1
|
Zadmajid V, Shahriar S, Gorelick DA. Testosterone acts through the membrane protein GPRC6A to cause cardiac edema in zebrafish embryos. Development 2024; 151:dev204390. [PMID: 39479956 PMCID: PMC11634029 DOI: 10.1242/dev.204390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 10/06/2024] [Indexed: 11/02/2024]
Abstract
Androgens are classically thought to act through intracellular androgen receptors (AR/NR3C4), but they can also trigger non-genomic effects via membrane proteins. Although several membrane androgen receptors have been characterized in vitro, their functions in vivo remain unclear. Using a chemical-genetic screen in zebrafish, we found that GPRC6A, a G-protein-coupled receptor, mediates non-genomic androgen actions during embryonic development. Exposure to androgens (androstanedione, DHT and testosterone) caused cardiac edema or tail curvature in wild-type embryos, as well as in ar mutants, suggesting AR-independent pathways. We then mutated putative membrane androgen receptors [gprc6a, hcar1-4 and zip9 (slc39a9)] and found that only gprc6a mutants exhibited a significant reduction in cardiac edema after testosterone exposure. Additionally, co-treatment of wild-type embryos with testosterone and GPRC6A antagonists significantly suppressed the cardiac edema phenotype. Using RNA-seq and RNA rescue approaches, we found that testosterone and GPRC6A cause cardiac phenotypes by reducing Pak1 signaling. Our results indicate that testosterone induces cardiac edema in zebrafish embryos through GPRC6A, independent of nuclear androgen receptors, highlighting a previously unappreciated non-genomic androgen signaling pathway in embryonic development.
Collapse
MESH Headings
- Animals
- Zebrafish/embryology
- Zebrafish/metabolism
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Testosterone/metabolism
- Testosterone/pharmacology
- Zebrafish Proteins/metabolism
- Zebrafish Proteins/genetics
- Edema, Cardiac/metabolism
- Edema, Cardiac/pathology
- Edema, Cardiac/genetics
- Embryo, Nonmammalian/metabolism
- Embryo, Nonmammalian/drug effects
- Receptors, Androgen/metabolism
- Receptors, Androgen/genetics
- Signal Transduction/drug effects
- Gene Expression Regulation, Developmental/drug effects
- Heart/embryology
- Heart/drug effects
- Androgens/pharmacology
- Androgens/metabolism
- Mutation/genetics
Collapse
Affiliation(s)
- Vahid Zadmajid
- Center for Precision Environmental Health, Department of Molecular & Cellular Biology, Baylor College of Medicine, One Baylor Plaza, BCM229, Houston, TX 77030, USA
| | - Shayan Shahriar
- Center for Precision Environmental Health, Department of Molecular & Cellular Biology, Baylor College of Medicine, One Baylor Plaza, BCM229, Houston, TX 77030, USA
| | - Daniel A. Gorelick
- Center for Precision Environmental Health, Department of Molecular & Cellular Biology, Baylor College of Medicine, One Baylor Plaza, BCM229, Houston, TX 77030, USA
| |
Collapse
|
2
|
Hollingworth D, Herold KF, Kelly G, Mykhaylyk VB, Xiang J, Zhang D, Wallace BA, Hemmings HC. Functionally important binding site for a volatile anesthetic in a voltage-gated sodium channel identified by X-ray crystallography. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621342. [PMID: 39574657 PMCID: PMC11580951 DOI: 10.1101/2024.11.04.621342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Volatile general anesthetics are used for inhalational anesthesia in hundreds of millions of surgical procedures annually, yet their mechanisms of action remain unclear. Membrane proteins involved in cell signaling are major targets for anesthetics, and voltage-gated ion channels that mediate neurotransmission, movement, and cognition are sensitive to volatile anesthetics (VAs). In many cases, the effects produced by VAs on mammalian ion channels are reproduced in prokaryotic orthologues, providing an opportunity to investigate VA interactions at high resolution using these structurally simpler prokaryotic proteins. We utilized the bacterial voltage-gated sodium channel (VGSC) NavMs from Magnetococcus marinus to investigate its interaction with the widely used VA sevoflurane. Sevoflurane interacted directly with NavMs, producing effects consistent with multisite binding models for VA actions on their targets. We report the identification of one of these interactions at atomic detail providing the first high-resolution structure of a VA bound to a voltage-gated ion channel. The X-ray crystal structure shows sevoflurane binding to NavMs within an intramembrane hydrophobic pocket formed by residues from the voltage sensor and channel pore, domains essential for channel gating. Mutation of the dominant sevoflurane binding-site residue within this pocket, and analogous residues found in similar sites in human VGSCs, profoundly affected channel properties, supporting a critical role for this site in VGSC function. These findings provide the basis for future work to understand the role of VA interactions with VGSCs in both the anesthetic and toxic effects associated with general anesthesia.
Collapse
Affiliation(s)
| | - Karl F Herold
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Geoff Kelly
- The Medical Research Council Biomedical NMR Centre, The Francis Crick Institute, London, UK
| | - Vitaliy B Mykhaylyk
- Diamond Light Source, Harwell Science and Innovation Campus, Chilton, Didcot, UK
| | - Jiaxin Xiang
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Donghang Zhang
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - B A Wallace
- School of Natural Sciences, Birkbeck University of London, London, UK
| | - Hugh C Hemmings
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
3
|
Hollingworth D, Thomas F, Page DA, Fouda MA, De Castro RLR, Sula A, Mykhaylyk VB, Kelly G, Ulmschneider MB, Ruben PC, Wallace BA. Structural basis for the rescue of hyperexcitable cells by the amyotrophic lateral sclerosis drug Riluzole. Nat Commun 2024; 15:8426. [PMID: 39341837 PMCID: PMC11438954 DOI: 10.1038/s41467-024-52539-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 09/09/2024] [Indexed: 10/01/2024] Open
Abstract
Neuronal hyperexcitability is a key element of many neurodegenerative disorders including the motor neuron disease Amyotrophic Lateral Sclerosis (ALS), where it occurs associated with elevated late sodium current (INaL). INaL results from incomplete inactivation of voltage-gated sodium channels (VGSCs) after their opening and shapes physiological membrane excitability. However, dysfunctional increases can cause hyperexcitability-associated diseases. Here we reveal the atypical binding mechanism which explains how the neuroprotective ALS-treatment drug riluzole stabilises VGSCs in their inactivated state to cause the suppression of INaL that leads to reversed cellular overexcitability. Riluzole accumulates in the membrane and enters VGSCs through openings to their membrane-accessible fenestrations. Riluzole binds within these fenestrations to stabilise the inactivated channel state, allowing for the selective allosteric inhibition of INaL without the physical block of Na+ conduction associated with traditional channel pore binding VGSC drugs. We further demonstrate that riluzole can reproduce these effects on a disease variant of the non-neuronal VGSC isoform Nav1.4, where pathologically increased INaL is caused directly by mutation. Overall, we identify a model for VGSC inhibition that produces effects consistent with the inhibitory action of riluzole observed in models of ALS. Our findings will aid future drug design and supports research directed towards riluzole repurposing.
Collapse
Affiliation(s)
- David Hollingworth
- School of Natural Sciences, Birkbeck University of London, London, United Kingdom
| | - Frances Thomas
- School of Natural Sciences, Birkbeck University of London, London, United Kingdom
| | - Dana A Page
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Mohamed A Fouda
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Raquel Lopez-Rios De Castro
- Department of Chemistry, King's College London, London, United Kingdom
- Biological Physics and Soft Matter Group, Department of Physics, King's College London, London, United Kingdom
| | - Altin Sula
- Syngenta Crop Protection, Jealott's Hill International Research Centre, Bracknell, Berkshire, United Kingdom
| | - Vitaliy B Mykhaylyk
- Diamond Light Source, Harwell Science and Innovation Campus, Chilton, Didcot, UK
| | - Geoff Kelly
- The Medical Research Council Biomedical NMR Centre, The Francis Crick Institute, London, UK
| | | | - Peter C Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada.
| | - B A Wallace
- School of Natural Sciences, Birkbeck University of London, London, United Kingdom.
| |
Collapse
|
4
|
Kranjčević JK, Čonkaš J, Ozretić P. The Role of Estrogen and Estrogen Receptors in Head and Neck Tumors. Cancers (Basel) 2024; 16:1575. [PMID: 38672656 PMCID: PMC11049451 DOI: 10.3390/cancers16081575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/12/2024] [Accepted: 04/18/2024] [Indexed: 04/21/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the most common histological form of head and neck tumors (HNTs), which originate from the epithelium of the lips and oral cavity, pharynx, larynx, salivary glands, nasal cavity, and sinuses. The main risk factors include consumption of tobacco in all forms and alcohol, as well as infections with high-risk human papillomaviruses or the Epstein-Barr virus. Regardless of the etiological agent, the risk of developing different types of HNTs is from two to more than six times higher in males than in females. The reason for such disparities probably lies in a combination of both biological and psychosocial factors. Therefore, it is hypothesized that exposure to female sex hormones, primarily estrogen, provides women with protection against the formation and metastasis of HNTs. In this review, we synthesized available knowledge on the role of estrogen and estrogen receptors (ERs) in the development and progression of HNTs, with special emphasis on membrane ERs, which are much less studied. We can summarize that in addition to epidemiologic studies unequivocally pointing to the protective effect of estrogen in women, an increased expression of both nuclear ERs, ERα, and ERβ, and membrane ERs, ERα36, GPER1, and NaV1.2, was present in different types of HNSCC, for which anti-estrogens could be used as an effective therapeutic approach.
Collapse
Affiliation(s)
| | | | - Petar Ozretić
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia (J.Č.)
| |
Collapse
|
5
|
Xie M, Zhu S, Liu G, Wu Y, Zhou W, Yu D, Wan J, Xing S, Wang S, Gan L, Li G, Chang D, Lai H, Liu N, Zhu P. A Novel Quantitative Electrocardiography Strategy Reveals the Electroinhibitory Effect of Tamoxifen on the Mouse Heart. J Cardiovasc Transl Res 2023; 16:1232-1248. [PMID: 37155136 DOI: 10.1007/s12265-023-10395-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 04/26/2023] [Indexed: 05/10/2023]
Abstract
Tamoxifen, a selective estrogen receptor modulator, was initially used to treat cancer in women and more recently to induce conditional gene editing in rodent hearts. However, little is known about the baseline biological effects of tamoxifen on the myocardium. In order to clarify the short-term effects of tamoxifen on cardiac electrophysiology of myocardium, we applied a single-chest-lead quantitative method and analyzed the short-term electrocardiographic phenotypes induced by tamoxifen in the heart of adult female mice. We found that tamoxifen prolonged the PP interval and caused a decreased heartbeat, and further induced atrioventricular block by gradually prolonging the PR interval. Further correlation analysis suggested that tamoxifen had a synergistic and dose-independent inhibition on the time course of the PP interval and PR interval. This prolongation of the critical time course may represent a tamoxifen-specific ECG excitatory-inhibitory mechanism, leading to a reduction in the number of supraventricular action potentials and thus bradycardia. Segmental reconstructions showed that tamoxifen induced a decrease in the conduction velocity of action potentials throughout the atria and parts of the ventricles, resulting in a flattening of the P wave and R wave. In addition, we detected the previously reported prolongation of the QT interval, which may be due to a prolonged duration of the ventricular repolarizing T wave rather than the depolarizing QRS complex. Our study highlights that tamoxifen can produce patterning alternations in the cardiac conduction system, including the formation of inhibitory electrical signals with reduced conduction velocity, implying its involvement in the regulation of myocardial ion transport and the mediation of arrhythmias. A Novel Quantitative Electrocardiography Strategy Reveals the Electroinhibitory Effect of Tamoxifen on the Mouse Heart(Figure 9). A working model of tamoxifen producing acute electrical disturbances in the myocardium. SN, sinus node; AVN, atrioventricular node; RA, right atrium; LA, left atrium; RV, right ventricle; LV, left ventricle.
Collapse
Affiliation(s)
- Ming Xie
- Department of Cardiac Surgery, School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, Guangdong, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, 510100, Guangdong, China
| | - Shuoji Zhu
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, Guangdong, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, 510100, Guangdong, China
- University of Tokyo, Tokyo, 113-8666, Japan
| | - Gang Liu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yijin Wu
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, Guangdong, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, 510100, Guangdong, China
| | - Wenkai Zhou
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, Guangdong, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, 510100, Guangdong, China
| | - Dingdang Yu
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, Guangdong, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, 510100, Guangdong, China
| | - Jinkai Wan
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Shenghui Xing
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Siqing Wang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Lin Gan
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, Guangdong, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, 510100, Guangdong, China
| | - Ge Li
- Department of Cardiac Surgery, School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, Guangdong, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, 510100, Guangdong, China
| | - Dehua Chang
- University of Tokyo Hospital Department of Cell Therapy in Regenerative Medicine, Tokyo, 113-8666, Japan.
| | - Hao Lai
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Nanbo Liu
- Department of Cardiac Surgery, School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China.
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, Guangdong, China.
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, 510100, Guangdong, China.
| | - Ping Zhu
- Department of Cardiac Surgery, School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China.
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, Guangdong, China.
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, 510100, Guangdong, China.
| |
Collapse
|
6
|
Negami T, Terada T. Calculations of the binding free energies of the Comprehensive in vitro Proarrhythmia Assay (CiPA) reference drugs to cardiac ion channels. Biophys Physicobiol 2023; 20:e200016. [PMID: 38496247 PMCID: PMC10941965 DOI: 10.2142/biophysico.bppb-v20.0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/24/2023] [Indexed: 03/19/2024] Open
Abstract
The evaluation of the inhibitory activities of drugs on multiple cardiac ion channels is required for the accurate assessment of proarrhythmic risks. Moreover, the in silico prediction of such inhibitory activities of drugs on cardiac channels can improve the efficiency of the drug-development process. Here, we performed molecular docking simulations to predict the complex structures of 25 reference drugs that were proposed by the Comprehensive in vitro Proarrhythmia Assay consortium using two cardiac ion channels, the human ether-a-go-go-related gene (hERG) potassium channel and human NaV1.5 (hNaV1.5) sodium channel, with experimentally available structures. The absolute binding free energy (ΔGbind) values of the predicted structures were calculated by a molecular dynamics-based method and compared with the experimental half-maximal inhibitory concentration (IC50) data. Furthermore, the regression analysis between the calculated values and negative of the common logarithm of the experimental IC50 values (pIC50) revealed that the calculated values of four and ten drugs deviated significantly from the regression lines of the hERG and hNaV1.5 channels, respectively. We reconsidered the docking poses and protonation states of the drugs based on the experimental data and recalculated their ΔGbind values. Finally, the calculated ΔGbind values of 24 and 19 drugs correlated with their experimental pIC50 values (coefficients of determination=0.791 and 0.613 for the hERG and hNaV1.5 channels, respectively). Thus, the regression analysis between the calculated ΔGbind and experimental IC50 data ensured the realization of an increased number of reliable complex structures.
Collapse
Affiliation(s)
- Tatsuki Negami
- Department of Biotechnology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Tohru Terada
- Department of Biotechnology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
7
|
Choudhury K, Howard RJ, Delemotte L. An α-π transition in S6 shapes the conformational cycle of the bacterial sodium channel NavAb. J Gen Physiol 2022; 155:213748. [PMID: 36515966 PMCID: PMC9754703 DOI: 10.1085/jgp.202213214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/17/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Voltage-gated sodium channels play an important role in electrical signaling in excitable cells. In response to changes in membrane potential, they cycle between nonconducting and conducting conformations. With recent advances in structural biology, structures of sodium channels have been captured in several distinct conformations, which are thought to represent different functional states. However, it has been difficult to capture the intrinsically transient open state. We recently showed that a proposed open state of the bacterial sodium channel NavMs was not conductive and that a conformational change involving a transition to a π-helix in the pore-lining S6 helix converted this structure into a conducting state. However, the relevance of this structural feature in other sodium channels, and its implications for the broader gating cycle, remained unclear. Here, we propose a comparable open state of another class of bacterial channel from Aliarcobacter butzleri (NavAb) with characteristic pore hydration, ion permeation, and drug binding properties. Furthermore, we show that a π-helix transition can lead to pore opening and that such a conformational change blocks fenestrations in the inner helix bundle. We also discover that a region in the C-terminal domain can undergo a disordering transition proposed to be important for pore opening. These results support a role for a π-helix transition in the opening of NavAb, enabling new proposals for the structural annotation and drug modulation mechanisms in this important sodium channel model.
Collapse
Affiliation(s)
- Koushik Choudhury
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Solna, Sweden
| | - Rebecca J. Howard
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Lucie Delemotte
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Solna, Sweden,Correspondence to Lucie Delemotte:
| |
Collapse
|
8
|
McCollum MM, Larmore M, Ishihara S, Ng LCT, Kimura LF, Guadarrama E, Ta MC, Vien TN, Frost GB, Scheidt KA, Miller RE, DeCaen PG. Targeting the tamoxifen receptor within sodium channels to block osteoarthritic pain. Cell Rep 2022; 40:111248. [PMID: 36001977 PMCID: PMC9523973 DOI: 10.1016/j.celrep.2022.111248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 06/29/2022] [Accepted: 08/01/2022] [Indexed: 01/11/2023] Open
Abstract
Voltage-gated sodium channels (NaV) in nociceptive neurons initiate action potentials required for transmission of aberrant painful stimuli observed in osteoarthritis (OA). Targeting NaV subtypes with drugs to produce analgesic effects for OA pain management is a developing therapeutic area. Previously, we determined the receptor site for the tamoxifen analog N-desmethyltamoxifen (ND-Tam) within a prokaryotic NaV. Here, we report the pharmacology of ND-Tam against eukaryotic NaVs natively expressed in nociceptive neurons. ND-Tam and analogs occupy two conserved intracellular receptor sites in domains II and IV of NaV1.7 to block ion entry using a "bind and plug" mechanism. We find that ND-Tam inhibition of the sodium current is state dependent, conferring a potent frequency- and voltage-dependent block of hyperexcitable nociceptive neuron action potentials implicated in OA pain. When evaluated using a mouse OA pain model, ND-Tam has long-lasting efficacy, which supports the potential of repurposing ND-Tam analogs as NaV antagonists for OA pain management.
Collapse
Affiliation(s)
- Megan M McCollum
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Megan Larmore
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Shingo Ishihara
- Division of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Leo C T Ng
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Louise F Kimura
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Eduardo Guadarrama
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - My C Ta
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Thuy N Vien
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Grant B Frost
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
| | - Karl A Scheidt
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
| | - Rachel E Miller
- Division of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Paul G DeCaen
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
9
|
Characterization in Inhibitory Effectiveness of Carbamazepine in Voltage-Gated Na + and Erg-Mediated K + Currents in a Mouse Neural Crest-Derived (Neuro-2a) Cell Line. Int J Mol Sci 2022; 23:ijms23147892. [PMID: 35887240 PMCID: PMC9321339 DOI: 10.3390/ijms23147892] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/15/2022] [Accepted: 07/15/2022] [Indexed: 12/18/2022] Open
Abstract
Carbamazepine (CBZ, Tegretol®) is an anticonvulsant used in the treatment of epilepsy and neuropathic pain; however, several unwanted effects of this drug have been noticed. Therefore, the regulatory actions of CBZ on ionic currents in electrically excitable cells need to be reappraised, although its efficacy in suppressing voltage-gated Na+ current (INa) has been disclosed. This study was undertaken to explore the modifications produced by CBZ on ionic currents (e.g., INa and erg-mediated K+ current [IK(erg)]) measured from Neuro-2a (N2a) cells. In these cells, we found that this drug differentially suppressed the peak (transient, INa(T)) and sustained (late, INa(L)) components of INa in a concentration-dependent manner with effective IC50 of 56 and 18 μM, respectively. The overall current–voltage relationship of INa(T) with or without the addition of CBZ remained unchanged; however, the strength (i.e., ∆area) in the window component of INa (INa(W)) evoked by the short ascending ramp pulse (Vramp) was overly lessened in the CBZ presence. Tefluthrin (Tef), a synthetic pyrethroid, known to stimulate INa, augmented the strength of the voltage-dependent hysteresis (Hys(V)) of persistent INa (INa(P)) in response to the isosceles-triangular Vramp; moreover, further application of CBZ attenuated Tef-mediated accentuation of INa(P)’s Hys(V). With a two-step voltage protocol, the recovery of INa(T) inactivation seen in Neuro-2a cells became progressively slowed by adding CBZ; however, the cumulative inhibition of INa(T) evoked by pulse train stimulation was enhanced during exposure to this drug. Neuro-2a-cell exposure to CBZ (100 μM), the magnitude of erg-mediated K+ current measured throughout the entire voltage-clamp steps applied was mildly inhibited. The docking results regarding the interaction of CBZ and voltage-gate Na+ (NaV) channel predicted the ability of CBZ to bind to some amino-acid residues in NaV due to the existence of a hydrogen bond or hydrophobic contact. It is conceivable from the current investigations that the INa (INa(T), INa(L), INa(W), and INa(P)) residing in Neuro-2a cells are susceptible to being suppressed by CBZ, and that its block on INa(L) is larger than that on INa(T). Collectively, the magnitude and gating of NaV channels produced by the CBZ presence might have an impact on its anticonvulsant and analgesic effects occurring in vivo.
Collapse
|
10
|
Djamgoz MBA. Combinatorial Therapy of Cancer: Possible Advantages of Involving Modulators of Ionic Mechanisms. Cancers (Basel) 2022; 14:2703. [PMID: 35681682 PMCID: PMC9179511 DOI: 10.3390/cancers14112703] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/22/2022] [Accepted: 05/27/2022] [Indexed: 02/04/2023] Open
Abstract
Cancer is a global health problem that 1 in 2-3 people can expect to experience during their lifetime. Several different modalities exist for cancer management, but all of these suffer from significant shortcomings in both diagnosis and therapy. Apart from developing completely new therapies, a viable way forward is to improve the efficacy of the existing modalities. One way is to combine these with each other or with other complementary approaches. An emerging latter approach is derived from ionic mechanisms, mainly ion channels and exchangers. We evaluate the evidence for this systematically for the main treatment methods: surgery, chemotherapy, radiotherapy and targeted therapies (including monoclonal antibodies, steroid hormones, tyrosine kinase inhibitors and immunotherapy). In surgery, the possible systemic use of local anesthetics to suppress subsequent relapse is still being discussed. For all the other methods, there is significant positive evidence for several cancers and a range of modulators of ionic mechanisms. This applies also to some of the undesirable side effects of the treatments. In chemotherapy, for example, there is evidence for co-treatment with modulators of the potassium channel (Kv11.1), pH regulation (sodium-hydrogen exchanger) and Na+-K+-ATPase (digoxin). Voltage-gated sodium channels, shown previously to promote metastasis, appear to be particularly useful for co-targeting with inhibitors of tyrosine kinases, especially epidermal growth factor. It is concluded that combining current orthodox treatment modalities with modulators of ionic mechanisms can produce beneficial effects including (i) making the treatment more effective, e.g., by lowering doses; (ii) avoiding the onset of resistance to therapy; (iii) reducing undesirable side effects. However, in many cases, prospective clinical trials are needed to put the findings firmly into clinical context.
Collapse
Affiliation(s)
- Mustafa B. A. Djamgoz
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK; ; Tel.: +44-796-181-6959
- Biotechnology Research Centre, Cyprus International University, Haspolat, Mersin 10, Turkey
| |
Collapse
|
11
|
Fouda MA, Ghovanloo MR, Ruben PC. Late sodium current: incomplete inactivation triggers seizures, myotonias, arrhythmias, and pain syndromes. J Physiol 2022; 600:2835-2851. [PMID: 35436004 DOI: 10.1113/jp282768] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/12/2022] [Indexed: 11/08/2022] Open
Abstract
Acquired and inherited dysfunction in voltage-gated sodium channels underlies a wide range of diseases. "In addition to the defects in trafficking and expression, sodium channelopathies are also caused by dysfunction in one or several gating properties, for instance activation or inactivation. Disruption of the channel inactivation leads to the increased late sodium current, which is a common defect in seizure disorders, cardiac arrhythmias skeletal muscle myotonia and pain. An increase in late sodium current leads to repetitive action potential in neurons and skeletal muscles, and prolonged action potential duration in the heart. In this topical review, we compare the effects of late sodium current in brain, heart, skeletal muscle, and peripheral nerves. Abstract figure legend Shows cartoon illustration of general Nav channel transitions between (1) resting, (2) open, and (3) fast inactivated states. Disruption of the inactivation process exacerbates (4) late sodium currents. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Mohamed A Fouda
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada.,Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt
| | | | - Peter C Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| |
Collapse
|
12
|
Choudhury K, Kasimova MA, McComas S, Howard RJ, Delemotte L. An open state of a voltage-gated sodium channel involving a π-helix and conserved pore-facing asparagine. Biophys J 2022; 121:11-22. [PMID: 34890580 PMCID: PMC8758419 DOI: 10.1016/j.bpj.2021.12.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/26/2021] [Accepted: 12/06/2021] [Indexed: 01/07/2023] Open
Abstract
Voltage-gated sodium (Nav) channels play critical roles in propagating action potentials and otherwise manipulating ionic gradients in excitable cells. These channels open in response to membrane depolarization, selectively permeating sodium ions until rapidly inactivating. Structural characterization of the gating cycle in this channel family has proved challenging, particularly due to the transient nature of the open state. A structure from the bacterium Magnetococcus marinus Nav (NavMs) was initially proposed to be open, based on its pore diameter and voltage-sensor conformation. However, the functional annotation of this model, and the structural details of the open state, remain disputed. In this work, we used molecular modeling and simulations to test possible open-state models of NavMs. The full-length experimental structure, termed here the α-model, was consistently dehydrated at the activation gate, indicating an inability to conduct ions. Based on a spontaneous transition observed in extended simulations, and sequence/structure comparison to other Nav channels, we built an alternative π-model featuring a helix transition and the rotation of a conserved asparagine residue into the activation gate. Pore hydration, ion permeation, and state-dependent drug binding in this model were consistent with an open functional state. This work thus offers both a functional annotation of the full-length NavMs structure and a detailed model for a stable Nav open state, with potential conservation in diverse ion-channel families.
Collapse
Affiliation(s)
- Koushik Choudhury
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Solna, Sweden
| | - Marina A. Kasimova
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Solna, Sweden
| | - Sarah McComas
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Rebecca J. Howard
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Lucie Delemotte
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Solna, Sweden,Corresponding author
| |
Collapse
|
13
|
Small molecule modulation of the Drosophila Slo channel elucidated by cryo-EM. Nat Commun 2021; 12:7164. [PMID: 34887422 PMCID: PMC8660915 DOI: 10.1038/s41467-021-27435-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/18/2021] [Indexed: 12/16/2022] Open
Abstract
Slowpoke (Slo) potassium channels display extraordinarily high conductance, are synergistically activated by a positive transmembrane potential and high intracellular Ca2+ concentrations and are important targets for insecticides and antiparasitic drugs. However, it is unknown how these compounds modulate ion translocation and whether there are insect-specific binding pockets. Here, we report structures of Drosophila Slo in the Ca2+-bound and Ca2+-free form and in complex with the fungal neurotoxin verruculogen and the anthelmintic drug emodepside. Whereas the architecture and gating mechanism of Slo channels are conserved, potential insect-specific binding pockets exist. Verruculogen inhibits K+ transport by blocking the Ca2+-induced activation signal and precludes K+ from entering the selectivity filter. Emodepside decreases the conductance by suboptimal K+ coordination and uncouples ion gating from Ca2+ and voltage sensing. Our results expand the mechanistic understanding of Slo regulation and lay the foundation for the rational design of regulators of Slo and other voltage-gated ion channels. Slowpoke (Slo) channels are voltage-gated potassium channels that are activated by high intracellular Ca2+ concentrations, and they are targets for insecticides and antiparasitic drugs. Here, the authors present the cryo-EM structures of the Drosophila melanogaster Slo channel in the Ca2+-bound and Ca2+-free conformations, as well as in complex with the fungal neurotoxin verruculogen and the anthelmintic drug emodepside and discuss the mechanisms by which they affect the activity of Slo.
Collapse
|
14
|
Garratt RC. Protein structure, dynamics, and function-a 20th IUPAB Congress symposium. Biophys Rev 2021; 13:867-869. [PMID: 35059010 PMCID: PMC8724499 DOI: 10.1007/s12551-021-00889-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 10/19/2022] Open
Abstract
A wide range of topics was raised by the four invited speakers who took part in the session on protein structure, dynamics, and function during the 20th IUPAB Congress. Most of the emphasis was placed on understanding the underlying biological phenomena of interest although applications in drug development were also mentioned. For both these purposes, it was clear that a complete description of the dynamics of the system was as important as the structures themselves. The subjects covered included antibiotic peptides, sodium channels, the synthesis of the bacterial cell wall, and protein dynamics using X-FELs.
Collapse
Affiliation(s)
- Richard C. Garratt
- São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil
| |
Collapse
|
15
|
Molecular Characterization of Membrane Steroid Receptors in Hormone-Sensitive Cancers. Cells 2021; 10:cells10112999. [PMID: 34831222 PMCID: PMC8616056 DOI: 10.3390/cells10112999] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 12/20/2022] Open
Abstract
Cancer is one of the most common causes of death worldwide, and its development is a result of the complex interaction of genetic factors, environmental cues, and aging. Hormone-sensitive cancers depend on the action of one or more hormones for their development and progression. Sex steroids and corticosteroids can regulate different physiological functions, including metabolism, growth, and proliferation, through their interaction with specific nuclear receptors, that can transcriptionally regulate target genes via their genomic actions. Therefore, interference with hormones’ activities, e.g., deregulation of their production and downstream pathways or the exposition to exogenous hormone-active substances such as endocrine-disrupting chemicals (EDCs), can affect the regulation of their correlated pathways and trigger the neoplastic transformation. Although nuclear receptors account for most hormone-related biologic effects and their slow genomic responses are well-studied, less-known membrane receptors are emerging for their ability to mediate steroid hormones effects through the activation of rapid non-genomic responses also involved in the development of hormone-sensitive cancers. This review aims to collect pre-clinical and clinical data on these extranuclear receptors not only to draw attention to their emerging role in cancer development and progression but also to highlight their dual role as tumor microenvironment players and potential candidate drug targets.
Collapse
|
16
|
Abstract
Steroid hormones bind receptors in the cell nucleus and in the cell membrane. The most widely studied class of steroid hormone receptors are the nuclear receptors, named for their function as ligand-dependent transcription factors in the cell nucleus. Nuclear receptors, such as estrogen receptor alpha, can also be anchored to the plasma membrane, where they respond to steroids by activating signaling pathways independent of their function as transcription factors. Steroids can also bind integral membrane proteins, such as the G protein-coupled estrogen receptor. Membrane estrogen and progestin receptors have been cloned and characterized in vitro and influence the development and function of many organ systems. Membrane androgen receptors were cloned and characterized in vitro, but their function as androgen receptors in vivo is unresolved. We review the identity and function of membrane proteins that bind estrogens, progestins, and androgens. We discuss evidence that membrane glucocorticoid and mineralocorticoid receptors exist, and whether glucocorticoid and mineralocorticoid nuclear receptors act at the cell membrane. In many cases, integral membrane steroid receptors act independently of nuclear steroid receptors, even though they may share a ligand.
Collapse
Affiliation(s)
- Lindsey S Treviño
- Department of Population Sciences, Division of Health Equities, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Daniel A Gorelick
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence: Daniel A Gorelick, PhD, One Baylor Plaza, Alkek Building N1317.07, Houston, TX, 77030-3411, USA.
| |
Collapse
|
17
|
Ghovanloo MR, Ruben PC. Cannabidiol and Sodium Channel Pharmacology: General Overview, Mechanism, and Clinical Implications. Neuroscientist 2021; 28:318-334. [PMID: 34027742 PMCID: PMC9344566 DOI: 10.1177/10738584211017009] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Voltage-gated sodium (Nav) channels initiate action potentials in excitable tissues. Altering these channels' function can lead to many pathophysiological conditions. Nav channels are composed of several functional and structural domains that could be targeted pharmacologically as potential therapeutic means against various neurological conditions. Mutations in Nav channels have been suggested to underlie various clinical syndromes in different tissues and in association with conditions ranging from epileptic to muscular problems. Treating those mutations that increase the excitability of Nav channels requires inhibitors that could effectively reduce channel firing. The main non-psychotropic constituent of the cannabis plant, cannabidiol (CBD), has recently gained interest as a viable compound to treat some of the conditions that are associated with Nav malfunctions. In this review, we discuss an overview of Nav channels followed by an in-depth description of the interactions of CBD and Nav channels. We conclude with some clinical implications of CBD use against Nav hyperexcitability based on a series of preclinical studies published to date, with a focus on Nav/CBD interactions.
Collapse
Affiliation(s)
- Mohammad-Reza Ghovanloo
- Department of Biomedical Physiology & Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada.,Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA
| | - Peter C Ruben
- Department of Biomedical Physiology & Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
18
|
Structural Pharmacology of Voltage-Gated Sodium Channels. J Mol Biol 2021; 433:166967. [PMID: 33794261 DOI: 10.1016/j.jmb.2021.166967] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/22/2021] [Accepted: 03/22/2021] [Indexed: 12/19/2022]
Abstract
Voltage-gated sodium (NaV) channels initiate and propagate action potentials in excitable tissues to mediate key physiological processes including heart contraction and nervous system function. Accordingly, NaV channels are major targets for drugs, toxins and disease-causing mutations. Recent breakthroughs in cryo-electron microscopy have led to the visualization of human NaV1.1, NaV1.2, NaV1.4, NaV1.5 and NaV1.7 channel subtypes at high-resolution. These landmark studies have greatly advanced our structural understanding of channel architecture, ion selectivity, voltage-sensing, electromechanical coupling, fast inactivation, and the molecular basis underlying NaV channelopathies. NaV channel structures have also been increasingly determined in complex with toxin and small molecule modulators that target either the pore module or voltage sensor domains. These structural studies have provided new insights into the mechanisms of pharmacological action and opportunities for subtype-selective NaV channel drug design. This review will highlight the structural pharmacology of human NaV channels as well as the potential use of engineered and chimeric channels in future drug discovery efforts.
Collapse
|
19
|
Paez Segala MG, Looger LL. Optogenetics. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00092-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|