PIP2-binding domain, a previously untargeted region of SK4 channels, at the interface of the proximal carboxyl terminus and the linker S4-S5. Site-directed mutagenesis, molecular docking, and patch-clamp electrophysiology indicate that BA6b9 inhibits SK4 channels by interacting with two specific residues, Arg191 and His192 in the linker S4-S5, not conserved in SK1-SK3 subunits, thereby conferring selectivity and preventing the Ca2+-CaM N-lobe from properly interacting with the channel linker region. Immunohistochemistry of the SK4 channel protein in rat hearts showed a widespread expression in the sarcolemma of atrial myocytes, with a sarcomeric striated Z-band pattern, and a weaker occurrence in the ventricle but a marked incidence at the intercalated discs. BA6b9 significantly prolonged atrial and atrioventricular effective refractory periods in rat isolated hearts and reduced atrial fibrillation induction ex vivo. Our work suggests that inhibition of SK4 K+ channels by targeting drugs to the CaM-PIP2-binding domain provides a promising anti-arrhythmic therapy.
Collapse
|
9
|
Ahmed M, Ganesan A, Barakat K. Leveraging structural and 2D-QSAR to investigate the role of functional group substitutions, conserved surface residues and desolvation in triggering the small molecule-induced dimerization of hPD-L1. BMC Chem 2022; 16:49. [PMID: 35761353 PMCID: PMC9238240 DOI: 10.1186/s13065-022-00842-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 06/21/2022] [Indexed: 12/02/2022] Open
Abstract
Small molecules are rising as a new generation of immune checkpoints’ inhibitors, with compounds targeting the human Programmed death-ligand 1 (hPD-L1) protein are pioneering this area of research. Promising examples include the recently disclosed compounds from Bristol-Myers-Squibb (BMS). These molecules bind specifically to hPD-L1 through a unique mode of action. They induce dimerization between two hPD-L1 monomers through the hPD-1 binding interface in each monomer, thereby inhibiting the PD-1/PD-L1 axis. While the recently reported crystal structures of such small molecules bound to hPD-L1 reveal valuable insights regarding their molecular interactions, there is still limited information about the dynamics driving this unusual complex formation. The current study provides an in-depth computational structural analysis to study the interactions of five small molecule compounds in complex with hPD-L1. By employing a combination of molecular dynamic simulations, binding energy calculations and computational solvent mapping techniques, our analyses quantified the dynamic roles of different hydrophilic and lipophilic residues at the surface of hPD-L1 in mediating these interactions. Furthermore, ligand-based analyses, including Free-Wilson 2D-QSAR was conducted to quantify the impact of R-group substitutions at different sites of the phenoxy-methyl biphenyl core. Our results emphasize the importance of a terminal phenyl ring that must be present in any hPD-L1 small molecule inhibitor. This phenyl moiety overlaps with a very unfavorable hydration site, which can explain the ability of such small molecules to trigger hPD-L1 dimerization.
Collapse
Affiliation(s)
- Marawan Ahmed
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Aravindhan Ganesan
- ArGan's Lab, School of Pharmacy, University of Waterloo, Kitchener, ON, Canada
| | - Khaled Barakat
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada. .,Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
10
|
Nam YW, Cui M, Salem El-Sayed N, Orfali R, Nguyen M, Yang G, Rahman MA, Lee J, Zhang M. Subtype-selective positive modulation of K Ca 2 channels depends on the HA/HB helices. Br J Pharmacol 2022; 179:460-472. [PMID: 34458981 PMCID: PMC8799485 DOI: 10.1111/bph.15676] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 06/29/2021] [Accepted: 07/29/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND AND PURPOSE In the activated state of small-conductance Ca2+ -activated potassium (KCa 2) channels, calmodulin interacts with the HA/HB helices and the S4-S5 linker. CyPPA potentiates KCa 2.2a and KCa 2.3 channel activity but not the KCa 2.1 and KCa 3.1 subtypes. EXPERIMENTAL APPROACH Site-directed mutagenesis, patch-clamp recordings and in silico modelling were utilised to explore the structural determinants for the subtype-selective modulation of KCa 2 channels by CyPPA. KEY RESULTS Mutating residues in the HA (V420) and HB (K467) helices of KCa 2.2a channels to their equivalent residues in KCa 3.1 channels diminished the potency of CyPPA. CyPPA elicited prominent responses on mutant KCa 3.1 channels with an arginine residue in the HB helix substituted for its equivalent lysine residue in the KCa 2.2a channels (R355K). KCa 2.1 channels harbouring a three-amino-acid insertion upstream of the cognate R438 residues in the HB helix showed no response to CyPPA, whereas the deletion mutant (KCa 2.1_ΔA434/Q435/K436) became sensitive to CyPPA. In molecular dynamics simulations, CyPPA docked between calmodulin C-lobe and the HA/HB helices widens the cytoplasmic gate of KCa 2.2a channels. CONCLUSION AND IMPLICATIONS Selectivity of CyPPA among KCa 2 and KCa 3.1 channel subtypes relies on the HA/HB helices.
Collapse
Affiliation(s)
- Young-Woo Nam
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, USA
| | - Meng Cui
- Department of Pharmaceutical Sciences, Northeastern University School of Pharmacy, Boston, Massachusetts, 02115, USA
| | - Naglaa Salem El-Sayed
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, USA
| | - Razan Orfali
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, USA
| | - Misa Nguyen
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, USA
| | - Grace Yang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, USA
| | - Mohammad Asikur Rahman
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, USA
| | - Judy Lee
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, USA
| | - Miao Zhang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, USA
| |
Collapse
|
11
|
Rayff da Silva P, do Nascimento Gonzaga TKS, Maia RE, Araújo da Silva B. Ionic Channels as Potential Targets for the Treatment of Autism Spectrum Disorder: A Review. Curr Neuropharmacol 2022; 20:1834-1849. [PMID: 34370640 PMCID: PMC9886809 DOI: 10.2174/1570159x19666210809102547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/23/2021] [Accepted: 07/24/2021] [Indexed: 11/22/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurological condition that directly affects brain functions and can culminate in delayed intellectual development, problems in verbal communication, difficulties in social interaction, and stereotyped behaviors. Its etiology reveals a genetic basis that can be strongly influenced by socio-environmental factors. Ion channels controlled by ligand voltage-activated calcium, sodium, and potassium channels may play important roles in modulating sensory and cognitive responses, and their dysfunctions may be closely associated with neurodevelopmental disorders such as ASD. This is due to ionic flow, which is of paramount importance to maintaining physiological conditions in the central nervous system and triggers action potentials, gene expression, and cell signaling. However, since ASD is a multifactorial disease, treatment is directed only to secondary symptoms. Therefore, this research aims to gather evidence concerning the principal pathophysiological mechanisms involving ion channels in order to recognize their importance as therapeutic targets for the treatment of central and secondary ASD symptoms.
Collapse
Affiliation(s)
| | | | | | - Bagnólia Araújo da Silva
- Address correspondence to this author at the Postgraduate Program in Natural Synthetic and Bioactive Products, Heath Sciences Center, Federal University of Paraíba - Campus I, 58051-085, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil; Tel: ++55-83-99352-5595; E-mail:
| |
Collapse
|
12
|
Tiffner A, Hopl V, Schober R, Sallinger M, Grabmayr H, Höglinger C, Fahrner M, Lunz V, Maltan L, Frischauf I, Krivic D, Bhardwaj R, Schindl R, Hediger MA, Derler I. Orai1 Boosts SK3 Channel Activation. Cancers (Basel) 2021; 13:6357. [PMID: 34944977 PMCID: PMC8699475 DOI: 10.3390/cancers13246357] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 12/13/2021] [Indexed: 12/20/2022] Open
Abstract
The interplay of SK3, a Ca2+ sensitive K+ ion channel, with Orai1, a Ca2+ ion channel, has been reported to increase cytosolic Ca2+ levels, thereby triggering proliferation of breast and colon cancer cells, although a molecular mechanism has remained elusive to date. We show in the current study, via heterologous protein expression, that Orai1 can enhance SK3 K+ currents, in addition to constitutively bound calmodulin (CaM). At low cytosolic Ca2+ levels that decrease SK3 K+ permeation, co-expressed Orai1 potentiates SK3 currents. This positive feedback mechanism of SK3 and Orai1 is enabled by their close co-localization. Remarkably, we discovered that loss of SK3 channel activity due to overexpressed CaM mutants could be restored by Orai1, likely via its interplay with the SK3-CaM binding site. Mapping for interaction sites within Orai1, we identified that the cytosolic strands and pore residues are critical for a functional communication with SK3. Moreover, STIM1 has a bimodal role in SK3-Orai1 regulation. Under physiological ionic conditions, STIM1 is able to impede SK3-Orai1 interplay by significantly decreasing their co-localization. Forced STIM1-Orai1 activity and associated Ca2+ influx promote SK3 K+ currents. The dynamic regulation of Orai1 to boost endogenous SK3 channels was also determined in the human prostate cancer cell line LNCaP.
Collapse
Affiliation(s)
- Adéla Tiffner
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Valentina Hopl
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Romana Schober
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
- Gottfried Schatz Research Centre, Medical University of Graz, A-8010 Graz, Austria; (D.K.); (R.S.)
| | - Matthias Sallinger
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Herwig Grabmayr
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Carmen Höglinger
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Marc Fahrner
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Victoria Lunz
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Lena Maltan
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Irene Frischauf
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Denis Krivic
- Gottfried Schatz Research Centre, Medical University of Graz, A-8010 Graz, Austria; (D.K.); (R.S.)
| | - Rajesh Bhardwaj
- Department of Nephrology and Hypertension, University of Bern, Inselspital, Freiburgstrasse 15, CH-3010 Bern, Switzerland; (R.B.); (M.A.H.)
- Department of Biomedical Research, University of Bern, Inselspital, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Rainer Schindl
- Gottfried Schatz Research Centre, Medical University of Graz, A-8010 Graz, Austria; (D.K.); (R.S.)
| | - Matthias A. Hediger
- Department of Nephrology and Hypertension, University of Bern, Inselspital, Freiburgstrasse 15, CH-3010 Bern, Switzerland; (R.B.); (M.A.H.)
- Department of Biomedical Research, University of Bern, Inselspital, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Isabella Derler
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| |
Collapse
|
13
|
Matos B, Howl J, Jerónimo C, Fardilha M. Modulation of serine/threonine-protein phosphatase 1 (PP1) complexes: A promising approach in cancer treatment. Drug Discov Today 2021; 26:2680-2698. [PMID: 34390863 DOI: 10.1016/j.drudis.2021.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/23/2021] [Accepted: 08/05/2021] [Indexed: 01/21/2023]
Abstract
Cancer is the second leading cause of death worldwide. Despite the availability of numerous therapeutic options, tumor heterogeneity and chemoresistance have limited the success of these treatments, and the development of effective anticancer therapies remains a major focus in oncology research. The serine/threonine-protein phosphatase 1 (PP1) and its complexes have been recognized as potential drug targets. Research on the modulation of PP1 complexes is currently at an early stage, but has immense potential. Chemically diverse compounds have been developed to disrupt or stabilize different PP1 complexes in various cancer types, with the objective of inhibiting disease progression. Beneficial results obtained in vitro now require further pre-clinical and clinical validation. In conclusion, the modulation of PP1 complexes seems to be a promising, albeit challenging, therapeutic strategy for cancer.
Collapse
Affiliation(s)
- Bárbara Matos
- Laboratory of Signal Transduction, Department of Medical Sciences, Institute of Biomedicine-iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal; Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal
| | - John Howl
- Molecular Pharmacology Group, Research Institute in Healthcare Science, University of Wolverhampton, Wolverhampton WV1 1LY, UK
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal; Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), 4050-513 Porto, Portugal
| | - Margarida Fardilha
- Laboratory of Signal Transduction, Department of Medical Sciences, Institute of Biomedicine-iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
14
|
Nam YW, Kong D, Wang D, Orfali R, Sherpa RT, Totonchy J, Nauli SM, Zhang M. Differential modulation of SK channel subtypes by phosphorylation. Cell Calcium 2021; 94:102346. [PMID: 33422768 PMCID: PMC8415101 DOI: 10.1016/j.ceca.2020.102346] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 01/01/2023]
Abstract
Small-conductance Ca2+-activated K+ (SK) channels are voltage-independent and are activated by Ca2+ binding to the calmodulin constitutively associated with the channels. Both the pore-forming subunits and the associated calmodulin are subject to phosphorylation. Here, we investigated the modulation of different SK channel subtypes by phosphorylation, using the cultured endothelial cells as a tool. We report that casein kinase 2 (CK2) negatively modulates the apparent Ca2+ sensitivity of SK1 and IK channel subtypes by more than 5-fold, whereas the apparent Ca2+ sensitivity of the SK3 and SK2 subtypes is only reduced by ∼2-fold, when heterologously expressed on the plasma membrane of cultured endothelial cells. The SK2 channel subtype exhibits limited cell surface expression in these cells, partly as a result of the phosphorylation of its C-terminus by cyclic AMP-dependent protein kinase (PKA). SK2 channels expressed on the ER and mitochondria membranes may protect against cell death. This work reveals the subtype-specific modulation of the apparent Ca2+ sensitivity and subcellular localization of SK channels by phosphorylation in cultured endothelial cells.
Collapse
Affiliation(s)
- Young-Woo Nam
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Dezhi Kong
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, Hebei, 050017, China
| | - Dong Wang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Razan Orfali
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Rinzhin T Sherpa
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Jennifer Totonchy
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Surya M Nauli
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Miao Zhang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA.
| |
Collapse
|
15
|
Nam YW, Cui M, Orfali R, Viegas A, Nguyen M, Mohammed EHM, Zoghebi KA, Rahighi S, Parang K, Zhang M. Hydrophobic interactions between the HA helix and S4-S5 linker modulate apparent Ca 2+ sensitivity of SK2 channels. Acta Physiol (Oxf) 2021; 231:e13552. [PMID: 32865319 PMCID: PMC7736289 DOI: 10.1111/apha.13552] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/09/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022]
Abstract
AIM Small-conductance Ca2+ -activated potassium (SK) channels are activated exclusively by increases in intracellular Ca2+ that binds to calmodulin constitutively associated with the channel. Wild-type SK2 channels are activated by Ca2+ with an EC50 value of ~0.3 μmol/L. Here, we investigate hydrophobic interactions between the HA helix and the S4-S5 linker as a major determinant of channel apparent Ca2+ sensitivity. METHODS Site-directed mutagenesis, electrophysiological recordings and molecular dynamic (MD) simulations were utilized. RESULTS Mutations that decrease hydrophobicity at the HA-S4-S5 interface lead to Ca2+ hyposensitivity of SK2 channels. Mutations that increase hydrophobicity result in hypersensitivity to Ca2+ . The Ca2+ hypersensitivity of the V407F mutant relies on the interaction of the cognate phenylalanine with the S4-S5 linker in the SK2 channel. Replacing the S4-S5 linker of the SK2 channel with the S4-S5 linker of the SK4 channel results in loss of the hypersensitivity caused by V407F. This difference between the S4-S5 linkers of SK2 and SK4 channels can be partially attributed to I295 equivalent to a valine in the SK4 channel. A N293A mutation in the S4-S5 linker also increases hydrophobicity at the HA-S4-S5 interface and elevates the channel apparent Ca2+ sensitivity. The double N293A/V407F mutations generate a highly Ca2+ sensitive channel, with an EC50 of 0.02 μmol/L. The MD simulations of this double-mutant channel revealed a larger channel cytoplasmic gate. CONCLUSION The electrophysiological data and MD simulations collectively suggest a crucial role of the interactions between the HA helix and S4-S5 linker in the apparent Ca2+ sensitivity of SK2 channels.
Collapse
Affiliation(s)
- Young-Woo Nam
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Meng Cui
- Department of Pharmaceutical Sciences, Northeastern University School of Pharmacy, Boston, MA, USA
| | - Razan Orfali
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Adam Viegas
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Misa Nguyen
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Eman H M Mohammed
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Khalid A Zoghebi
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Simin Rahighi
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Keykavous Parang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Miao Zhang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| |
Collapse
|
16
|
Tiffner A, Derler I. Molecular Choreography and Structure of Ca 2+ Release-Activated Ca 2+ (CRAC) and K Ca2+ Channels and Their Relevance in Disease with Special Focus on Cancer. MEMBRANES 2020; 10:E425. [PMID: 33333945 PMCID: PMC7765462 DOI: 10.3390/membranes10120425] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/16/2022]
Abstract
Ca2+ ions play a variety of roles in the human body as well as within a single cell. Cellular Ca2+ signal transduction processes are governed by Ca2+ sensing and Ca2+ transporting proteins. In this review, we discuss the Ca2+ and the Ca2+-sensing ion channels with particular focus on the structure-function relationship of the Ca2+ release-activated Ca2+ (CRAC) ion channel, the Ca2+-activated K+ (KCa2+) ion channels, and their modulation via other cellular components. Moreover, we highlight their roles in healthy signaling processes as well as in disease with a special focus on cancer. As KCa2+ channels are activated via elevations of intracellular Ca2+ levels, we summarize the current knowledge on the action mechanisms of the interplay of CRAC and KCa2+ ion channels and their role in cancer cell development.
Collapse
Affiliation(s)
| | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria;
| |
Collapse
|
17
|
Diniz AFA, Ferreira RC, de Souza ILL, da Silva BA. Ionic Channels as Potential Therapeutic Targets for Erectile Dysfunction: A Review. Front Pharmacol 2020; 11:1120. [PMID: 32848741 PMCID: PMC7396897 DOI: 10.3389/fphar.2020.01120] [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: 05/06/2020] [Accepted: 07/10/2020] [Indexed: 12/12/2022] Open
Abstract
Erectile dysfunction (ED) is a prevalent condition, especially in men over 40 years old, characterized by the inability to obtain and/or maintain penile erection sufficient for satisfactory sexual intercourse. Several psychological and/or organic factors are involved in the etiopathogenesis of ED. In this context, we gathered evidence of the involvement of Large-conductance, Ca2+-activated K+ channels (BKCa), Small-conductance, Ca2+-activated K+ channels (SKCa), KCNQ-encoded voltage-dependent K+ channels (KV7), Transient Receptor Potential channels (TRP), and Calcium-activated Chloride channels (CaCC) dysfunctions on ED. In addition, the use of modulating agents of these channels are involved in relaxation of the cavernous smooth muscle cell and, consequent penile erection, suggesting that these channels are promising therapeutic targets for the treatment of erectile dysfunction.
Collapse
Affiliation(s)
- Anderson Fellyp Avelino Diniz
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Rafael Carlos Ferreira
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Iara Leão Luna de Souza
- Departamento de Ciências Biológicas e da Saúde, Universidade Estadual de Roraima, Boa Vista, Brazil
| | - Bagnólia Araújo da Silva
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, João Pessoa, Brazil
| |
Collapse
|
18
|
Núñez E, Muguruza-Montero A, Villarroel A. Atomistic Insights of Calmodulin Gating of Complete Ion Channels. Int J Mol Sci 2020; 21:ijms21041285. [PMID: 32075037 PMCID: PMC7072864 DOI: 10.3390/ijms21041285] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/07/2020] [Accepted: 02/12/2020] [Indexed: 12/13/2022] Open
Abstract
Intracellular calcium is essential for many physiological processes, from neuronal signaling and exocytosis to muscle contraction and bone formation. Ca2+ signaling from the extracellular medium depends both on membrane potential, especially controlled by ion channels selective to K+, and direct permeation of this cation through specialized channels. Calmodulin (CaM), through direct binding to these proteins, participates in setting the membrane potential and the overall permeability to Ca2+. Over the past years many structures of complete channels in complex with CaM at near atomic resolution have been resolved. In combination with mutagenesis-function, structural information of individual domains and functional studies, different mechanisms employed by CaM to control channel gating are starting to be understood at atomic detail. Here, new insights regarding four types of tetrameric channels with six transmembrane (6TM) architecture, Eag1, SK2/SK4, TRPV5/TRPV6 and KCNQ1–5, and its regulation by CaM are described structurally. Different CaM regions, N-lobe, C-lobe and EF3/EF4-linker play prominent signaling roles in different complexes, emerging the realization of crucial non-canonical interactions between CaM and its target that are only evidenced in the full-channel structure. Different mechanisms to control gating are used, including direct and indirect mechanical actuation over the pore, allosteric control, indirect effect through lipid binding, as well as direct plugging of the pore. Although each CaM lobe engages through apparently similar alpha-helices, they do so using different docking strategies. We discuss how this allows selective action of drugs with great therapeutic potential.
Collapse
|
19
|
Khandai P, Forcelli PA, N'Gouemo P. Activation of small conductance calcium-activated potassium channels suppresses seizure susceptibility in the genetically epilepsy-prone rats. Neuropharmacology 2019; 163:107865. [PMID: 31783064 DOI: 10.1016/j.neuropharm.2019.107865] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/14/2019] [Accepted: 11/25/2019] [Indexed: 12/19/2022]
Abstract
Small conductance calcium-activated potassium (SK) channels dampen neuronal excitability by contributing to slow afterhyperpolarization (AHP) that follows a series of action potentials, and therefore may represent an intrinsic inhibitory mechanism to prevent seizures. We have previously reported that susceptibility to acoustically evoked seizures was associated with downregulation of SK1 and SK3 subtypes of SK channels in the inferior colliculus of the moderated seizure severity strain of the genetically epilepsy-prone rats (GEPR-3s). Here, we evaluated the effects of 1-ethyl-2-benzimidazolinone (1-EBIO), a potent activator of SK channels, on acoustically evoked seizures in both male and female adult GEPR-3s at various time points post-treatment. Systemic administration of 1-EBIO at various tested doses suppressed seizure susceptibility in both male and female GEPR-3s; however, the complete seizure suppression was only observed following administration of relatively higher doses of 1-EBIO in females. These findings indicate that activation of SK channels results in anticonvulsive action against generalized tonic-clonic seizures in both male and female GEPR-3s, with males exhibiting higher sensitivity than females.
Collapse
Affiliation(s)
- Padmini Khandai
- Departments of Pediatrics, Georgetown University Medical Center, Washington, USA
| | - Patrick A Forcelli
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington DC, USA; Department of Neuroscience, Georgetown University Medical Center, Washington DC, USA; Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington DC, USA
| | - Prosper N'Gouemo
- Departments of Pediatrics, Georgetown University Medical Center, Washington, USA; Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington DC, USA; Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington DC, USA.
| |
Collapse
|
20
|
Shim H, Brown BM, Singh L, Singh V, Fettinger JC, Yarov-Yarovoy V, Wulff H. The Trials and Tribulations of Structure Assisted Design of K Ca Channel Activators. Front Pharmacol 2019; 10:972. [PMID: 31616290 PMCID: PMC6764326 DOI: 10.3389/fphar.2019.00972] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 07/29/2019] [Indexed: 11/13/2022] Open
Abstract
Calcium-activated K+ channels constitute attractive targets for the treatment of neurological and cardiovascular diseases. To explain why certain 2-aminobenzothiazole/oxazole-type KCa activators (SKAs) are KCa3.1 selective we previously generated homology models of the C-terminal calmodulin-binding domain (CaM-BD) of KCa3.1 and KCa2.3 in complex with CaM using Rosetta modeling software. We here attempted to employ this atomistic level understanding of KCa activator binding to switch selectivity around and design KCa2.2 selective activators as potential anticonvulsants. In this structure-based drug design approach we used RosettaLigand docking and carefully compared the binding poses of various SKA compounds in the KCa2.2 and KCa3.1 CaM-BD/CaM interface pocket. Based on differences between residues in the KCa2.2 and KCa.3.1 models we virtually designed 168 new SKA compounds. The compounds that were predicted to be both potent and KCa2.2 selective were synthesized, and their activity and selectivity tested by manual or automated electrophysiology. However, we failed to identify any KCa2.2 selective compounds. Based on the full-length KCa3.1 structure it was recently demonstrated that the C-terminal crystal dimer was an artefact and suggested that the "real" binding pocket for the KCa activators is located at the S4-S5 linker. We here confirmed this structural hypothesis through mutagenesis and now offer a new, corrected binding site model for the SKA-type KCa channel activators. SKA-111 (5-methylnaphtho[1,2-d]thiazol-2-amine) is binding in the interface between the CaM N-lobe and the S4-S5 linker where it makes van der Waals contacts with S181 and L185 in the S45A helix of KCa3.1.
Collapse
Affiliation(s)
- Heesung Shim
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States.,Department of Chemistry, University of California, Davis, Davis, CA, United States
| | - Brandon M Brown
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Latika Singh
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Vikrant Singh
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - James C Fettinger
- Department of Chemistry, University of California, Davis, Davis, CA, United States
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
21
|
Kim HJ, Yin MZ, Cho S, Kim SE, Choi SW, Ye SK, Yoo HY, Kim SJ. Increased inward rectifier K + current of coronary artery smooth muscle cells in spontaneously hypertensive rats; partial compensation of the attenuated endothelium-dependent relaxation via Ca 2+ -activated K + channels. Clin Exp Pharmacol Physiol 2019; 47:38-48. [PMID: 31444788 DOI: 10.1111/1440-1681.13168] [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: 06/13/2019] [Revised: 08/06/2019] [Accepted: 08/19/2019] [Indexed: 11/28/2022]
Abstract
Endothelium-dependent vasorelaxation is partly mediated by small-conductance (SK3) and intermediate-conductance Ca2+ -activated K+ channels (SK4) in the endothelium that results in endothelium-dependent hyperpolarization (EDH). Apart from the electrical propagation through myoendothelial gap junctions, the K+ released from the endothelium facilitates EDH by increasing inward rectifier K+ channel (Kir) conductance in smooth muscle cells. The EDH-dependent relaxation of coronary artery (CA) and Kir current in smooth muscle cells (CASMCs) of hypertensive animals are poorly understood despite the critical role of coronary flow in the hypertrophic heart. In spontaneously hypertensive (SHR) and control (WKY) rats, we found attenuation of the CA relaxation by activators of SK3 and SK4 (NS309 and 1-EBIO) in SHR. In isolated CASMCs, whole-cell patch-clamp study revealed larger IKir in SHR than WKY, whereas the myocytes of skeletal and cerebral arteries showed smaller IKir in SHR than WKY. While the treatment with IKir inhibitor (0.1 mmol/L Ba2+ ) alone did not affect the WKY-CA, the SHR-CA showed significant contractile response, suggesting relaxing influence of the higher IK ir in the CASMCs of SHR. Furthermore, the attenuation of NS309-induced relaxation of CA by the combined treatment with 0.1 mmol/L Ba2+ was more prominent in SHR than WKY. Our study firstly shows a distinct increase of IK ir in the CASMCs of SHR, which could partly compensate for the attenuated relaxation via endothelial SK3 and SK4.
Collapse
Affiliation(s)
- Hae Jin Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Ming Zhe Yin
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Suhan Cho
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Sung Eun Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Seong Woo Choi
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Sang Kyu Ye
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea
| | - Hae Young Yoo
- Chung-Ang University Red Cross College of Nursing, Seoul, Korea
| | - Sung Joon Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
22
|
Brown BM, Shim H, Christophersen P, Wulff H. Pharmacology of Small- and Intermediate-Conductance Calcium-Activated Potassium Channels. Annu Rev Pharmacol Toxicol 2019; 60:219-240. [PMID: 31337271 DOI: 10.1146/annurev-pharmtox-010919-023420] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The three small-conductance calcium-activated potassium (KCa2) channels and the related intermediate-conductance KCa3.1 channel are voltage-independent K+ channels that mediate calcium-induced membrane hyperpolarization. When intracellular calcium increases in the channel vicinity, it calcifies the flexible N lobe of the channel-bound calmodulin, which then swings over to the S4-S5 linker and opens the channel. KCa2 and KCa3.1 channels are highly druggable and offer multiple binding sites for venom peptides and small-molecule blockers as well as for positive- and negative-gating modulators. In this review, we briefly summarize the physiological role of KCa channels and then discuss the pharmacophores and the mechanism of action of the most commonly used peptidic and small-molecule KCa2 and KCa3.1 modulators. Finally, we describe the progress that has been made in advancing KCa3.1 blockers and KCa2.2 negative- and positive-gating modulators toward the clinic for neurological and cardiovascular diseases and discuss the remaining challenges.
Collapse
Affiliation(s)
- Brandon M Brown
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| | - Heesung Shim
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| | | | - Heike Wulff
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| |
Collapse
|
23
|
Rational modulator design by exploitation of protein-protein complex structures. Future Med Chem 2019; 11:1015-1033. [PMID: 31141413 DOI: 10.4155/fmc-2018-0433] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The horizon of drug discovery is currently expanding to target and modulate protein-protein interactions (PPIs) in globular proteins and intrinsically disordered proteins that are involved in various diseases. To either interrupt or stabilize PPIs, the 3D structure of target protein-protein (or protein-peptide) complexes can be exploited to rationally design PPI modulators (inhibitors or stabilizers) through structure-based molecular design. In this review, we present an overview of experimental and computational methods that can be used to determine 3D structures of protein-protein complexes. Several approaches including rational and in silico methods that can be applied to design peptides, peptidomimetics and small compounds by utilization of determined 3D protein-protein/peptide complexes are summarized and illustrated.
Collapse
|
24
|
Sakamoto K. [A Cellular Pharmacological Approach to the Development of Drugs to Treat Muscle Wasting]. YAKUGAKU ZASSHI 2018; 138:1271-1275. [PMID: 30270271 DOI: 10.1248/yakushi.18-00091-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Skeletal muscle atrophy reduces quality of life and increases mortality. However, there are few available drugs for the treatment of muscle atrophy. Recently, cell signaling pathways involved in skeletal muscle atrophy or hypertrophy have been determined. To develop drugs for skeletal muscle atrophy, we have studied compounds which modulate pathways of myogenic differentiation, a pivotal step for the maintenance of skeletal muscle mass. First, we examined a K+ channel opener on myogenic differentiation, since hyperpolarization is a trigger for skeletal muscle differentiation. 5,6-Dichloro-1-ethyl-1,3-dihydro-2H-benzimidazol-2-one (DCEBIO), an opener of the small/intermediate conductance Ca2+ activated K+ (SKCa/IKCa) channels, increases myogenic differentiation in C2C12 mouse skeletal myoblasts. This effect was inhibited by TRAM-34, an IKCa channel blocker. This suggests that K+ channels in skeletal muscle stem cells are potential targets for an anti-muscle atrophy drug. Next, we searched for drugs which prevent sepsis-induced muscle atrophy. Lipopolysaccharide (LPS), an inducer of sepsis, attenuates myogenic differentiation in C2C12 myoblasts. LPS also increases the protein expression of myostatin and activates NFκB during differentiation. The TLR4 signal inhibitor TAK-242, and an anti-TNFα neutralizing antibody, reduce these inflammatory responses. Our data suggest that LPS inhibits myogenic differentiation via the NFκB/TNFα pathway. This pathway may be involved in the development of muscle wasting caused by sepsis.
Collapse
Affiliation(s)
- Kazuho Sakamoto
- Department of Pharmacology, School of Medicine, Fukushima Medical University
| |
Collapse
|
25
|
Li Z, Tremble SM, Cipolla MJ. Implications for understanding ischemic stroke as a sexually dimorphic disease: the role of pial collateral circulations. Am J Physiol Heart Circ Physiol 2018; 315:H1703-H1712. [PMID: 30239233 DOI: 10.1152/ajpheart.00402.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We investigated structural and functional differences in primary and pial collateral circulations in adult normotensive male and female Wistar rats. Male ( n = 10) and female ( n = 7) rats were subjected to middle cerebral artery (MCA) occlusion and changes in relative cerebral blood flow in MCA and pial collateral territories were measured by multisite laser-Doppler flowmetry. Rats were then transcardially perfused with a mixture of carbon black and latex, perfusion fixed, and imaged to compare primary and pial collateral structure between male ( n = 4) and female ( n = 3) rats, including lumen diameters and number. To study pial collateral function, leptomeningeal anastomoses (LMAs) were isolated and pressurized from male ( n = 7) and female ( n = 6) rats. Myogenic tone and reactivity to pressure, vascular function to pharmacological activator, or inhibitor of ion channels was measured and compared. There was no difference between relative cerebral blood flow in both MCA and pial collateral territories during occlusion and reperfusion between groups. Compared with male LMAs, female LMAs had similar myogenic tone (24.0 ± 7.3% vs. 16.0 ± 3.7%, P > 0.05) and reactivity to increased pressure and similar vascular responses to vasoconstrictive and vasodilatory stimuli. Additionally, compared with female LMAs, male LMAs had similar numbers (21 ± 1 vs. 20 ± 2, P > 0.05) and diameters (30.5 ± 2.0 vs. 26.2 ± 0.6 μm, P > 0.05), and no sex difference was detected in the diameter of arterial segments of circle of Willis. Together, our data establish no sex difference of cerebral collateral structure or function, suggesting that the reduced severity of stroke outcome in female rats is not likely due to differences in the cerebral collateral circulation. NEW & NOTEWORTHY Our work compared the function of leptomeningeal anastomoses between male and female adult normotensive rats with no sex difference found. We also confirmed no sex difference in primary and pial collateral structure in Wistar rats. Our findings suggest that the reduced severity of stroke in premenopausal women and reproductively intact female rodents is not likely due to improved primary and pial collateral circulations.
Collapse
Affiliation(s)
- Zhaojin Li
- Department of Neurological Sciences, University of Vermont Robert Larner College of Medicine , Burlington, Vermont
| | - Sarah M Tremble
- Department of Neurological Sciences, University of Vermont Robert Larner College of Medicine , Burlington, Vermont
| | - Marilyn J Cipolla
- Department of Neurological Sciences, University of Vermont Robert Larner College of Medicine , Burlington, Vermont.,Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Vermont Robert Larner College of Medicine , Burlington, Vermont.,Department of Pharmacology, University of Vermont Robert Larner College of Medicine , Burlington, Vermont
| |
Collapse
|
26
|
Kshatri AS, Gonzalez-Hernandez A, Giraldez T. Physiological Roles and Therapeutic Potential of Ca 2+ Activated Potassium Channels in the Nervous System. Front Mol Neurosci 2018; 11:258. [PMID: 30104956 PMCID: PMC6077210 DOI: 10.3389/fnmol.2018.00258] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/06/2018] [Indexed: 12/21/2022] Open
Abstract
Within the potassium ion channel family, calcium activated potassium (KCa) channels are unique in their ability to couple intracellular Ca2+ signals to membrane potential variations. KCa channels are diversely distributed throughout the central nervous system and play fundamental roles ranging from regulating neuronal excitability to controlling neurotransmitter release. The physiological versatility of KCa channels is enhanced by alternative splicing and co-assembly with auxiliary subunits, leading to fundamental differences in distribution, subunit composition and pharmacological profiles. Thus, understanding specific KCa channels’ mechanisms in neuronal function is challenging. Based on their single channel conductance, KCa channels are divided into three subtypes: small (SK, 4–14 pS), intermediate (IK, 32–39 pS) and big potassium (BK, 200–300 pS) channels. This review describes the biophysical characteristics of these KCa channels, as well as their physiological roles and pathological implications. In addition, we also discuss the current pharmacological strategies and challenges to target KCa channels for the treatment of various neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Aravind S Kshatri
- Department of Basic Medical Sciences, Medical School, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologias Biomedicas, Universidad de La Laguna, Tenerife, Spain
| | - Alberto Gonzalez-Hernandez
- Department of Basic Medical Sciences, Medical School, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologias Biomedicas, Universidad de La Laguna, Tenerife, Spain
| | - Teresa Giraldez
- Department of Basic Medical Sciences, Medical School, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologias Biomedicas, Universidad de La Laguna, Tenerife, Spain
| |
Collapse
|
27
|
Lee CH, MacKinnon R. Activation mechanism of a human SK-calmodulin channel complex elucidated by cryo-EM structures. Science 2018; 360:508-513. [PMID: 29724949 DOI: 10.1126/science.aas9466] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 03/16/2018] [Indexed: 12/17/2022]
Abstract
Small-conductance Ca2+-activated K+ (SK) channels mediate neuron excitability and are associated with synaptic transmission and plasticity. They also regulate immune responses and the size of blood cells. Activation of SK channels requires calmodulin (CaM), but how CaM binds and opens SK channels has been unclear. Here we report cryo-electron microscopy (cryo-EM) structures of a human SK4-CaM channel complex in closed and activated states at 3.4- and 3.5-angstrom resolution, respectively. Four CaM molecules bind to one channel tetramer. Each lobe of CaM serves a distinct function: The C-lobe binds to the channel constitutively, whereas the N-lobe interacts with the S4-S5 linker in a Ca2+-dependent manner. The S4-S5 linker, which contains two distinct helices, undergoes conformational changes upon CaM binding to open the channel pore. These structures reveal the gating mechanism of SK channels and provide a basis for understanding SK channel pharmacology.
Collapse
Affiliation(s)
- Chia-Hsueh Lee
- Laboratory of Molecular Neurobiology and Biophysics, The Rockefeller University, Howard Hughes Medical Institute, 1230 York Avenue, New York, NY 10065, USA
| | - Roderick MacKinnon
- Laboratory of Molecular Neurobiology and Biophysics, The Rockefeller University, Howard Hughes Medical Institute, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
28
|
A V-to-F substitution in SK2 channels causes Ca 2+ hypersensitivity and improves locomotion in a C. elegans ALS model. Sci Rep 2018; 8:10749. [PMID: 30013223 PMCID: PMC6048120 DOI: 10.1038/s41598-018-28783-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 06/29/2018] [Indexed: 11/09/2022] Open
Abstract
Small-conductance Ca2+-activated K+ (SK) channels mediate medium afterhyperpolarization in the neurons and play a key role in the regulation of neuronal excitability. SK channels are potential drug targets for ataxia and Amyotrophic Lateral Sclerosis (ALS). SK channels are activated exclusively by the Ca2+-bound calmodulin. Previously, we identified an intrinsically disordered fragment that is essential for the mechanical coupling between Ca2+/calmodulin binding and channel opening. Here, we report that substitution of a valine to phenylalanine (V407F) in the intrinsically disordered fragment caused a ~6 fold increase in the Ca2+ sensitivity of SK2-a channels. This substitution resulted in a novel interaction between the ectopic phenylalanine and M411, which stabilized PIP2-interacting residue K405, and subsequently enhanced Ca2+ sensitivity. Also, equivalent valine to phenylalanine substitutions in SK1 or SK3 channels conferred Ca2+ hypersensitivity. An equivalent phenylalanine substitution in the Caenorhabditis elegans (C. elegans) SK2 ortholog kcnl-2 partially rescued locomotion defects in an existing C. elegans ALS model, in which human SOD1G85R is expressed at high levels in neurons, confirming that this phenylalanine substitution impacts channel function in vivo. This work for the first time provides a critical reagent for future studies: an SK channel that is hypersensitive to Ca2+ with increased activity in vivo.
Collapse
|
29
|
Skarsfeldt MA, Bomholtz SH, Lundegaard PR, Lopez-Izquierdo A, Tristani-Firouzi M, Bentzen BH. Atrium-specific ion channels in the zebrafish-A role of I KACh in atrial repolarization. Acta Physiol (Oxf) 2018; 223:e13049. [PMID: 29412518 DOI: 10.1111/apha.13049] [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: 10/04/2017] [Revised: 01/26/2018] [Accepted: 01/29/2018] [Indexed: 12/16/2022]
Abstract
AIM The zebrafish has emerged as a novel model for investigating cardiac physiology and pathology. The aim of this study was to investigate the atrium-specific ion channels responsible for shaping the atrial cardiac action potential in zebrafish. METHODS Using quantitative polymerase chain reaction, we assessed the expression level of atrium-specific potassium channels. The functional role of these channels was studied by patch clamp experiments on isolated atrial and ventricular cardiomyocytes and by optical mapping of explanted adult zebrafish hearts. Finally, surface ECGs were recorded to establish possible in vivo roles of atrial ion channels. RESULTS In isolated adult zebrafish hearts, we identified the expression of kcnk3, kcnk9, kcnn1, kcnn2, kcnn3, kcnj3 and kcnj5, the genes that encode the atrium-specific K2P , KCa 2.x and Kir 3.1/4 (KACh ) ion channels. The electrophysiological data indicate that the acetylcholine-activated inward-rectifying current, IKACh, plays a major role in the zebrafish atrium, whereas K2P 3.1/9.1 and KCa 2.x channels do not appear to be involved in regulating the action potential in the zebrafish heart. CONCLUSION We demonstrate that the acetylcholine-activated inward-rectifying current (IKACh ) current plays a major role in the zebrafish atrium and that the zebrafish could potentially be a cost-effective and reliable model for pharmacological testing of atrium-specific IKACh modulating compounds.
Collapse
Affiliation(s)
- M. A. Skarsfeldt
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen N Denmark
- Nora Eccles Harrison Cardiovascular Research and Training Institute; University of Utah; Salt Lake City UT USA
| | - S. H. Bomholtz
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen N Denmark
| | - P. R. Lundegaard
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen N Denmark
| | - A. Lopez-Izquierdo
- Nora Eccles Harrison Cardiovascular Research and Training Institute; University of Utah; Salt Lake City UT USA
| | - M. Tristani-Firouzi
- Nora Eccles Harrison Cardiovascular Research and Training Institute; University of Utah; Salt Lake City UT USA
| | - B. H. Bentzen
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen N Denmark
| |
Collapse
|
30
|
Cho LTY, Alexandrou AJ, Torella R, Knafels J, Hobbs J, Taylor T, Loucif A, Konopacka A, Bell S, Stevens EB, Pandit J, Horst R, Withka JM, Pryde DC, Liu S, Young GT. An Intracellular Allosteric Modulator Binding Pocket in SK2 Ion Channels Is Shared by Multiple Chemotypes. Structure 2018; 26:533-544.e3. [PMID: 29576321 DOI: 10.1016/j.str.2018.02.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/03/2017] [Accepted: 02/23/2018] [Indexed: 01/22/2023]
Abstract
Small conductance potassium (SK) ion channels define neuronal firing rates by conducting the after-hyperpolarization current. They are key targets in developing therapies where neuronal firing rates are dysfunctional, such as in epilepsy, Parkinson's, and amyotrophic lateral sclerosis (ALS). Here, we characterize a binding pocket situated at the intracellular interface of SK2 and calmodulin, which we show to be shared by multiple small-molecule chemotypes. Crystallization of this complex revealed that riluzole (approved for ALS) and an analog of the anti-ataxic agent (4-chloro-phenyl)-[2-(3,5-dimethyl-pyrazol-1-yl)-pyrimidin-4-yl]-amine (CyPPA) bind to and allosterically modulate via this site. Solution-state nuclear magnetic resonance demonstrates that riluzole, NS309, and CyPPA analogs bind at this bipartite pocket. We demonstrate, by patch-clamp electrophysiology, that both classes of ligand interact with overlapping but distinct residues within this pocket. These data define a clinically important site, laying the foundations for further studies of the mechanism of action of riluzole and related molecules.
Collapse
Affiliation(s)
- Lily T-Y Cho
- Pfizer Neuroscience and Pain Research Unit, Granta Park, Great Abington, Cambridge CB21 6GS, UK
| | - Aristos J Alexandrou
- Pfizer Neuroscience and Pain Research Unit, Granta Park, Great Abington, Cambridge CB21 6GS, UK
| | - Rubben Torella
- Pfizer Worldwide Medicinal Chemistry, Neuroscience and Pain Research Unit, Granta Park, Great Abington, Cambridge CB21 6GS, UK
| | - John Knafels
- Pfizer Structural Biology and Biophysics, Groton, Eastern Point Road, Groton, CT 06340, USA
| | - Jake Hobbs
- Pfizer Neuroscience and Pain Research Unit, Granta Park, Great Abington, Cambridge CB21 6GS, UK
| | - Toni Taylor
- Pfizer Neuroscience and Pain Research Unit, Granta Park, Great Abington, Cambridge CB21 6GS, UK
| | - Alex Loucif
- Pfizer Neuroscience and Pain Research Unit, Granta Park, Great Abington, Cambridge CB21 6GS, UK
| | - Agnieszka Konopacka
- Pfizer Neuroscience and Pain Research Unit, Granta Park, Great Abington, Cambridge CB21 6GS, UK
| | - Sigourney Bell
- Pfizer Neuroscience and Pain Research Unit, Granta Park, Great Abington, Cambridge CB21 6GS, UK
| | - Edward B Stevens
- Pfizer Neuroscience and Pain Research Unit, Granta Park, Great Abington, Cambridge CB21 6GS, UK
| | - Jay Pandit
- Pfizer Structural Biology and Biophysics, Groton, Eastern Point Road, Groton, CT 06340, USA
| | - Reto Horst
- Pfizer Structural Biology and Biophysics, Groton, Eastern Point Road, Groton, CT 06340, USA
| | - Jane M Withka
- Pfizer Structural Biology and Biophysics, Groton, Eastern Point Road, Groton, CT 06340, USA
| | - David C Pryde
- Pfizer Worldwide Medicinal Chemistry, Neuroscience and Pain Research Unit, Granta Park, Great Abington, Cambridge CB21 6GS, UK
| | - Shenping Liu
- Pfizer Structural Biology and Biophysics, Groton, Eastern Point Road, Groton, CT 06340, USA.
| | - Gareth T Young
- Pfizer Neuroscience and Pain Research Unit, Granta Park, Great Abington, Cambridge CB21 6GS, UK.
| |
Collapse
|
31
|
Mathew John C, Khaddaj Mallat R, George G, Kim T, Mishra RC, Braun AP. Pharmacologic targeting of endothelial Ca 2+-activated K + channels: A strategy to improve cardiovascular function. Channels (Austin) 2018; 12:126-136. [PMID: 29577810 PMCID: PMC5972810 DOI: 10.1080/19336950.2018.1454814] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 03/15/2018] [Indexed: 12/17/2022] Open
Abstract
Endothelial small and intermediate-conductance, Ca2+-activated K+ channels (KCa2.3 and KCa3.1, respectively) play an important role in the regulation of vascular function and systemic blood pressure. Growing evidence indicates that they are intimately involved in agonist-evoked vasodilation of small resistance arteries throughout the circulation. Small molecule activators of KCa2.x and 3.1 channels, such as SKA-31, can acutely inhibit myogenic tone in isolated resistance arteries, induce effective vasodilation in intact vascular beds, such as the coronary circulation, and acutely decrease systemic blood pressure in vivo. The blood pressure-lowering effect of SKA-31, and early indications of improvement in endothelial dysfunction suggest that endothelial KCa channel activators could eventually be developed into a new class of endothelial targeted agents to combat hypertension or atherosclerosis. This review summarises recent insights into the activation of endothelial Ca2+ activated K+ channels in various vascular beds, and how tools, such as SKA-31, may be beneficial in disease-related conditions.
Collapse
Affiliation(s)
- Cini Mathew John
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rayan Khaddaj Mallat
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Grace George
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Taeyeob Kim
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ramesh C. Mishra
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Andrew P. Braun
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
32
|
Nam YW, Orfali R, Liu T, Yu K, Cui M, Wulff H, Zhang M. Structural insights into the potency of SK channel positive modulators. Sci Rep 2017; 7:17178. [PMID: 29214998 PMCID: PMC5719431 DOI: 10.1038/s41598-017-16607-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 11/15/2017] [Indexed: 12/26/2022] Open
Abstract
Small-conductance Ca2+-activated K+ (SK) channels play essential roles in the regulation of cellular excitability and have been implicated in neurological and cardiovascular diseases through both animal model studies and human genetic association studies. Over the past two decades, positive modulators of SK channels such as NS309 and 1-EBIO have been developed. Our previous structural studies have identified the binding pocket of 1-EBIO and NS309 that is located at the interface between the channel and calmodulin. In this study, we took advantage of four compounds with potencies varying over three orders of magnitude, including 1-EBIO, NS309, SKS-11 (6-bromo-5-methyl-1H-indole-2,3-dione-3-oxime) and SKS-14 (7-fluoro-3-(hydroxyimino)indolin-2-one). A combination of x-ray crystallographic, computational and electrophysiological approaches was utilized to investigate the interactions between the positive modulators and their binding pocket. A strong trend exists between the interaction energy of the compounds within their binding site calculated from the crystal structures, and the potency of these compounds in potentiating the SK2 channel current determined by electrophysiological recordings. Our results further reveal that the difference in potency of the positive modulators in potentiating SK2 channel activity may be attributed primarily to specific electrostatic interactions between the modulators and their binding pocket.
Collapse
Affiliation(s)
- Young-Woo Nam
- Department of Biomedical and Pharmaceutical Sciences & Structural Biology Research Center, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Razan Orfali
- Department of Biomedical and Pharmaceutical Sciences & Structural Biology Research Center, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Tingting Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kunqian Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Meng Cui
- Department of Pharmaceutical Sciences, Northeastern University School of Pharmacy, Boston, MA, 02115, USA
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California, Davis, CA, 95616, USA
| | - Miao Zhang
- Department of Biomedical and Pharmaceutical Sciences & Structural Biology Research Center, Chapman University School of Pharmacy, Irvine, CA, 92618, USA.
| |
Collapse
|
33
|
Affiliation(s)
- Brandon M Brown
- a Department of Pharmacology , University of California, Davis , Davis , CA , USA
| | - Heesung Shim
- a Department of Pharmacology , University of California, Davis , Davis , CA , USA
| | - Heike Wulff
- a Department of Pharmacology , University of California, Davis , Davis , CA , USA
| |
Collapse
|
34
|
Brown BM, Shim H, Zhang M, Yarov-Yarovoy V, Wulff H. Structural Determinants for the Selectivity of the Positive KCa3.1 Gating Modulator 5-Methylnaphtho[2,1- d]oxazol-2-amine (SKA-121). Mol Pharmacol 2017; 92:469-480. [PMID: 28760780 DOI: 10.1124/mol.117.109421] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/27/2017] [Indexed: 12/20/2022] Open
Abstract
Intermediate-conductance (KCa3.1) and small-conductance (KCa2) calcium-activated K+ channels are gated by calcium binding to calmodulin (CaM) molecules associated with the calmodulin-binding domain (CaM-BD) of these channels. The existing KCa activators, such as naphtho[1,2-d]thiazol-2-ylamine (SKA-31), 6,7-dichloro-1H-indole-2,3-dione 3-oxime (NS309), and 1-ethylbenzimidazolin-2-one (EBIO), activate both channel types with similar potencies. In a previous chemistry effort, we optimized the benzothiazole pharmacophore of SKA-31 toward KCa3.1 selectivity and identified 5-methylnaphtho[2,1-d]oxazol-2-amine (SKA-121), which exhibits 40-fold selectivity for KCa3.1 over KCa2.3. To understand why introduction of a single CH3 group in five-position of the benzothiazole/oxazole system could achieve such a gain in selectivity for KCa3.1 over KCa2.3, we first localized the binding site of the benzothiazoles/oxazoles to the CaM-BD/CaM interface and then used computational modeling software to generate models of the KCa3.1 and KCa2.3 CaM-BD/CaM complexes with SKA-121. Based on a combination of mutagenesis and structural modeling, we suggest that all benzothiazole/oxazole-type KCa activators bind relatively "deep" in the CaM-BD/CaM interface and hydrogen bond with E54 on CaM. In KCa3.1, SKA-121 forms an additional hydrogen bond network with R362. In contrast, NS309 sits more "forward" and directly hydrogen bonds with R362 in KCa3.1. Mutating R362 to serine, the corresponding residue in KCa2.3 reduces the potency of SKA-121 by 7-fold, suggesting that R362 is responsible for the generally greater potency of KCa activators on KCa3.1. The increase in SKA-121's KCa3.1 selectivity compared with its parent, SKA-31, seems to be due to better overall shape complementarity and hydrophobic interactions with S372 and M368 on KCa3.1 and M72 on CaM at the KCa3.1-CaM-BD/CaM interface.
Collapse
Affiliation(s)
- Brandon M Brown
- Department of Pharmacology (B.M.B., H.S., H.W.), Department of Physiology and Membrane Biology (V.Y.-Y.), School of Medicine, and Department of Chemistry (H.S.), University of California, Davis, California; and Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (M.Z.)
| | - Heesung Shim
- Department of Pharmacology (B.M.B., H.S., H.W.), Department of Physiology and Membrane Biology (V.Y.-Y.), School of Medicine, and Department of Chemistry (H.S.), University of California, Davis, California; and Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (M.Z.)
| | - Miao Zhang
- Department of Pharmacology (B.M.B., H.S., H.W.), Department of Physiology and Membrane Biology (V.Y.-Y.), School of Medicine, and Department of Chemistry (H.S.), University of California, Davis, California; and Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (M.Z.)
| | - Vladimir Yarov-Yarovoy
- Department of Pharmacology (B.M.B., H.S., H.W.), Department of Physiology and Membrane Biology (V.Y.-Y.), School of Medicine, and Department of Chemistry (H.S.), University of California, Davis, California; and Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (M.Z.)
| | - Heike Wulff
- Department of Pharmacology (B.M.B., H.S., H.W.), Department of Physiology and Membrane Biology (V.Y.-Y.), School of Medicine, and Department of Chemistry (H.S.), University of California, Davis, California; and Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (M.Z.)
| |
Collapse
|
35
|
Andrei SA, Sijbesma E, Hann M, Davis J, O’Mahony G, Perry MWD, Karawajczyk A, Eickhoff J, Brunsveld L, Doveston RG, Milroy LG, Ottmann C. Stabilization of protein-protein interactions in drug discovery. Expert Opin Drug Discov 2017; 12:925-940. [DOI: 10.1080/17460441.2017.1346608] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Sebastian A. Andrei
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Eline Sijbesma
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Michael Hann
- Platform Technology and Science, Medicines Research Centre, GlaxoSmithKline R&D, Stevenage, UK
| | - Jeremy Davis
- Department of Chemistry, UCB Celltech, Slough, UK
| | - Gavin O’Mahony
- CVMD Medicinal Chemistry, Innovative Medicines and Early Development, AstraZeneca Gothenburg, Pepparedsleden, Mölndal, Sweden
| | - Matthew W. D. Perry
- RIA Medicinal Chemistry, Innovative Medicines and Early Development, AstraZeneca Gothenburg, Pepparedsleden, Mölndal, Sweden
| | - Anna Karawajczyk
- Medicinal Chemistry, Taros Chemicals GmbH & Co. KG, Dortmund, Germany
| | - Jan Eickhoff
- Assay development & screening, Lead Discovery Center GmbH, Dortmund, Germany
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Richard G. Doveston
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Lech-Gustav Milroy
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
- Department of Chemistry, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
36
|
Kaczmarek LK, Aldrich RW, Chandy KG, Grissmer S, Wei AD, Wulff H. International Union of Basic and Clinical Pharmacology. C. Nomenclature and Properties of Calcium-Activated and Sodium-Activated Potassium Channels. Pharmacol Rev 2017; 69:1-11. [PMID: 28267675 PMCID: PMC11060434 DOI: 10.1124/pr.116.012864] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024] Open
Abstract
A subset of potassium channels is regulated primarily by changes in the cytoplasmic concentration of ions, including calcium, sodium, chloride, and protons. The eight members of this subfamily were originally all designated as calcium-activated channels. More recent studies have clarified the gating mechanisms for these channels and have documented that not all members are sensitive to calcium. This article describes the molecular relationships between these channels and provides an introduction to their functional properties. It also introduces a new nomenclature that differentiates between calcium- and sodium-activated potassium channels.
Collapse
Affiliation(s)
- Leonard K Kaczmarek
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| | - Richard W Aldrich
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| | - K George Chandy
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| | - Stephan Grissmer
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| | - Aguan D Wei
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| | - Heike Wulff
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| |
Collapse
|
37
|
Köhler R, Oliván-Viguera A, Wulff H. Endothelial Small- and Intermediate-Conductance K Channels and Endothelium-Dependent Hyperpolarization as Drug Targets in Cardiovascular Disease. ADVANCES IN PHARMACOLOGY 2016; 77:65-104. [DOI: 10.1016/bs.apha.2016.04.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
38
|
Abstract
Modulation of protein-protein interactions (PPIs) is becoming increasingly important in drug discovery and chemical biology. While a few years ago this 'target class' was deemed to be largely undruggable an impressing number of publications and success stories now show that targeting PPIs with small, drug-like molecules indeed is a feasible approach. Here, we summarize the current state of small-molecule inhibition and stabilization of PPIs and review the active molecules from a structural and medicinal chemistry angle, especially focusing on the key examples of iNOS, LFA-1 and 14-3-3.
Collapse
|
39
|
Zarzycka B, Kuenemann MA, Miteva MA, Nicolaes GAF, Vriend G, Sperandio O. Stabilization of protein-protein interaction complexes through small molecules. Drug Discov Today 2015; 21:48-57. [PMID: 26434617 DOI: 10.1016/j.drudis.2015.09.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 09/09/2015] [Accepted: 09/25/2015] [Indexed: 12/17/2022]
Abstract
Most of the small molecules that have been identified thus far to modulate protein-protein interactions (PPIs) are inhibitors. Another promising way to interfere with PPI-associated biological processes is to promote PPI stabilization. Even though PPI stabilizers are still scarce, stabilization of PPIs by small molecules is gaining momentum and offers new pharmacological options. Therefore, we have performed a literature survey of PPI stabilization using small molecules. From this, we propose a classification of PPI stabilizers based on their binding mode and the architecture of the complex to facilitate the structure-based design of stabilizers.
Collapse
Affiliation(s)
- Barbara Zarzycka
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Mélaine A Kuenemann
- Université Paris Diderot, Sorbonne Paris Cité, UMRS 973 Inserm, Paris 75013, France; Inserm, U973, Paris 75013, France
| | - Maria A Miteva
- Université Paris Diderot, Sorbonne Paris Cité, UMRS 973 Inserm, Paris 75013, France; Inserm, U973, Paris 75013, France
| | - Gerry A F Nicolaes
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Gert Vriend
- Centre for Molecular and Biomolecular Informatics (CMBI), Radboudumc, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Olivier Sperandio
- Université Paris Diderot, Sorbonne Paris Cité, UMRS 973 Inserm, Paris 75013, France; Inserm, U973, Paris 75013, France; Faculté de Pharmacie, CDithem, 1 rue du Prof. Laguesse, 59000 Lille, France.
| |
Collapse
|
40
|
Increased Basolateral Amygdala Pyramidal Cell Excitability May Contribute to the Anxiogenic Phenotype Induced by Chronic Early-Life Stress. J Neurosci 2015; 35:9730-40. [PMID: 26134655 DOI: 10.1523/jneurosci.0384-15.2015] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
UNLABELLED Adolescence represents a particularly vulnerable period during which exposure to stressors can precipitate the onset of psychiatric disorders and addiction. The basolateral amygdala (BLA) plays an integral role in the pathophysiology of anxiety and addiction. Acute and chronic stress promote increases in BLA pyramidal cell firing, and decreasing BLA excitability alleviates anxiety measures in humans and rodents. Notably, the impact of early-life stress on the mechanisms that govern BLA excitability is unknown. To address this gap in our knowledge, we used a rodent model of chronic early-life stress that engenders robust and enduring increases in anxiety-like behaviors and ethanol intake and examined the impact of this model on the intrinsic excitability of BLA pyramidal cells. Adolescent social isolation was associated with a significant increase in the intrinsic excitability of BLA pyramidal cells and a blunting of the medium component of the afterhyperpolarization potential, a voltage signature of calcium-activated potassium (Kca) channel activity. Western blot analysis revealed reduced expression of small-conductance Kca (SK) channel protein in the BLA of socially isolated (SI) rats. Bath application of a positive SK channel modulator (1-EBIO) normalized firing in ex vivo recordings from SI rats, and in vivo intra-BLA 1-EBIO infusion reduced anxiety-like behaviors. These findings reveal that chronic adolescent stress impairs SK channel function, which contributes to an increase in BLA pyramidal cell excitability and highlights BLA SK channels as promising targets for the treatment of anxiety disorders and comorbid addiction. SIGNIFICANCE STATEMENT Although anxiety disorders and alcohol addiction frequently co-occur, the mechanisms that contribute to this comorbidity are poorly understood. Here, we used a rodent early-life stress model that leads to robust and longlasting increases in behaviors associated with elevated risk of anxiety disorders and addiction to identify novel neurobiological substrates that may underlie these behaviors. Our studies focused on the primary output neurons of the basolateral amygdala, a brain region that plays a key role in anxiety and addiction. We discovered that early-life stress decreases the activity of a specific class of potassium channels and increases the intrinsic excitability of BLA neurons and present evidence that enhancing the function of these channels normalizes BLA excitability and attenuates anxiety-like behaviors.
Collapse
|
41
|
Christophersen P, Wulff H. Pharmacological gating modulation of small- and intermediate-conductance Ca(2+)-activated K(+) channels (KCa2.x and KCa3.1). Channels (Austin) 2015. [PMID: 26217968 DOI: 10.1080/19336950.2015.1071748] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
This short review discusses pharmacological modulation of the opening/closing properties (gating) of small- and intermediate-conductance Ca(2+)-activated K(+) channels (KCa2 and KCa3.1) with special focus on mechanisms-of-action, selectivity, binding sites, and therapeutic potentials. Despite KCa channel gating-modulation being a relatively novel field in drug discovery, efforts in this area have already revealed a surprising plethora of pharmacological sites-of-actions and channel subtype selectivity exerted by different chemical classes. The currently published positive modulators show that such molecules are potentially useful for the treatment of various neurodegenerative disorders such as ataxia, alcohol dependence, and epilepsy as well as hypertension. The negative KCa2 modulators are very effective agents for atrial fibrillation. The prediction is that further unraveling of the molecular details of gating pharmacology will allow for the design of even more potent and subtype selective KCa modulators entering into drug development for these indications.
Collapse
Affiliation(s)
| | - Heike Wulff
- b Department of Pharmacology ; University of California, Davis ; Davis , CA USA
| |
Collapse
|
42
|
Zhang M, Meng XY, Zhang JF, Cui M, Logothetis DE. Molecular overlap in the regulation of SK channels by small molecules and phosphoinositides. SCIENCE ADVANCES 2015; 1:e1500008. [PMID: 26366439 PMCID: PMC4563807 DOI: 10.1126/sciadv.1500008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 05/29/2015] [Indexed: 06/05/2023]
Abstract
Phosphatidylinositol 4,5-bisphosphate (PIP2) directly interacts with the small-conductance Ca2+-activated K+ 2-a (SK2-a) channel/calmodulin complex, serving as a critical element in the regulation of channel activity. We report that changes of protein conformation in close proximity to the PIP2 binding site induced by a small-molecule SK channel modulator, NS309, can effectively enhance the interaction between the protein and PIP2 to potentiate channel activity. This novel modulation of PIP2 sensitivity by small-molecule drugs is likely not to be limited in its application to SK channels, representing an intriguing strategy to develop drugs controlling the activity of the large number of PIP2-dependent proteins.
Collapse
Affiliation(s)
- Miao Zhang
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, 1101 East Marshall Street, Richmond, VA 23298, USA
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, 9401 Jeronimo Road, Irvine, CA 92618, USA
| | - Xuan-Yu Meng
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, 1101 East Marshall Street, Richmond, VA 23298, USA
| | - Ji-fang Zhang
- Department of Molecular Physiology and Biophysics, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA
- Farber Institute for Neurosciences and Graduate Program in Neuroscience, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA
| | - Meng Cui
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, 1101 East Marshall Street, Richmond, VA 23298, USA
| | - Diomedes E. Logothetis
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, 1101 East Marshall Street, Richmond, VA 23298, USA
| |
Collapse
|
43
|
Oliván-Viguera A, Valero MS, Coleman N, Brown BM, Laría C, Murillo MD, Gálvez JA, Díaz-de-Villegas MD, Wulff H, Badorrey R, Köhler R. A novel pan-negative-gating modulator of KCa2/3 channels, fluoro-di-benzoate, RA-2, inhibits endothelium-derived hyperpolarization-type relaxation in coronary artery and produces bradycardia in vivo. Mol Pharmacol 2014; 87:338-48. [PMID: 25468883 DOI: 10.1124/mol.114.095745] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Small/intermediate conductance KCa channels (KCa2/3) are Ca(2+)/calmodulin regulated K(+) channels that produce membrane hyperpolarization and shape neurologic, epithelial, cardiovascular, and immunologic functions. Moreover, they emerged as therapeutic targets to treat cardiovascular disease, chronic inflammation, and some cancers. Here, we aimed to generate a new pharmacophore for negative-gating modulation of KCa2/3 channels. We synthesized a series of mono- and dibenzoates and identified three dibenzoates [1,3-phenylenebis(methylene) bis(3-fluoro-4-hydroxybenzoate) (RA-2), 1,2-phenylenebis(methylene) bis(3-fluoro-4-hydroxybenzoate), and 1,4-phenylenebis(methylene) bis(3-fluoro-4-hydroxybenzoate)] with inhibitory efficacy as determined by patch clamp. Among them, RA-2 was the most drug-like and inhibited human KCa3.1 with an IC50 of 17 nM and all three human KCa2 subtypes with similar potencies. RA-2 at 100 nM right-shifted the KCa3.1 concentration-response curve for Ca(2+) activation. The positive-gating modulator naphtho[1,2-d]thiazol-2-ylamine (SKA-31) reversed channel inhibition at nanomolar RA-2 concentrations. RA-2 had no considerable blocking effects on distantly related large-conductance KCa1.1, Kv1.2/1.3, Kv7.4, hERG, or inwardly rectifying K(+) channels. In isometric myography on porcine coronary arteries, RA-2 inhibited bradykinin-induced endothelium-derived hyperpolarization (EDH)-type relaxation in U46619-precontracted rings. Blood pressure telemetry in mice showed that intraperitoneal application of RA-2 (≤100 mg/kg) did not increase blood pressure or cause gross behavioral deficits. However, RA-2 decreased heart rate by ≈145 beats per minute, which was not seen in KCa3.1(-/-) mice. In conclusion, we identified the KCa2/3-negative-gating modulator, RA-2, as a new pharmacophore with nanomolar potency. RA-2 may be of use to generate structurally new types of negative-gating modulators that could help to define the physiologic and pathomechanistic roles of KCa2/3 in the vasculature, central nervous system, and during inflammation in vivo.
Collapse
Affiliation(s)
- Aida Oliván-Viguera
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.)
| | - Marta Sofía Valero
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.)
| | - Nicole Coleman
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.)
| | - Brandon M Brown
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.)
| | - Celia Laría
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.)
| | - María Divina Murillo
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.)
| | - José A Gálvez
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.)
| | - María D Díaz-de-Villegas
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.)
| | - Heike Wulff
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.)
| | - Ramón Badorrey
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.)
| | - Ralf Köhler
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.).
| |
Collapse
|
44
|
Giordanetto F, Schäfer A, Ottmann C. Stabilization of protein–protein interactions by small molecules. Drug Discov Today 2014; 19:1812-1821. [DOI: 10.1016/j.drudis.2014.08.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 07/03/2014] [Accepted: 08/18/2014] [Indexed: 12/23/2022]
|
45
|
Cui M, Qin G, Yu K, Bowers MS, Zhang M. Targeting the Small- and Intermediate-Conductance Ca-Activated Potassium Channels: The Drug-Binding Pocket at the Channel/Calmodulin Interface. Neurosignals 2014; 22:65-78. [PMID: 25300231 DOI: 10.1159/000367896] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 08/26/2014] [Indexed: 11/19/2022] Open
Abstract
The small- and intermediate-conductance Ca(2+)-activated potassium (SK/IK) channels play important roles in the regulation of excitable cells in both the central nervous and cardiovascular systems. Evidence from animal models has implicated SK/IK channels in neurological conditions such as ataxia and alcohol use disorders. Further, genome-wide association studies have suggested that cardiovascular abnormalities such as arrhythmias and hypertension are associated with single nucleotide polymorphisms that occur within the genes encoding the SK/IK channels. The Ca(2+) sensitivity of the SK/IK channels stems from a constitutively bound Ca(2+)-binding protein: calmodulin. Small-molecule positive modulators of SK/IK channels have been developed over the past decade, and recent structural studies have revealed that the binding pocket of these positive modulators is located at the interface between the channel and calmodulin. SK/IK channel positive modulators can potentiate channel activity by enhancing the coupling between Ca(2+) sensing via calmodulin and mechanical opening of the channel. Here, we review binding pocket studies that have provided structural insight into the mechanism of action for SK/IK channel positive modulators. These studies lay the foundation for structure-based drug discovery efforts that can identify novel SK/IK channel positive modulators.
Collapse
Affiliation(s)
- Meng Cui
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Va., USA
| | | | | | | | | |
Collapse
|
46
|
Huang P, Zhang Y, Chen X, Zhu L, Yin D, Zeng X, Liang S. The activation effect of hainantoxin-I, a peptide toxin from the Chinese spider, Ornithoctonus hainana, on intermediate-conductance Ca2+-activated K+ channels. Toxins (Basel) 2014; 6:2568-79. [PMID: 25153257 PMCID: PMC4147597 DOI: 10.3390/toxins6082568] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 07/28/2014] [Accepted: 08/14/2014] [Indexed: 11/24/2022] Open
Abstract
Intermediate-conductance Ca2+-activated K+ (IK) channels are calcium/calmodulin-regulated voltage-independent K+ channels. Activation of IK currents is important in vessel and respiratory tissues, rendering the channels potential drug targets. A variety of small organic molecules have been synthesized and found to be potent activators of IK channels. However, the poor selectivity of these molecules limits their therapeutic value. Venom-derived peptides usually block their targets with high specificity. Therefore, we searched for novel peptide activators of IK channels by testing a series of toxins from spiders. Using electrophysiological experiments, we identified hainantoxin-I (HNTX-I) as an IK-channel activator. HNTX-I has little effect on voltage-gated Na+ and Ca2+ channels from rat dorsal root ganglion neurons and on the heterologous expression of voltage-gated rapidly activating delayed rectifier K+ channels (human ether-à-go-go-related gene; human ERG) in HEK293T cells. Only 35.2% ± 0.4% of the currents were activated in SK channels, and there was no effect on BK channels. We demonstrated that HNTX-I was not a phrenic nerve conduction blocker or acutely toxic. This is believed to be the first report of a peptide activator effect on IK channels. Our study suggests that the activity and selectivity of HNTX-I on IK channels make HNTX-I a promising template for designing new drugs for cardiovascular diseases.
Collapse
Affiliation(s)
- Pengfei Huang
- Key Laboratory of Protein Chemistry and Developmental Biology of the Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China.
| | - Yiya Zhang
- Key Laboratory of Protein Chemistry and Developmental Biology of the Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China.
| | - Xinyi Chen
- Key Laboratory of Protein Chemistry and Developmental Biology of the Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China.
| | - Li Zhu
- Key Laboratory of Protein Chemistry and Developmental Biology of the Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China.
| | - Dazhong Yin
- Key Laboratory of Protein Chemistry and Developmental Biology of the Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China.
| | - Xiongzhi Zeng
- Key Laboratory of Protein Chemistry and Developmental Biology of the Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China.
| | - Songping Liang
- Key Laboratory of Protein Chemistry and Developmental Biology of the Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China.
| |
Collapse
|
47
|
Zhang M, Meng XY, Cui M, Pascal JM, Logothetis DE, Zhang JF. Selective phosphorylation modulates the PIP2 sensitivity of the CaM-SK channel complex. Nat Chem Biol 2014; 10:753-9. [PMID: 25108821 DOI: 10.1038/nchembio.1592] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 06/19/2014] [Indexed: 11/09/2022]
Abstract
Phosphatidylinositol bisphosphate (PIP2) regulates the activities of many membrane proteins, including ion channels, through direct interactions. However, the affinity of PIP2 is so high for some channel proteins that its physiological role as a modulator has been questioned. Here we show that PIP2 is a key cofactor for activation of small conductance Ca2+-activated potassium channels (SKs) by Ca(2+)-bound calmodulin (CaM). Removal of the endogenous PIP2 inhibits SKs. The PIP2-binding site resides at the interface of CaM and the SK C terminus. We further demonstrate that the affinity of PIP2 for its target proteins can be regulated by cellular signaling. Phosphorylation of CaM T79, located adjacent to the PIP2-binding site, by casein kinase 2 reduces the affinity of PIP2 for the CaM-SK channel complex by altering the dynamic interactions among amino acid residues surrounding the PIP2-binding site. This effect of CaM phosphorylation promotes greater channel inhibition by G protein-mediated hydrolysis of PIP2.
Collapse
Affiliation(s)
- Miao Zhang
- 1] Department of Molecular Physiology and Biophysics, Thomas Jefferson University, Philadelphia, Pennsylvania, USA. [2] Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Xuan-Yu Meng
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Meng Cui
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - John M Pascal
- Department of Biochemistry &Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Diomedes E Logothetis
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Ji-Fang Zhang
- 1] Department of Molecular Physiology and Biophysics, Thomas Jefferson University, Philadelphia, Pennsylvania, USA. [2] Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA. [3] Graduate Program in Neuroscience, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
48
|
Coleman N, Brown BM, Oliván-Viguera A, Singh V, Olmstead MM, Valero MS, Köhler R, Wulff H. New positive Ca2+-activated K+ channel gating modulators with selectivity for KCa3.1. Mol Pharmacol 2014; 86:342-57. [PMID: 24958817 DOI: 10.1124/mol.114.093286] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Small-conductance (KCa2) and intermediate-conductance (KCa3.1) calcium-activated K(+) channels are voltage-independent and share a common calcium/calmodulin-mediated gating mechanism. Existing positive gating modulators like EBIO, NS309, or SKA-31 activate both KCa2 and KCa3.1 channels with similar potency or, as in the case of CyPPA and NS13001, selectively activate KCa2.2 and KCa2.3 channels. We performed a structure-activity relationship (SAR) study with the aim of optimizing the benzothiazole pharmacophore of SKA-31 toward KCa3.1 selectivity. We identified SKA-111 (5-methylnaphtho[1,2-d]thiazol-2-amine), which displays 123-fold selectivity for KCa3.1 (EC50 111 ± 27 nM) over KCa2.3 (EC50 13.7 ± 6.9 μM), and SKA-121 (5-methylnaphtho[2,1-d]oxazol-2-amine), which displays 41-fold selectivity for KCa3.1 (EC50 109 nM ± 14 nM) over KCa2.3 (EC50 4.4 ± 1.6 μM). Both compounds are 200- to 400-fold selective over representative KV (KV1.3, KV2.1, KV3.1, and KV11.1), NaV (NaV1.2, NaV1.4, NaV1.5, and NaV1.7), as well as CaV1.2 channels. SKA-121 is a typical positive-gating modulator and shifts the calcium-concentration response curve of KCa3.1 to the left. In blood pressure telemetry experiments, SKA-121 (100 mg/kg i.p.) significantly lowered mean arterial blood pressure in normotensive and hypertensive wild-type but not in KCa3.1(-/-) mice. SKA-111, which was found in pharmacokinetic experiments to have a much longer half-life and to be much more brain penetrant than SKA-121, not only lowered blood pressure but also drastically reduced heart rate, presumably through cardiac and neuronal KCa2 activation when dosed at 100 mg/kg. In conclusion, with SKA-121, we generated a KCa3.1-specific positive gating modulator suitable for further exploring the therapeutical potential of KCa3.1 activation.
Collapse
Affiliation(s)
- Nichole Coleman
- Department of Pharmacology (N.C., B.M.B., V.S., H.W.), School of Medicine, and Department of Chemistry (M.M.O.), University of California, Davis, California; Aragon Institute of Health Sciences, Instituto de Investigación Sanitaria, Fundación Agencia Aragonesa para la Investigación y el Desarrollo, Zaragoza, Spain (A.O.-V., R.K.); and Grupo de Investigación del Medio Ambiente del Centro de Estudios Superiores, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V.)
| | - Brandon M Brown
- Department of Pharmacology (N.C., B.M.B., V.S., H.W.), School of Medicine, and Department of Chemistry (M.M.O.), University of California, Davis, California; Aragon Institute of Health Sciences, Instituto de Investigación Sanitaria, Fundación Agencia Aragonesa para la Investigación y el Desarrollo, Zaragoza, Spain (A.O.-V., R.K.); and Grupo de Investigación del Medio Ambiente del Centro de Estudios Superiores, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V.)
| | - Aida Oliván-Viguera
- Department of Pharmacology (N.C., B.M.B., V.S., H.W.), School of Medicine, and Department of Chemistry (M.M.O.), University of California, Davis, California; Aragon Institute of Health Sciences, Instituto de Investigación Sanitaria, Fundación Agencia Aragonesa para la Investigación y el Desarrollo, Zaragoza, Spain (A.O.-V., R.K.); and Grupo de Investigación del Medio Ambiente del Centro de Estudios Superiores, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V.)
| | - Vikrant Singh
- Department of Pharmacology (N.C., B.M.B., V.S., H.W.), School of Medicine, and Department of Chemistry (M.M.O.), University of California, Davis, California; Aragon Institute of Health Sciences, Instituto de Investigación Sanitaria, Fundación Agencia Aragonesa para la Investigación y el Desarrollo, Zaragoza, Spain (A.O.-V., R.K.); and Grupo de Investigación del Medio Ambiente del Centro de Estudios Superiores, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V.)
| | - Marilyn M Olmstead
- Department of Pharmacology (N.C., B.M.B., V.S., H.W.), School of Medicine, and Department of Chemistry (M.M.O.), University of California, Davis, California; Aragon Institute of Health Sciences, Instituto de Investigación Sanitaria, Fundación Agencia Aragonesa para la Investigación y el Desarrollo, Zaragoza, Spain (A.O.-V., R.K.); and Grupo de Investigación del Medio Ambiente del Centro de Estudios Superiores, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V.)
| | - Marta Sofia Valero
- Department of Pharmacology (N.C., B.M.B., V.S., H.W.), School of Medicine, and Department of Chemistry (M.M.O.), University of California, Davis, California; Aragon Institute of Health Sciences, Instituto de Investigación Sanitaria, Fundación Agencia Aragonesa para la Investigación y el Desarrollo, Zaragoza, Spain (A.O.-V., R.K.); and Grupo de Investigación del Medio Ambiente del Centro de Estudios Superiores, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V.)
| | - Ralf Köhler
- Department of Pharmacology (N.C., B.M.B., V.S., H.W.), School of Medicine, and Department of Chemistry (M.M.O.), University of California, Davis, California; Aragon Institute of Health Sciences, Instituto de Investigación Sanitaria, Fundación Agencia Aragonesa para la Investigación y el Desarrollo, Zaragoza, Spain (A.O.-V., R.K.); and Grupo de Investigación del Medio Ambiente del Centro de Estudios Superiores, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V.)
| | - Heike Wulff
- Department of Pharmacology (N.C., B.M.B., V.S., H.W.), School of Medicine, and Department of Chemistry (M.M.O.), University of California, Davis, California; Aragon Institute of Health Sciences, Instituto de Investigación Sanitaria, Fundación Agencia Aragonesa para la Investigación y el Desarrollo, Zaragoza, Spain (A.O.-V., R.K.); and Grupo de Investigación del Medio Ambiente del Centro de Estudios Superiores, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V.)
| |
Collapse
|
49
|
Milroy LG, Grossmann TN, Hennig S, Brunsveld L, Ottmann C. Modulators of Protein–Protein Interactions. Chem Rev 2014; 114:4695-748. [DOI: 10.1021/cr400698c] [Citation(s) in RCA: 352] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Lech-Gustav Milroy
- Laboratory
of Chemical Biology and Institute of Complex Molecular Systems, Department
of Biomedical Engineering, Technische Universiteit Eindhoven, Den Dolech
2, 5612 AZ Eindhoven, The Netherlands
| | - Tom N. Grossmann
- Chemical Genomics Centre of the Max Planck Society, Otto-Hahn Straße 15, 44227 Dortmund, Germany
- Department
of Chemistry and Chemical Biology, Technical University Dortmund, Otto-Hahn-Strasse 6, 44227 Dortmund, Germany
| | - Sven Hennig
- Chemical Genomics Centre of the Max Planck Society, Otto-Hahn Straße 15, 44227 Dortmund, Germany
| | - Luc Brunsveld
- Laboratory
of Chemical Biology and Institute of Complex Molecular Systems, Department
of Biomedical Engineering, Technische Universiteit Eindhoven, Den Dolech
2, 5612 AZ Eindhoven, The Netherlands
| | - Christian Ottmann
- Laboratory
of Chemical Biology and Institute of Complex Molecular Systems, Department
of Biomedical Engineering, Technische Universiteit Eindhoven, Den Dolech
2, 5612 AZ Eindhoven, The Netherlands
| |
Collapse
|
50
|
Halling DB, Kenrick SA, Riggs AF, Aldrich RW. Calcium-dependent stoichiometries of the KCa2.2 (SK) intracellular domain/calmodulin complex in solution. ACTA ACUST UNITED AC 2014; 143:231-52. [PMID: 24420768 PMCID: PMC4001768 DOI: 10.1085/jgp.201311007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Biophysical analyses indicate that the Ca2+-activated K+ channel SK2 binds calmodulin with multiple stoichiometries, distinct from the two SK2-two calmodulin stoichiometry identified by crystallography. Ca2+ activates SK Ca2+-activated K+ channels through the protein Ca2+ sensor, calmodulin (CaM). To understand how SK channels operate, it is necessary to determine how Ca2+ regulates CaM binding to its target on SK. Tagless, recombinant SK peptide (SKp), was purified for binding studies with CaM at low and high Ca2+ concentrations. Composition gradient multi-angle light scattering accurately measures the molar mass, stoichiometry, and affinity of protein complexes. In 2 mM Ca2+, SKp and CaM bind with three different stoichiometries that depend on the molar ratio of SKp:CaM in solution. These complexes include 28 kD 1SKp/1CaM, 39 kD 2SKp/1CaM, and 44 kD 1SKp/2CaM. A 2SKp/2CaM complex, observed in prior crystallographic studies, is absent. At <5 nM Ca2+, 1SKp/1CaM and 2SKp/1CaM were observed; however, 1SKp/2CaM was absent. Analytical ultracentrifugation was used to characterize the physical properties of the three SKp/CaM stoichiometries. In high Ca2+, the sedimentation coefficient is smaller for a 1SKp:1CaM solution than it is for either 2SKp:1CaM or 1SKp:2CaM. At low Ca2+ and at >100 µM protein concentrations, a molar excess of SKp over CaM causes aggregation. Aggregation is not observed in Ca2+ or with CaM in molar excess. In low Ca2+ both 1SKp:1CaM and 1SKp:2CaM solutions have similar sedimentation coefficients, which is consistent with the absence of a 1SKp/2CaM complex in low Ca2+. These results suggest that complexes with stoichiometries other than 2SKp/2CaM are important in gating.
Collapse
Affiliation(s)
- D Brent Halling
- Department of Neuroscience, 2 Center for Learning and Memory, The University of Texas at Austin, Austin, TX 78712
| | | | | | | |
Collapse
|