1
|
González-Ponce KS, Celaya-Herrera S, Mendoza-Acosta MF, Casados-Vázquez LE. Cell-Free Systems and Their Importance in the Study of Membrane Proteins. J Membr Biol 2025:10.1007/s00232-024-00333-0. [PMID: 39760767 DOI: 10.1007/s00232-024-00333-0] [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: 11/05/2024] [Accepted: 12/14/2024] [Indexed: 01/07/2025]
Abstract
The Cell-Free Protein Synthesis (CFPS) is an innovative technique used to produce various proteins. It has several advantages, including short expression times, no strain engineering is required, and toxic proteins such as membrane proteins can be produced. However, the most important advantage is that it eliminates the need for a living cell as a production system. Membrane proteins (MPs) are difficult to express in heterologous strains such as Escherichia coli. Modified strains must be used, and sometimes the strain produces them as inclusion bodies, which makes purification difficult. CFPS can avoid the problem of toxicity and, with the use of additives, allows the production of folded and functional membrane proteins. In this review, we focus on describing what cell-free systems are. We address the advantages and disadvantages of the different organisms that can be used to obtain cell extracts, including PURE systems, where the components are obtained recombinantly, and the methodologies that allow the synthesis of membrane proteins in cell-free systems, which, given their hydrophobic nature, require additives for their correct folding.
Collapse
Affiliation(s)
- Karen Stephania González-Ponce
- Departamento de Alimentos. División de Ciencias de la Vida, Campus Irapuato-Salamanca, Universidad de Guanajuato, Carretera Irapuato Silao km 9, 36500, Irapuato, Guanajuato, México
| | - Samuel Celaya-Herrera
- Departamento de Formación Integral e Institucional, Fraccionamiento Industrial Puerto Interior, Unidad Profesional Interdisciplinaria de Ingeniería Campus Guanajuato, Instituto Politécnico Nacional. Avenida Mineral de Valenciana 200, C.P. 36275, Silao de La Victoria, Guanajuato, México
| | - María Fernanda Mendoza-Acosta
- Posgrado en Biociencias, Campus Irapuato-Salamanca, Universidad de Guanajuato, Carretera Irapuato Silao km 9, 36500, Irapuato, Guanajuato, México
| | - Luz Edith Casados-Vázquez
- Departamento de Alimentos. División de Ciencias de la Vida, Campus Irapuato-Salamanca, Universidad de Guanajuato, Carretera Irapuato Silao km 9, 36500, Irapuato, Guanajuato, México.
- Posgrado en Biociencias, Campus Irapuato-Salamanca, Universidad de Guanajuato, Carretera Irapuato Silao km 9, 36500, Irapuato, Guanajuato, México.
- Investigadoras e Investigadores por México CONAHCYT, Consejo Nacional de Humanidades Ciencias y Tecnologías, Av. Insurgentes Sur 1582, Col. Crédito Constructor, Benito Juárez, 03940, Mexico, México.
| |
Collapse
|
2
|
Hassan MT, Tayara H, Chong KT. Possum: identification and interpretation of potassium ion inhibitors using probabilistic feature vectors. Arch Toxicol 2025; 99:225-235. [PMID: 39438319 DOI: 10.1007/s00204-024-03888-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
The flow of potassium ions through cell membranes plays a crucial role in facilitating various cell processes such as hormone secretion, epithelial function, maintenance of electrochemical gradients, and electrical impulse formation. Potassium ion inhibitors are considered promising alternatives in treating cancer, muscle weakness, renal dysfunction, endocrine disorders, impaired cellular function, and cardiac arrhythmia. Thus, it becomes essential to identify and understand potassium ion inhibitors in order to regulate the ion flow across ion channels. In this study, we created a meta-model, POSSUM, for the identification of potassium ion inhibitors. Two distinct datasets were used for training, testing, and evaluation of the meta-model. We employed seven feature descriptors and five distinctive classifiers to construct 35 baseline models. We used the mean Gini index score to select the optimal base models and classifiers. The POSSUM method was trained on the optimal probabilistic feature vectors. The proposed optimal model, POSSUM, outperforms the baseline models and the existing methods on both datasets. We anticipate POSSUM will be a very useful tool and will be essential in the process of finding and screening possible potassium ion inhibitors.
Collapse
Affiliation(s)
- Mir Tanveerul Hassan
- Department of Electronics and Information Engineering, Jeonbuk National University, Jeonju, 54896, Jeollabuk-do, South Korea
| | - Hilal Tayara
- School of International Engineering and Science, Jeonbuk National University, Jeonju, 54896, Jeollabuk-do, South Korea.
| | - Kil To Chong
- Department of Electronics and Information Engineering, Jeonbuk National University, Jeonju, 54896, Jeollabuk-do, South Korea.
- Advances Electronics and Information Research Centre, Jeonbuk National University, Jeonju, 54896, Jeollabuk-do, South Korea.
| |
Collapse
|
3
|
Lopez-Mateos D, Harris BJ, Hernández-González A, Narang K, Yarov-Yarovoy V. Harnessing Deep Learning Methods for Voltage-Gated Ion Channel Drug Discovery. Physiology (Bethesda) 2025; 40:0. [PMID: 39189871 DOI: 10.1152/physiol.00029.2024] [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: 06/11/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 08/28/2024] Open
Abstract
Voltage-gated ion channels (VGICs) are pivotal in regulating electrical activity in excitable cells and are critical pharmaceutical targets for treating many diseases including cardiac arrhythmia and neuropathic pain. Despite their significance, challenges such as achieving target selectivity persist in VGIC drug development. Recent progress in deep learning, particularly diffusion models, has enabled the computational design of protein binders for any clinically relevant protein based solely on its structure. These developments coincide with a surge in experimental structural data for VGICs, providing a rich foundation for computational design efforts. This review explores the recent advancements in computational protein design using deep learning and diffusion methods, focusing on their application in designing protein binders to modulate VGIC activity. We discuss the potential use of these methods to computationally design protein binders targeting different regions of VGICs, including the pore domain, voltage-sensing domains, and interface with auxiliary subunits. We provide a comprehensive overview of the different design scenarios, discuss key structural considerations, and address the practical challenges in developing VGIC-targeting protein binders. By exploring these innovative computational methods, we aim to provide a framework for developing novel strategies that could significantly advance VGIC pharmacology and lead to the discovery of effective and safe therapeutics.
Collapse
Affiliation(s)
- Diego Lopez-Mateos
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, California, United States
- Biophysics Graduate Group, University of California School of Medicine, Davis, California, United States
| | - Brandon John Harris
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, California, United States
- Biophysics Graduate Group, University of California School of Medicine, Davis, California, United States
| | - Adriana Hernández-González
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, California, United States
- Biophysics Graduate Group, University of California School of Medicine, Davis, California, United States
| | - Kush Narang
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, California, United States
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, California, United States
- Biophysics Graduate Group, University of California School of Medicine, Davis, California, United States
- Department of Anesthesiology and Pain Medicine, University of California School of Medicine, Davis, California, United States
| |
Collapse
|
4
|
Wei S, Shou Z, Yang D, Sun L, Guo Y, Wang Y, Zan X, Li L, Zhang C. Ultra-Long-Term Anti-Inflammatory Polyphenol Capsule to Remodel the Microenvironment for Accelerating Osteoarthritis Healing by Single Dosage. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407425. [PMID: 39556697 DOI: 10.1002/advs.202407425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/03/2024] [Indexed: 11/20/2024]
Abstract
Osteoarthritis (OA) is a common chronic inflammatory disease that leads to disability and death. Existing therapeutic agents often require frequent use, which can lead to drug resistance and long-term side effects. Polyphenols have anti-inflammatory and antioxidant potential. However, they are limited by their short half-life and low bioavailability. This work presents a novel pure polyphenol capsule for sustained release of polyphenols, which is self-assembled via hydrophobic and hydrogen bonds. The capsule enhances cellular uptake, scavenges reactive oxygen and nitrogen species, reduces inflammatory markers, and remodels the OA microenvironment by inhibiting the p38 MAPK pathway. The capsule overcomes the limitations of short half-life and low bioavailability of polyphenols and achieves single-dose cure in mouse and dog OA models, providing an optimal therapeutic window for OA repair. Taking advantage of simple manufacturing, convenient administration, and pure polyphenol composition, these capsules show great potential for clinical treatment of osteoarthritis and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Shaoyin Wei
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325035, China
- Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Zeyu Shou
- Department of Orthopedics, Zhuji Affiliated Hospital of Wenzhou Medical University, Shaoxing, 311800, China
| | - Dong Yang
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325035, China
- Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Linxiao Sun
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yan Guo
- Hunan Provincial Key Laboratory of Advanced Materials for New Energy Storage and Conversion, School of Materials Science and Engineering, Hunan University of Science and Technology, Xiangtan, 411201, China
| | - Yang Wang
- Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Xingjie Zan
- Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Lianxin Li
- Department of Orthopaedics Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Chunwu Zhang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| |
Collapse
|
5
|
Bhuvaneshwari S, Venkataraman K, Sankaranarayanan K. Exploring potential ion channel targets for rheumatoid arthritis: combination of network analysis and gene expression analysis. Biotechnol Appl Biochem 2024; 71:1405-1427. [PMID: 39049164 DOI: 10.1002/bab.2638] [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: 12/28/2023] [Accepted: 06/29/2024] [Indexed: 07/27/2024]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic inflammation of the synovial membrane that leads to the destruction of cartilage and bone. Currently, pharmacological targeting of ion channels is being increasingly recognized as an attractive and feasible strategy for the treatment of RA. The present work employs a network analysis approach to predict the most promising ion channel target for potential RA-treating drugs. A protein-protein interaction map was generated for 343 genes associated with inflammation in RA and ion channel genes using Search Tool for the Retrieval of Interacting Genes and visualized using Cytoscape. Based on the betweenness centrality and traffic values as key topological parameters, 17 hub nodes were identified, including FOS (9800.85), tumor necrosis factor (3654.60), TGFB1 (3305.75), and VEGFA (3052.88). The backbone network constructed with these 17 hub genes was intensely analyzed to identify the most promising ion channel target using network analyzer. Calcium permeating ion channels, especially store-operated calcium entry channels, and their associated regulatory proteins were found to highly interact with RA inflammatory hub genes. This significant ion channel target for RA identified by theoretical and statistical studies was further validated by a pilot case-control gene expression study. Experimental verification of the above findings in 75 RA cases and 25 controls showed increased ORAI1 expression. Thus, with a combination of network analysis approach and gene expression studies, we have explored potential targets for RA treatment.
Collapse
Affiliation(s)
- Sampath Bhuvaneshwari
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology, Anna University, Chennai, India
| | | | - Kavitha Sankaranarayanan
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology, Anna University, Chennai, India
| |
Collapse
|
6
|
Zhu W, Xu Z, Zhang W, Jia Q, Hao H, Gu Y, Zhao Y. Bioinspired Ion Host with Buried and Consecutive Binding Sites for Controlled Ion Dislocation. JACS AU 2024; 4:4415-4422. [PMID: 39610723 PMCID: PMC11600180 DOI: 10.1021/jacsau.4c00752] [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] [Received: 08/17/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 11/30/2024]
Abstract
This study presents a bioinspired ion host featuring continuous binding sites arranged in a tunnel-like structure, closely resembling the selectivity filter of natural ion channels. Our investigation reveals that ions traverse these sites in a controlled, sequential manner due to the structural constraints, effectively mimicking the ion translocation process observed in natural channels. Unlike systems with open binding sites, our model facilitates sequential ion recognition state transitions, enabled by the deliberate design of the tunnel. Notably, we observe dual ion release kinetics, highlighting the system's capacity to maintain ion balance in complex environments and adapt to changing conditions. Additionally, we demonstrate selective binding of two different ions-a challenging task for systems lacking structured tunnels.
Collapse
Affiliation(s)
- Wenjie Zhu
- Key Laboratory
of Fluorine and Nitrogen Chemistry and Advanced Materials, Shanghai Institute of Organic Chemistry, University
of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Ling-Ling Road, Shanghai 200032, China
| | - Zhenchuang Xu
- Key Laboratory
of Fluorine and Nitrogen Chemistry and Advanced Materials, Shanghai Institute of Organic Chemistry, University
of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Ling-Ling Road, Shanghai 200032, China
| | - Wei Zhang
- Key Laboratory
of Fluorine and Nitrogen Chemistry and Advanced Materials, Shanghai Institute of Organic Chemistry, University
of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Ling-Ling Road, Shanghai 200032, China
| | - Qi Jia
- Key Laboratory
of Fluorine and Nitrogen Chemistry and Advanced Materials, Shanghai Institute of Organic Chemistry, University
of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Ling-Ling Road, Shanghai 200032, China
| | - Haoliang Hao
- Key Laboratory
of Fluorine and Nitrogen Chemistry and Advanced Materials, Shanghai Institute of Organic Chemistry, University
of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Ling-Ling Road, Shanghai 200032, China
| | - Yucheng Gu
- Jealott’s
Hill International Research Centre, Syngenta, Bracknell, Berkshire RG42
6EY, U.K.
| | - Yanchuan Zhao
- Key Laboratory
of Fluorine and Nitrogen Chemistry and Advanced Materials, Shanghai Institute of Organic Chemistry, University
of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Ling-Ling Road, Shanghai 200032, China
| |
Collapse
|
7
|
Zhang Y, Ding Y, Zeng Z, Zhu R, Zheng P, Fan S, Cao Q, Chen H, Ren W, Wu M, Wang L, Du J. Intra-channel bi-epitopic crosslinking unleashes ultrapotent antibodies targeting Na V1.7 for pain alleviation. Cell Rep Med 2024; 5:101800. [PMID: 39461335 PMCID: PMC11604545 DOI: 10.1016/j.xcrm.2024.101800] [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: 03/19/2024] [Revised: 09/01/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024]
Abstract
Crucial for cell activities, ion channels are key drug discovery targets. Although small-molecule and peptide modulators dominate ion channel drug discovery, antibodies are emerging as an alternative modality. However, challenges persist in generating potent antibodies, especially for channels with limited extracellular epitopes. We herein present a bi-epitopic crosslinking strategy to overcome these challenges, focusing on NaV1.7, a potential analgesic target. Aiming to crosslink two non-overlapping epitopes on voltage-sensing domains II and IV, we construct bispecific antibodies and ligand-antibody conjugates. Enhanced affinity and potency are observed in comparison to the monospecific controls. Among them, a ligand-antibody conjugate (1080-PEG7-ACDTB) displays a two-orders-of-magnitude improvement in potency (IC50 of 0.06 ± 0.01 nM) and over 1,000-fold selectivity for NaV1.7. Additionally, this conjugate demonstrates robust analgesic effects in mouse pain models. Our study introduces an approach to developing effective antibodies against NaV1.7, thereby initiating a promising direction for the advancement of pain therapeutics.
Collapse
Affiliation(s)
- Yaning Zhang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China; Peking University-Tsinghua University-National Institute Biological Sciences (PTN) Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yanchao Ding
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Ziyan Zeng
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Rui Zhu
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Peiyuan Zheng
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Shilong Fan
- The Technology Center for Protein Sciences, Tsinghua University, Beijing 100084, China
| | - Qingjuan Cao
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Hang Chen
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Weishuai Ren
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Mengling Wu
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Luyao Wang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Juanjuan Du
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
8
|
Huang J, Pan X, Yan N. Structural biology and molecular pharmacology of voltage-gated ion channels. Nat Rev Mol Cell Biol 2024; 25:904-925. [PMID: 39103479 DOI: 10.1038/s41580-024-00763-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2024] [Indexed: 08/07/2024]
Abstract
Voltage-gated ion channels (VGICs), including those for Na+, Ca2+ and K+, selectively permeate ions across the cell membrane in response to changes in membrane potential, thus participating in physiological processes involving electrical signalling, such as neurotransmission, muscle contraction and hormone secretion. Aberrant function or dysregulation of VGICs is associated with a diversity of neurological, psychiatric, cardiovascular and muscular disorders, and approximately 10% of FDA-approved drugs directly target VGICs. Understanding the structure-function relationship of VGICs is crucial for our comprehension of their working mechanisms and role in diseases. In this Review, we discuss how advances in single-particle cryo-electron microscopy have afforded unprecedented structural insights into VGICs, especially on their interactions with clinical and investigational drugs. We present a comprehensive overview of the recent advances in the structural biology of VGICs, with a focus on how prototypical drugs and toxins modulate VGIC activities. We explore how these structures elucidate the molecular basis for drug actions, reveal novel pharmacological sites, and provide critical clues to future drug discovery.
Collapse
Affiliation(s)
- Jian Huang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Xiaojing Pan
- Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, Guangdong, China.
| | - Nieng Yan
- Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, Guangdong, China.
- Beijing Frontier Research Center for Biological Structure, Tsinghua-Peking Joint Center for Life Sciences, State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, China.
| |
Collapse
|
9
|
Stephens AD, Wilkinson T. Discovery of Therapeutic Antibodies Targeting Complex Multi-Spanning Membrane Proteins. BioDrugs 2024; 38:769-794. [PMID: 39453540 PMCID: PMC11530565 DOI: 10.1007/s40259-024-00682-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 10/26/2024]
Abstract
Complex integral membrane proteins, which are embedded in the cell surface lipid bilayer by multiple transmembrane spanning polypeptides, encompass families of proteins that are important target classes for drug discovery. These protein families include G protein-coupled receptors, ion channels, transporters, enzymes, and adhesion molecules. The high specificity of monoclonal antibodies and the ability to engineer their properties offers a significant opportunity to selectively bind these target proteins, allowing direct modulation of pharmacology or enabling other mechanisms of action such as cell killing. Isolation of antibodies that bind these types of membrane proteins and exhibit the desired pharmacological function has, however, remained challenging due to technical issues in preparing membrane protein antigens suitable for enabling and driving antibody drug discovery strategies. In this article, we review progress and emerging themes in defining discovery strategies for a generation of antibodies that target these complex membrane protein antigens. We also comment on how this field may develop with the emerging implementation of computational techniques, artificial intelligence, and machine learning.
Collapse
Affiliation(s)
- Amberley D Stephens
- Department of Biologics Engineering, Oncology R&D, The Discovery Centre, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0AA, UK
| | - Trevor Wilkinson
- Department of Biologics Engineering, Oncology R&D, The Discovery Centre, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0AA, UK.
| |
Collapse
|
10
|
Centonze E, Kellenberger S. Voltage-clamp fluorometry for advancing mechanistic understanding of ion channel mechanisms with a focus on acid-sensing ion channels. Biochem Soc Trans 2024; 52:2167-2177. [PMID: 39400205 PMCID: PMC11555705 DOI: 10.1042/bst20240165] [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: 07/23/2024] [Revised: 09/13/2024] [Accepted: 09/20/2024] [Indexed: 10/15/2024]
Abstract
Voltage-clamp fluorometry (VCF) has revolutionized the study of ion channels by combining electrophysiology with fluorescence spectroscopy. VCF allows ion channel researchers to link dynamic structural changes, measured in real time, to function. Acid-sensing ion channels (ASICs) are Na+-permeable non-voltage-gated ion channels of the central and peripheral nervous system. They function as pH sensors, triggering neuronal excitation when pH decreases. Animal studies have shown the importance of ASICs for pain and fear sensation, learning, and neurodegeneration following ischaemic stroke. This review explores the technical bases and various developments of VCF, including fluorescence resonance energy transfer and the use of unnatural fluorescent amino acids. We provide an overview of VCF applications with a focus on ASICs, detailing how VCF has unveiled proton-induced conformational changes in key regions such as the acid pocket, wrist, and pore, crucial for understanding transitions between closed, open, and desensitized states.
Collapse
Affiliation(s)
- Eleonora Centonze
- Department of Biomedical Sciences, University of Lausanne, 1011 Lausanne, Switzerland
| | - Stephan Kellenberger
- Department of Biomedical Sciences, University of Lausanne, 1011 Lausanne, Switzerland
| |
Collapse
|
11
|
Shi W, Zhao Q, Gao H, Yang C, Tan Z, Li N, Jiang F, Wang H, Ji Y, Zhou Y. Involvement of BK Channels and Ryanodine Receptors in Salicylate-induced Tinnitus. Mol Neurobiol 2024:10.1007/s12035-024-04533-6. [PMID: 39397241 DOI: 10.1007/s12035-024-04533-6] [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: 12/17/2023] [Accepted: 10/04/2024] [Indexed: 10/15/2024]
Abstract
Neural hyperexcitability of the central auditory system is a key pathological characteristic of tinnitus, but its underlying molecular mechanisms remain elusive. The large-conductance Ca2+-activated K+ channel (BK) plays a crucial role in down- or upregulating neuronal activity. This study aims to investigate the role of BK channels in mediating tinnitus-associated neural hyperexcitability and elucidate the mechanisms behind it. Immunofluorescent staining revealed extensive expression of the BK channels on neurons within the central auditory system of rats. After long-term systemic administration of salicylate, a stable tinnitus inducer, we observed a significant change in the expression levels of BKα and β4 subunits in the rat central auditory system. In addition, salicylate was found to enhance the outward potassium currents mediated by the BK channel when exogenously expressed in HEK293 cells. Interestingly, this effect could be blocked by ryanodine, a potent inhibitor of ryanodine receptors (RyRs). Molecular docking identified Gln4020 within the central domain of RyR as a key residue in RyR-salicylate interactions. The results indicated that salicylate might directly activate RyRs leading to Ca2+ release from endoplasmic reticulum, and increased BK currents subsequently. Systemic treatment with paxilline, a potent blocker of BK channel, selectively reversed the increased P4/P1 amplitude ratios in the frequency region of tinnitus perception induced by single-dose salicylate administration. These results suggest that BK channels and ryanodine receptors may play a selective role in salicylate-induced tinnitus.
Collapse
Affiliation(s)
- Wenying Shi
- School of Basic Medical Sciences, Hebei University, Baoding, 071000, China
| | - Qi Zhao
- School of Basic Medical Sciences, Hebei University, Baoding, 071000, China
| | - Hongwei Gao
- School of Basic Medical Sciences, Hebei University, Baoding, 071000, China
| | - Chao Yang
- Shanghai Chongming Xinhua Translational Medical Institute for Cancer Pain, Shanghai, 202150, China
| | - Zhiyong Tan
- School of Basic Medical Sciences, Hebei University, Baoding, 071000, China
| | - Na Li
- School of Basic Medical Sciences, Hebei University, Baoding, 071000, China
| | - Feng Jiang
- Shanghai Chongming Xinhua Translational Medical Institute for Cancer Pain, Shanghai, 202150, China
| | - Hongjie Wang
- School of Basic Medical Sciences, Hebei University, Baoding, 071000, China
| | - Yonghua Ji
- School of Basic Medical Sciences, Hebei University, Baoding, 071000, China
- Shanghai Chongming Xinhua Translational Medical Institute for Cancer Pain, Shanghai, 202150, China
| | - You Zhou
- School of Basic Medical Sciences, Hebei University, Baoding, 071000, China.
| |
Collapse
|
12
|
Gertsch J, Chicca A. CNS Drug Discovery in Academia: Where Basic Research Meets Innovation. Chembiochem 2024; 25:e202400397. [PMID: 38958639 DOI: 10.1002/cbic.202400397] [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] [Revised: 05/27/2024] [Indexed: 07/04/2024]
Abstract
The involvement of academic research in drug discovery is consistently growing. However, academic projects seldom advance to clinical trials. Here, we assess the landscape of drug discovery within the National Centre of Competence in Research (NCCR) TransCure launched by the Swiss National Science Foundation to foster basic research and early-stage drug discovery on membrane transporters. This included transporters in central nervous system (CNS) disorders, which represent a huge unmet medical need. While idea championship, sustainable funding, collaborations between disciplines at the interface of academia and industry are important for translational research, Popperian falsifiability, strong intellectual property and a motivated startup team are key elements for innovation. This is exemplified by the NCCR TransCure spin-off company Synendos Therapeutics, a clinical stage biotech company developing the first selective endocannabinoid reuptake inhibitors (SERIs) as novel treatment for neuropsychiatric disorders. We provide a perspective on the challenges related to entering an uncharted druggable space and bridging the often mentioned "valley of death". The high attrition rate of drug discovery projects in the CNS field within academia is often due to the lack of meaningful animal models that can provide pharmacological proof-of-concept for potentially disruptive technologies at the earliest stages, and the absence of solid intellectual property.
Collapse
Affiliation(s)
- Jürg Gertsch
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012, Bern, Switzerland
| | - Andrea Chicca
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012, Bern, Switzerland
- Synendos Therapeutics, Barfüsserplatz, 3, 4051, Basel, Switzerland
| |
Collapse
|
13
|
Kwak J, Kim W, Cho H, Han J, Sim SJ, Song HG, Pak Y, Song HS. Label-free optical detection of calcium ion influx in cell-derived nanovesicles using a conical Au/PDMS biosensor. LAB ON A CHIP 2024; 24:4138-4146. [PMID: 39072370 DOI: 10.1039/d4lc00421c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Ion channels, which are key to physiological regulation and drug discovery, control ion flux across membranes, and their dysregulation leads to various diseases. Ca2+ monitoring is crucial for cellular signaling when performing Ca-based assays in ion channel research; these assays are widely utilized in both academic and pharmaceutical contexts for drug screening and pharmacological profiling. However, existing detection methods are limited by slow detection speeds, low throughput, complex processes, and low analyte viability. In this study, we developed a label-free optical biosensing method using a conical Au/polydimethylsiloxane platform tailored to detect Ca2+ influx in A549-originated nanovesicles facilitated by the transient receptor potential ankyrin 1 (TRPA1) channel. Nanovesicles expressing cellular signaling components mimic TRPA1 signal transduction in cell membranes and improve analyte viability. The conical Au/polydimethylsiloxane sensor converted Ca2+ influx events induced by specific agonist exposure into noticeable changes in relative transmittance under visible light. The optical transmittance change accompanying Ca2+ influx resulted in an enhanced sensing response, high accuracy and reliability, and rapid detection (∼5 s) without immobilization or ligand treatments. In the underlying sensing mechanism, morphological variations in nanovesicles, which depend on Ca2+ influx, induce a considerable light scattering change at an interface between the nanovesicle and Au, revealed by optical simulation. This study provides a foundation for developing biosensors based on light-matter interactions. These sensors are simple and cost-effective with superior performance and diverse functionality.
Collapse
Affiliation(s)
- Jisung Kwak
- Sensor System Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Woochul Kim
- Sensor System Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hyerim Cho
- Sensor System Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Jiyun Han
- Center of Water Cycle Research, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Sang Jun Sim
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hyun Gyu Song
- Sensor System Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Yusin Pak
- Sensor System Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hyun Seok Song
- Sensor System Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| |
Collapse
|
14
|
Chow CY, King GF. Shining a Light on Venom-Peptide Receptors: Venom Peptides as Targeted Agents for In Vivo Molecular Imaging. Toxins (Basel) 2024; 16:307. [PMID: 39057947 PMCID: PMC11281729 DOI: 10.3390/toxins16070307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Molecular imaging has revolutionised the field of biomedical research by providing a non-invasive means to visualise and understand biochemical processes within living organisms. Optical fluorescent imaging in particular allows researchers to gain valuable insights into the dynamic behaviour of a target of interest in real time. Ion channels play a fundamental role in cellular signalling, and they are implicated in diverse pathological conditions, making them an attractive target in the field of molecular imaging. Many venom peptides exhibit exquisite selectivity and potency towards ion channels, rendering them ideal agents for molecular imaging applications. In this review, we illustrate the use of fluorescently-labelled venom peptides for disease diagnostics and intraoperative imaging of brain tumours and peripheral nerves. Finally, we address challenges for the development and clinical translation of venom peptides as nerve-targeted imaging agents.
Collapse
Affiliation(s)
- Chun Yuen Chow
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
- Australia Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Glenn F. King
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
- Australia Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, St. Lucia, QLD 4072, Australia
| |
Collapse
|
15
|
Piga M, Varga Z, Feher A, Papp F, Korpos E, Bangera KC, Frlan R, Ilaš J, Dernovšek J, Tomašič T, Zidar N. Identification of a Novel Structural Class of H V1 Inhibitors by Structure-Based Virtual Screening. J Chem Inf Model 2024; 64:4850-4862. [PMID: 38850237 PMCID: PMC11200261 DOI: 10.1021/acs.jcim.4c00240] [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: 02/09/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/10/2024]
Abstract
The human voltage-gated proton channel, hHV1, is highly expressed in various cell types including macrophages, B lymphocytes, microglia, sperm cells and also in various cancer cells. Overexpression of HV1 has been shown to promote tumor formation by highly metastatic cancer cells, and has been associated with neuroinflammatory diseases, immune response disorders and infertility, suggesting a potential use of hHV1 inhibitors in numerous therapeutic areas. To identify compounds targeting this channel, we performed a structure-based virtual screening on an open structure of the human HV1 channel. Twenty selected virtual screening hits were tested on Chinese hamster ovary (CHO) cells transiently expressing hHV1, with compound 13 showing strong block of the proton current with an IC50 value of 8.5 μM. Biological evaluation of twenty-three additional analogs of 13 led to the discovery of six other compounds that blocked the proton current by more than 50% at 50 μM concentration. This allowed for an investigation of structure-activity relationships. The antiproliferative activity of the selected promising hHV1 inhibitors was investigated in the cell lines MDA-MB-231 and THP-1, where compound 13 inhibited growth with an IC50 value of 9.0 and 8.1 μM, respectively. The identification of a new structural class of HV1 inhibitors contributes to our understanding of the structural requirements for inhibition of this ion channel and opens up the possibility of investigating the role of HV1 inhibitors in various pathological conditions and in cancer therapy.
Collapse
Affiliation(s)
- Martina Piga
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Zoltan Varga
- Faculty
of Medicine, University of Debrecen, Egyetem tér 1, Debrecen H-4032, Hungary
| | - Adam Feher
- Faculty
of Medicine, University of Debrecen, Egyetem tér 1, Debrecen H-4032, Hungary
| | - Ferenc Papp
- Faculty
of Medicine, University of Debrecen, Egyetem tér 1, Debrecen H-4032, Hungary
| | - Eva Korpos
- Faculty
of Medicine, University of Debrecen, Egyetem tér 1, Debrecen H-4032, Hungary
- HUN-REN−UD
Cell Biology and Signaling Research Group, Egyetem tér 1, Debrecen H-4032, Hungary
| | - Kavya C. Bangera
- Faculty
of Medicine, University of Debrecen, Egyetem tér 1, Debrecen H-4032, Hungary
| | - Rok Frlan
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Janez Ilaš
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Jaka Dernovšek
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Tihomir Tomašič
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Nace Zidar
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| |
Collapse
|
16
|
Cen J, Hu N, Shen J, Gao Y, Lu H. Pathological Functions of Lysosomal Ion Channels in the Central Nervous System. Int J Mol Sci 2024; 25:6565. [PMID: 38928271 PMCID: PMC11203704 DOI: 10.3390/ijms25126565] [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: 04/03/2024] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Lysosomes are highly dynamic organelles that maintain cellular homeostasis and regulate fundamental cellular processes by integrating multiple metabolic pathways. Lysosomal ion channels such as TRPML1-3, TPC1/2, ClC6/7, CLN7, and TMEM175 mediate the flux of Ca2+, Cl-, Na+, H+, and K+ across lysosomal membranes in response to osmotic stimulus, nutrient-dependent signals, and cellular stresses. These ion channels serve as the crucial transducers of cell signals and are essential for the regulation of lysosomal biogenesis, motility, membrane contact site formation, and lysosomal homeostasis. In terms of pathophysiology, genetic variations in these channel genes have been associated with the development of lysosomal storage diseases, neurodegenerative diseases, inflammation, and cancer. This review aims to discuss the current understanding of the role of these ion channels in the central nervous system and to assess their potential as drug targets.
Collapse
Affiliation(s)
| | | | | | - Yongjing Gao
- Institute of Pain Medicine and Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China; (J.C.); (N.H.); (J.S.)
| | - Huanjun Lu
- Institute of Pain Medicine and Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China; (J.C.); (N.H.); (J.S.)
| |
Collapse
|
17
|
Harris BJ, Nguyen PT, Zhou G, Wulff H, DiMaio F, Yarov-Yarovoy V. Toward high-resolution modeling of small molecule-ion channel interactions. Front Pharmacol 2024; 15:1411428. [PMID: 38919257 PMCID: PMC11196768 DOI: 10.3389/fphar.2024.1411428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/13/2024] [Indexed: 06/27/2024] Open
Abstract
Ion channels are critical drug targets for a range of pathologies, such as epilepsy, pain, itch, autoimmunity, and cardiac arrhythmias. To develop effective and safe therapeutics, it is necessary to design small molecules with high potency and selectivity for specific ion channel subtypes. There has been increasing implementation of structure-guided drug design for the development of small molecules targeting ion channels. We evaluated the performance of two RosettaLigand docking methods, RosettaLigand and GALigandDock, on the structures of known ligand-cation channel complexes. Ligands were docked to voltage-gated sodium (NaV), voltage-gated calcium (CaV), and transient receptor potential vanilloid (TRPV) channel families. For each test case, RosettaLigand and GALigandDock methods frequently sampled a ligand-binding pose within a root mean square deviation (RMSD) of 1-2 Å relative to the experimental ligand coordinates. However, RosettaLigand and GALigandDock scoring functions cannot consistently identify experimental ligand coordinates as top-scoring models. Our study reveals that the proper scoring criteria for RosettaLigand and GALigandDock modeling of ligand-ion channel complexes should be assessed on a case-by-case basis using sufficient ligand and receptor interface sampling, knowledge about state-specific interactions of the ion channel, and inherent receptor site flexibility that could influence ligand binding.
Collapse
Affiliation(s)
- Brandon J. Harris
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
- Biophysics Graduate Group, University of California, Davis, Davis, CA, United States
| | - Phuong T. Nguyen
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
| | - Guangfeng Zhou
- Department of Biochemistry, University of Washington, Seattle, WA, United States
- Institute for Protein Design, University of Washington, Seattle, WA, United States
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Frank DiMaio
- Department of Biochemistry, University of Washington, Seattle, WA, United States
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
- Biophysics Graduate Group, University of California, Davis, Davis, CA, United States
- Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
18
|
Tabassum S, Shorter S, Ovsepian SV. Analysis of the action mechanisms and targets of herbal anticonvulsants highlights opportunities for therapeutic engagement with refractory epilepsy. J Mol Med (Berl) 2024; 102:761-771. [PMID: 38653825 PMCID: PMC11106186 DOI: 10.1007/s00109-024-02445-5] [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: 12/11/2023] [Revised: 03/26/2024] [Accepted: 04/05/2024] [Indexed: 04/25/2024]
Abstract
Epilepsy is a neurological disorder characterized by spontaneous and recurring seizures. It poses significant therapeutic challenges due to diverse etiology, pathobiology, and pharmacotherapy-resistant variants. The anticonvulsive effects of herbal leads with biocompatibility and toxicity considerations have attracted much interest, inspiring mechanistic analysis with the view of their use for engagement of new targets and combination with antiseizure pharmacotherapies. This article presents a comprehensive overview of the key molecular players and putative action mechanisms of the most common antiepileptic herbals demonstrated in tissue culture and preclinical models. From the review of the literature, it emerges that their effects are mediated via five distinct mechanisms: (1) reduction of membrane excitability through inhibition of cation channels, (2) improvement of mitochondrial functions with antioxidant effects, (3) enhancement in synaptic transmission mediated by GABAA receptors, (4) improvement of immune response with anti-inflammatory action, and (5) suppression of protein synthesis and metabolism. While some of the primary targets and action mechanisms of herbal anticonvulsants (1, 3) are shared with antiseizure pharmacotherapies, herbal leads also engage with distinct mechanisms (2, 4, and 5), suggesting new drug targets and opportunities for their integration with antiseizure medications. Addressing outstanding questions through research and in silico modeling should facilitate the future use of herbals as auxiliary therapy in epilepsy and guide the development of treatment of pharmacoresistant seizures through rigorous trials and regulatory approval.
Collapse
Affiliation(s)
- Sobia Tabassum
- Department of Biological Sciences, Faculty of Sciences, International Islamic University, Islamabad, Pakistan
| | - Susan Shorter
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, ME4 4TB, UK
| | - Saak V Ovsepian
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, ME4 4TB, UK.
- Faculty of Medicine, Tbilisi State University, Tbilisi, 0177, Republic of Georgia.
| |
Collapse
|
19
|
Rödström KEJ, Cloake A, Sörmann J, Baronina A, Smith KHM, Pike ACW, Ang J, Proks P, Schewe M, Holland-Kaye I, Bushell SR, Elliott J, Pardon E, Baukrowitz T, Owens RJ, Newstead S, Steyaert J, Carpenter EP, Tucker SJ. Extracellular modulation of TREK-2 activity with nanobodies provides insight into the mechanisms of K2P channel regulation. Nat Commun 2024; 15:4173. [PMID: 38755204 PMCID: PMC11099193 DOI: 10.1038/s41467-024-48536-2] [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: 12/15/2023] [Accepted: 05/03/2024] [Indexed: 05/18/2024] Open
Abstract
Potassium channels of the Two-Pore Domain (K2P) subfamily, KCNK1-KCNK18, play crucial roles in controlling the electrical activity of many different cell types and represent attractive therapeutic targets. However, the identification of highly selective small molecule drugs against these channels has been challenging due to the high degree of structural and functional conservation that exists not only between K2P channels, but across the whole K+ channel superfamily. To address the issue of selectivity, here we generate camelid antibody fragments (nanobodies) against the TREK-2 (KCNK10) K2P K+ channel and identify selective binders including several that directly modulate channel activity. X-ray crystallography and CryoEM data of these nanobodies in complex with TREK-2 also reveal insights into their mechanisms of activation and inhibition via binding to the extracellular loops and Cap domain, as well as their suitability for immunodetection. These structures facilitate design of a biparatropic inhibitory nanobody with markedly improved sensitivity. Together, these results provide important insights into TREK channel gating and provide an alternative, more selective approach to modulation of K2P channel activity via their extracellular domains.
Collapse
Affiliation(s)
- Karin E J Rödström
- Clarendon Laboratory, Department of Physics, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
- Centre for Medicines Discovery, University of Oxford, Oxford, UK
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Alexander Cloake
- Clarendon Laboratory, Department of Physics, University of Oxford, Oxford, UK
| | - Janina Sörmann
- Clarendon Laboratory, Department of Physics, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Agnese Baronina
- Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Kathryn H M Smith
- Clarendon Laboratory, Department of Physics, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Ashley C W Pike
- Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Jackie Ang
- Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Peter Proks
- Clarendon Laboratory, Department of Physics, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Marcus Schewe
- Institute of Physiology, Medical Faculty, Kiel University, Kiel, Germany
| | | | - Simon R Bushell
- Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Jenna Elliott
- Clarendon Laboratory, Department of Physics, University of Oxford, Oxford, UK
| | - Els Pardon
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
- VIB-VUB Center for Structural Biology, VIB, Brussels, Belgium
| | - Thomas Baukrowitz
- Institute of Physiology, Medical Faculty, Kiel University, Kiel, Germany
| | - Raymond J Owens
- The Rosalind Franklin Institute, Harwell Campus, Didcot, UK
- Division of Structural Biology, University of Oxford, Oxford, UK
| | - Simon Newstead
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
- Department of Biochemistry, University of Oxford, Oxford, UK
- OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford, UK
| | - Jan Steyaert
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
- VIB-VUB Center for Structural Biology, VIB, Brussels, Belgium
| | - Elisabeth P Carpenter
- Centre for Medicines Discovery, University of Oxford, Oxford, UK.
- OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford, UK.
| | - Stephen J Tucker
- Clarendon Laboratory, Department of Physics, University of Oxford, Oxford, UK.
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK.
- OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford, UK.
| |
Collapse
|
20
|
Wang Q, Ye Y, Yang L, Xiao L, Liu J, Zhang W, Du G. Painful diabetic neuropathy: The role of ion channels. Biomed Pharmacother 2024; 173:116417. [PMID: 38490158 DOI: 10.1016/j.biopha.2024.116417] [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: 11/30/2023] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 03/17/2024] Open
Abstract
Painful diabetic neuropathy (PDN) is a common chronic complication of diabetes that causes neuropathic pain and negatively affects the quality of life. The management of PDN is far from satisfactory. At present, interventions are primarily focused on symptomatic treatment. Ion channel disorders are a major cause of PDN, and a complete understanding of their roles and mechanisms may provide better options for the clinical treatment of PDN. Therefore, this review summarizes the important role of ion channels in PDN and the current drug development targeting these ion channels.
Collapse
Affiliation(s)
- Qi Wang
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Centre, West China Hospital, Sichuan University, Chengdu, China; National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yifei Ye
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Centre, West China Hospital, Sichuan University, Chengdu, China; National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Linghui Yang
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Centre, West China Hospital, Sichuan University, Chengdu, China; National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Lifan Xiao
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Centre, West China Hospital, Sichuan University, Chengdu, China; National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Liu
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Centre, West China Hospital, Sichuan University, Chengdu, China; National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Wensheng Zhang
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Centre, West China Hospital, Sichuan University, Chengdu, China; National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China.
| | - Guizhi Du
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Centre, West China Hospital, Sichuan University, Chengdu, China; National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
21
|
Huang J, Korsunsky A, Yazdani M, Chen J. Targeting TRP channels: recent advances in structure, ligand binding, and molecular mechanisms. Front Mol Neurosci 2024; 16:1334370. [PMID: 38273937 PMCID: PMC10808746 DOI: 10.3389/fnmol.2023.1334370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Abstract
Transient receptor potential (TRP) channels are a large and diverse family of transmembrane ion channels that are widely expressed, have important physiological roles, and are associated with many human diseases. These proteins are actively pursued as promising drug targets, benefitting greatly from advances in structural and mechanistic studies of TRP channels. At the same time, the complex, polymodal activation and regulation of TRP channels have presented formidable challenges. In this short review, we summarize recent progresses toward understanding the structural basis of TRP channel function, as well as potential ligand binding sites that could be targeted for therapeutics. A particular focus is on the current understanding of the molecular mechanisms of TRP channel activation and regulation, where many fundamental questions remain unanswered. We believe that a deeper understanding of the functional mechanisms of TRP channels will be critical and likely transformative toward developing successful therapeutic strategies targeting these exciting proteins. This endeavor will require concerted efforts from computation, structural biology, medicinal chemistry, electrophysiology, pharmacology, drug safety and clinical studies.
Collapse
Affiliation(s)
- Jian Huang
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| | - Aron Korsunsky
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| | - Mahdieh Yazdani
- Modeling and Informatics, Merck & Co., Inc., West Point, PA, United States
| | - Jianhan Chen
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| |
Collapse
|
22
|
Rajendran R, Krishnan R, Kim JO, Oh MJ. Regulatory effects of potassium channel blockers on potassium channel genes upon nervous necrosis virus infection in sevenband grouper Hyporthodus septumfasciatus. Gene 2024; 890:147815. [PMID: 37739197 DOI: 10.1016/j.gene.2023.147815] [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: 06/26/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023]
Abstract
Ion channels in fishes regulate the flow of important ions that play an active role in the excitation and transmission of impulses through neuronal cells. Specific housekeeping genes translates into proteins and selectively permeabilize and facilitate ion crossover transmissions. Potassium (K+) channels play a crucial role in a wide range of functions such as cell volume regulation, hormone secretion, synaptic transmission and muscle contraction. The dysfunction of ion channels result in channelopathies, which hinder critical cellular activities. Recent studies have indicated that viral pathogens tend to regulate cellular ion channels for entry into host cells. Hence, the present study aimed to elucidate the role of K+ channels during nervous necrosis virus (NNV) infections in the sevenband grouper (Hyporthodus septumfasciatus). Real-time PCR with the standardized potassium genes revealed the downregulation of potassium two pore domain channel subfamily member - KCNK10, KCNK9, KCNK2, and KCNK1 genes post infection at both 17 °C and 25 °C whereas an upregulation was noted in the case of gill tissues. SMART analysis revealed a transmembrane region in all genes. Multiple sequence alignment using MultAlin and phylogenetic analysis revealed true homology of potassium genes with other higher vertebrates. In vitro and in vivo challenge study of NNV using Tetra ethyl ammonium (TEA) as potential drug showed inverse relation to that of viral replication and a corresponding downregulation of K+ channel gene expression was observed which was further confirmed by an immunofluorescence assay. These findings indicate that K+ channels play a crucial role during viral infection. Moreover, the observed downregulation can be related to rapid endocytosis resulting from recycling endosomes during a viral infection. Hence, further studies are warranted to better understand the role of K+ channel genes during NNV infection.
Collapse
Affiliation(s)
- Rahul Rajendran
- Department of Aqualife Medicine, Chonnam National University, Yeosu, 50626, Republic of Korea
| | - Rahul Krishnan
- Department of Aquatic Animal Health Management, Faculty of Fisheries, Kerala University of Fisheries and Ocean Studies, Ernakulam, Kerala, India
| | - Jong-Oh Kim
- Department of Microbiology, Pukyong National University, Busan, Republic of Korea
| | - Myung-Joo Oh
- Department of Aqualife Medicine, Chonnam National University, Yeosu, 50626, Republic of Korea.
| |
Collapse
|
23
|
Shang W, Zhu L, Li Z, Xu W, Xiong B, Liu Y, Tang KW, Qian PC, Yin SF, Wong WY. Ruthenium-Catalyzed 1,6-Hydroalkylation of para-Quinone Methides with Ketones via the in Situ Activation of C( sp3)-H Bonds. J Org Chem 2023; 88:16196-16215. [PMID: 37955519 DOI: 10.1021/acs.joc.3c01661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
A simple and efficient method for the ruthenium-catalyzed 1,6-hydroalkylation of para-quinone methides (p-QMs) with ketones via the in situ activation of C(sp3)-H bonds has been disclosed. Without the need for preactivation of the substrates and oxidant, a broad range of p-QMs and ketones are well tolerated, producing the expected 1,6-hydroalkylation products with moderate to good yields. Step-by-step control experiments and DFT calculation were conducted systematically to gain insights for the plausible reaction mechanism. This finding may have potential application in the selective diarylmethylation of ketones at the α-C position in organic synthesis.
Collapse
Affiliation(s)
- Wenli Shang
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, P. R. China
| | - Longzhi Zhu
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, P. R. China
| | - Zikang Li
- Department of Applied Biology and Chemical Technology and Research Institute for Smart Energy, The Hong Kong Polytechnic University, Hung Hom, Hong Kong 999077, P. R. China
| | - Weifeng Xu
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, P. R. China
| | - Biquan Xiong
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, P. R. China
- Department of Applied Biology and Chemical Technology and Research Institute for Smart Energy, The Hong Kong Polytechnic University, Hung Hom, Hong Kong 999077, P. R. China
| | - Yu Liu
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, P. R. China
| | - Ke-Wen Tang
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, P. R. China
| | - Peng-Cheng Qian
- Key Laboratory of Environmental Functional Materials Technology and Application of Wenzhou City, Institute of New Materials & Industry Technology, College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325035, P. R. China
| | - Shuang-Feng Yin
- Advanced Catalytic Engineering Research Center of the Ministry of Education, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
- College of Science, Central South University of Forestry and Technology, Changsha 410004, P. R. China
| | - Wai-Yeung Wong
- Department of Applied Biology and Chemical Technology and Research Institute for Smart Energy, The Hong Kong Polytechnic University, Hung Hom, Hong Kong 999077, P. R. China
| |
Collapse
|
24
|
Dewaker V, Sharma AR, Debnath U, Park ST, Kim HS. Insights from molecular dynamics simulations of TRPV1 channel modulators in pain. Drug Discov Today 2023; 28:103798. [PMID: 37838068 DOI: 10.1016/j.drudis.2023.103798] [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: 07/28/2023] [Revised: 09/28/2023] [Accepted: 10/09/2023] [Indexed: 10/16/2023]
Abstract
TRPV1 is a nonselective cation channel vital for detecting noxious stimuli (heat, acid, capsaicin). Its role in pain makes it a potential drug target for chronic pain management, migraines, and related disorders. This review updates molecular dynamics (MD) simulation studies on the TRPV1 channel, focusing on its gating mechanism, ligand-binding sites, and implications for drug design. The article also explores challenges in developing modulators, SAR optimization, and clinical trial studies. Efforts have been undertaken to concisely present MD simulation findings, with a focus on their relevance to drug discovery.
Collapse
Affiliation(s)
- Varun Dewaker
- Institute of New Frontier Research Team, Hallym University, Chuncheon-si 24252, Gangwon-do, Republic of Korea
| | - Ashish R Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si 24252, Gangwon-do, Republic of Korea
| | - Utsab Debnath
- School of Health Sciences & Technology, UPES, Dehradun, Uttarakhand 248007, India
| | - Sung Taek Park
- Institute of New Frontier Research Team, Hallym University, Chuncheon-si 24252, Gangwon-do, Republic of Korea; Department of Obstetrics and Gynecology, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea; EIONCELL Inc., Chuncheon 24252, Republic of Korea
| | - Hyeong Su Kim
- Institute of New Frontier Research Team, Hallym University, Chuncheon-si 24252, Gangwon-do, Republic of Korea; Division of Hemato-Oncology, Department of Internal Medicine, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea; EIONCELL Inc., Chuncheon 24252, Republic of Korea.
| |
Collapse
|
25
|
Papadourakis M, Sinenka H, Matricon P, Hénin J, Brannigan G, Pérez-Benito L, Pande V, van Vlijmen H, de Graaf C, Deflorian F, Tresadern G, Cecchini M, Cournia Z. Alchemical Free Energy Calculations on Membrane-Associated Proteins. J Chem Theory Comput 2023; 19:7437-7458. [PMID: 37902715 PMCID: PMC11017255 DOI: 10.1021/acs.jctc.3c00365] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Indexed: 10/31/2023]
Abstract
Membrane proteins have diverse functions within cells and are well-established drug targets. The advances in membrane protein structural biology have revealed drug and lipid binding sites on membrane proteins, while computational methods such as molecular simulations can resolve the thermodynamic basis of these interactions. Particularly, alchemical free energy calculations have shown promise in the calculation of reliable and reproducible binding free energies of protein-ligand and protein-lipid complexes in membrane-associated systems. In this review, we present an overview of representative alchemical free energy studies on G-protein-coupled receptors, ion channels, transporters as well as protein-lipid interactions, with emphasis on best practices and critical aspects of running these simulations. Additionally, we analyze challenges and successes when running alchemical free energy calculations on membrane-associated proteins. Finally, we highlight the value of alchemical free energy calculations calculations in drug discovery and their applicability in the pharmaceutical industry.
Collapse
Affiliation(s)
- Michail Papadourakis
- Biomedical
Research Foundation, Academy of Athens, 4 Soranou Ephessiou, 11527 Athens, Greece
| | - Hryhory Sinenka
- Institut
de Chimie de Strasbourg, UMR7177, CNRS, Université de Strasbourg, F-67083 Strasbourg Cedex, France
| | - Pierre Matricon
- Sosei
Heptares, Steinmetz Building,
Granta Park, Great Abington, Cambridge CB21 6DG, United
Kingdom
| | - Jérôme Hénin
- Laboratoire
de Biochimie Théorique UPR 9080, CNRS and Université Paris Cité, 75005 Paris, France
| | - Grace Brannigan
- Center
for Computational and Integrative Biology, Rutgers University−Camden, Camden, New Jersey 08103, United States of America
- Department
of Physics, Rutgers University−Camden, Camden, New Jersey 08102, United States
of America
| | - Laura Pérez-Benito
- CADD,
In Silico Discovery, Janssen Research &
Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Vineet Pande
- CADD,
In Silico Discovery, Janssen Research &
Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Herman van Vlijmen
- CADD,
In Silico Discovery, Janssen Research &
Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Chris de Graaf
- Sosei
Heptares, Steinmetz Building,
Granta Park, Great Abington, Cambridge CB21 6DG, United
Kingdom
| | - Francesca Deflorian
- Sosei
Heptares, Steinmetz Building,
Granta Park, Great Abington, Cambridge CB21 6DG, United
Kingdom
| | - Gary Tresadern
- CADD,
In Silico Discovery, Janssen Research &
Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Marco Cecchini
- Institut
de Chimie de Strasbourg, UMR7177, CNRS, Université de Strasbourg, F-67083 Strasbourg Cedex, France
| | - Zoe Cournia
- Biomedical
Research Foundation, Academy of Athens, 4 Soranou Ephessiou, 11527 Athens, Greece
| |
Collapse
|
26
|
Bian Y, Tuo J, He L, Li W, Li S, Chu H, Zhao Y. Voltage-gated sodium channels in cancer and their specific inhibitors. Pathol Res Pract 2023; 251:154909. [PMID: 37939447 DOI: 10.1016/j.prp.2023.154909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/10/2023]
Abstract
Voltage-gated sodium channels (VGSCs) participate in generating and spreading action potentials in electrically excited cells such as neurons and muscle fibers. Abnormal expression of VGSCs has been observed in various types of tumors, while they are either not expressed or expressed at a low level in the matching normal tissue. Hence, this abnormal expression suggests that VGSCs confer some advantage or viability on tumor cells, making them a valuable indicator for identifying tumor cells. In addition, overexpression of VGSCs increased the ability of cancer cells to metastasize and invade, as well as correlated with the metastatic behavior of different cancers. Therefore, blocking VGSCs presents a new strategy for the treatment of cancers. A portion of this review summarizes the structure and function of VGSCs and also describes the correlation between VGSCs and cancers. Most importantly, we provide an overview of current research on various subtype-selective VGSC inhibitors and updates on ongoing clinical studies.
Collapse
Affiliation(s)
- Yuan Bian
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Jiale Tuo
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Liangpeng He
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Wenwen Li
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Shangxiao Li
- School of Medical Devices, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, PR China
| | - Huiying Chu
- Laboratory of Molecular Modeling and Design, State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yongshan Zhao
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China.
| |
Collapse
|
27
|
Li MR, Luo XJ, Peng J. Role of sonic hedgehog signaling pathway in the regulation of ion channels: focus on its association with cardio-cerebrovascular diseases. J Physiol Biochem 2023; 79:719-730. [PMID: 37676576 DOI: 10.1007/s13105-023-00982-0] [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: 01/19/2023] [Accepted: 08/25/2023] [Indexed: 09/08/2023]
Abstract
Sonic hedgehog (SHH) signaling is vital for cell differentiation and proliferation during embryonic development, yet its role in cardiac, cerebral, and vascular pathophysiology is under debate. Recent studies have demonstrated that several compounds of SHH signaling regulate ion channels, which in turn affect the behavior of target cells. Some of these ion channels are involved in the cardio-cerebrovascular system. Here, we first reviewed the SHH signaling cascades, then its interaction with ion channels, and their impact on cardio-cerebrovascular diseases. Considering the complex cross talk of SHH signaling with other pathways that also affect ion channels and their potential impact on the cardio-cerebrovascular system, we highlight the necessity of thoroughly studying the effect of SHH signaling on ion homeostasis, which could serve as a novel mechanism for cardio-cerebrovascular diseases. Activation of SHH signaling influence ion channels activity, which in turn influence ion homeostasis, membrane potential, and electrophysiology, could serve as a novel strategy for cardio-cerebrovascular diseases.
Collapse
Affiliation(s)
- Ming-Rui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
| |
Collapse
|
28
|
Silva MP, Rodrigues CG, Machado DC, Nogueira RA. Long-term memory in Staphylococcus aureus α-hemolysin ion channel kinetics. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2023; 52:661-671. [PMID: 37542583 DOI: 10.1007/s00249-023-01675-8] [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: 06/20/2022] [Revised: 07/03/2023] [Accepted: 07/20/2023] [Indexed: 08/07/2023]
Abstract
The kinetics of an ion channel are classically understood as a random process. However, studies have shown that in complex ion channels, formed by multiple subunits, this process can be deterministic, presenting long-term memory. Staphylococcus aureus α-hemolysin (α-HL) is a toxin that acts as the major factor in Staphylococcus aureus virulence. α-HL is a water-soluble protein capable of forming ion channels into lipid bilayers, by insertion of an amphipathic β-barrel. Here, the α-HL was used as an experimental model to study memory in ion channel kinetics. We applied the approximate entropy (ApEn) approach to analyze randomness and the Detrended Fluctuation Analysis (DFA) to investigate the existence of long memory in α-HL channel kinetics. Single-channel currents were measured through experiments with α-HL channels incorporated in planar lipid bilayers. All experiments were carried out under the following conditions: 1 M NaCl solution, pH 4.5; transmembrane potential of + 40 mV and temperature 25 ± 1 °C. Single-channel currents were recorded in real-time in the memory of a microcomputer coupled to an A/D converter and a patch-clamp amplifier. The conductance value of the α-HL channels was 0.82 ± 0.0025 nS (n = 128). The DFA analysis showed that the kinetics of α-HL channels presents long-term memory ([Formula: see text] = 0.63 ± 0.04). The ApEn outcomes showed low complexity to dwell times when open (ApEno = 0.5514 ± 0.28) and closed (ApEnc = 0.1145 ± 0.08), corroborating the results of the DFA method.
Collapse
Affiliation(s)
- M P Silva
- Department of Animal Morphology and Physiology, Federal Rural University of Pernambuco, Recife, Pernambuco, Brazil
| | - C G Rodrigues
- Department of Biophysics and Radiobiology, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - D C Machado
- Department of Biophysics and Radiobiology, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - R A Nogueira
- Department of Animal Morphology and Physiology, Federal Rural University of Pernambuco, Recife, Pernambuco, Brazil.
| |
Collapse
|
29
|
Huang J, Fan X, Jin X, Teng L, Yan N. Dual-pocket inhibition of Na v channels by the antiepileptic drug lamotrigine. Proc Natl Acad Sci U S A 2023; 120:e2309773120. [PMID: 37782796 PMCID: PMC10576118 DOI: 10.1073/pnas.2309773120] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/01/2023] [Indexed: 10/04/2023] Open
Abstract
Voltage-gated sodium (Nav) channels govern membrane excitability, thus setting the foundation for various physiological and neuronal processes. Nav channels serve as the primary targets for several classes of widely used and investigational drugs, including local anesthetics, antiepileptic drugs, antiarrhythmics, and analgesics. In this study, we present cryogenic electron microscopy (cryo-EM) structures of human Nav1.7 bound to two clinical drugs, riluzole (RLZ) and lamotrigine (LTG), at resolutions of 2.9 Å and 2.7 Å, respectively. A 3D EM reconstruction of ligand-free Nav1.7 was also obtained at 2.1 Å resolution. RLZ resides in the central cavity of the pore domain and is coordinated by residues from repeats III and IV. Whereas one LTG molecule also binds to the central cavity, the other is found beneath the intracellular gate, known as site BIG. Therefore, LTG, similar to lacosamide and cannabidiol, blocks Nav channels via a dual-pocket mechanism. These structures, complemented with docking and mutational analyses, also explain the structure-activity relationships of the LTG-related linear 6,6 series that have been developed for improved efficacy and subtype specificity on different Nav channels. Our findings reveal the molecular basis for these drugs' mechanism of action and will aid the development of novel antiepileptic and pain-relieving drugs.
Collapse
Affiliation(s)
- Jian Huang
- Department of Molecular Biology, Princeton University, Princeton, NJ08544
| | - Xiao Fan
- Department of Molecular Biology, Princeton University, Princeton, NJ08544
| | - Xueqin Jin
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing100084, China
| | - Liming Teng
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing100084, China
| | - Nieng Yan
- Department of Molecular Biology, Princeton University, Princeton, NJ08544
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing100084, China
- Shenzhen Medical Academy of Research and Translation, Shenzhen, Guangdong Province518107, China
| |
Collapse
|
30
|
Dong W, Fekete A, Chen X, Liu H, Beilhartz GL, Chen X, Bahrampour S, Xiong Y, Yang Q, Zhao H, Kong T, Morioka MS, Jung G, Kim JE, Schramek D, Dirks PB, Song Y, Kim TH, He Y, Wanggou S, Li X, Melnyk RA, Wang LY, Huang X. A designer peptide against the EAG2-Kvβ2 potassium channel targets the interaction of cancer cells and neurons to treat glioblastoma. NATURE CANCER 2023; 4:1418-1436. [PMID: 37697045 DOI: 10.1038/s43018-023-00626-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/02/2023] [Indexed: 09/13/2023]
Abstract
Glioblastoma (GBM) is an incurable brain cancer that lacks effective therapies. Here we show that EAG2 and Kvβ2, which are predominantly expressed by GBM cells at the tumor-brain interface, physically interact to form a potassium channel complex due to a GBM-enriched Kvβ2 isoform. In GBM cells, EAG2 localizes at neuron-contacting regions in a Kvβ2-dependent manner. Genetic knockdown of the EAG2-Kvβ2 complex decreases calcium transients of GBM cells, suppresses tumor growth and invasion and extends the survival of tumor-bearing mice. We engineered a designer peptide to disrupt EAG2-Kvβ2 interaction, thereby mitigating tumor growth in patient-derived xenograft and syngeneic mouse models across GBM subtypes without overt toxicity. Neurons upregulate chemoresistant genes in GBM cells in an EAG2-Kvβ2-dependent manner. The designer peptide targets neuron-associated GBM cells and possesses robust efficacy in treating temozolomide-resistant GBM. Our findings may lead to the next-generation therapeutic agent to benefit patients with GBM.
Collapse
Affiliation(s)
- Weifan Dong
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Adam Fekete
- Neurosciences and Mental Health Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Xiaodi Chen
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Hongwei Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Greg L Beilhartz
- Molecular Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Xin Chen
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Shahrzad Bahrampour
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Yi Xiong
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Qi Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Hongyu Zhao
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Tian Kong
- Neurosciences and Mental Health Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Malia S Morioka
- Macaulay Honors College, City College of New York, New York, NY, USA
- Advanced Science Research Center at the Graduate Center, City University of New York, New York, NY, USA
| | - Geena Jung
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ji-Eun Kim
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Daniel Schramek
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Peter B Dirks
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Yuanquan Song
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tae-Hee Kim
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Ye He
- Macaulay Honors College, City College of New York, New York, NY, USA
- Advanced Science Research Center at the Graduate Center, City University of New York, New York, NY, USA
| | - Siyi Wanggou
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Roman A Melnyk
- Molecular Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Lu-Yang Wang
- Neurosciences and Mental Health Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Xi Huang
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada.
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
31
|
Eskandari N, Gentile S. Potassium channels activity unveils cancer vulnerability. CURRENT TOPICS IN MEMBRANES 2023; 92:1-14. [PMID: 38007264 DOI: 10.1016/bs.ctm.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
"No cell could exist without ion channels" (Clay Armstrong; 1999). Since the discovery in the early 1950s, that ions move across biological membranes, the idea that changes of ionic gradients can generate biological signals has fascinated scientists in any fields. Soon later (1960s) it was found that ionic flows were controlled by a class of specific and selective proteins called ion channels. Thus, it became clear that the concerted activities of these proteins can initiate, arrest, and finely tune a variety of biochemical cascades which offered the opportunity to better understand both biology and pathology. Cancer is a disease that is notoriously difficult to treat due its heterogeneous nature which makes it the deadliest disease in the developed world. Recently, emerging evidence has established that potassium channels are critical modulators of several hallmarks of cancer including tumor growth, metastasis, and metabolism. Nevertheless, the role of potassium ion channels in cancer biology and the therapeutic potential offered by targeting these proteins has not been explored thoroughly. This chapter is addressed to both cancer biologists and ion channels scientists and it aims to shine a light on the established and potential roles of potassium ion channels in cancer biology and on the therapeutic benefit of targeting potassium channels with activator molecules.
Collapse
Affiliation(s)
- Najmeh Eskandari
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
| | - Saverio Gentile
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
32
|
Lu HJ, Wu XB, Wei QQ. Ion channels in cancer-induced bone pain: from molecular mechanisms to clinical applications. Front Mol Neurosci 2023; 16:1239599. [PMID: 37664239 PMCID: PMC10469682 DOI: 10.3389/fnmol.2023.1239599] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023] Open
Abstract
Cancer-induced bone pain (CIBP) caused by bone metastasis is one of the most prevalent diseases, and current treatments rely primarily on opioids, which have significant side effects. However, recent developments in pharmaceutical science have identified several new mechanisms for CIBP, including the targeted modification of certain ion channels and receptors. Ion channels are transmembrane proteins, which are situated on biological cell membranes, which facilitate passive transport of inorganic ions across membranes. They are involved in various physiological processes, including transmission of pain signals in the nervous system. In recent years, there has been an increasing interest in the role of ion channels in chronic pain, including CIBP. Therefore, in this review, we summarize the current literature on ion channels, related receptors, and drugs and explore the mechanism of CIBP. Targeting ion channels and regulating their activity might be key to treating pain associated with bone cancer and offer new treatment avenues.
Collapse
Affiliation(s)
- Huan-Jun Lu
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, China
| | - Xiao-Bo Wu
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, China
| | - Qian-Qi Wei
- Department of Infectious Diseases, General Hospital of Tibet Military Command, Xizang, China
| |
Collapse
|
33
|
Pliushcheuskaya P, Künze G. Recent Advances in Computer-Aided Structure-Based Drug Design on Ion Channels. Int J Mol Sci 2023; 24:ijms24119226. [PMID: 37298178 DOI: 10.3390/ijms24119226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/16/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Ion channels play important roles in fundamental biological processes, such as electric signaling in cells, muscle contraction, hormone secretion, and regulation of the immune response. Targeting ion channels with drugs represents a treatment option for neurological and cardiovascular diseases, muscular degradation disorders, and pathologies related to disturbed pain sensation. While there are more than 300 different ion channels in the human organism, drugs have been developed only for some of them and currently available drugs lack selectivity. Computational approaches are an indispensable tool for drug discovery and can speed up, especially, the early development stages of lead identification and optimization. The number of molecular structures of ion channels has considerably increased over the last ten years, providing new opportunities for structure-based drug development. This review summarizes important knowledge about ion channel classification, structure, mechanisms, and pathology with the main focus on recent developments in the field of computer-aided, structure-based drug design on ion channels. We highlight studies that link structural data with modeling and chemoinformatic approaches for the identification and characterization of new molecules targeting ion channels. These approaches hold great potential to advance research on ion channel drugs in the future.
Collapse
Affiliation(s)
- Palina Pliushcheuskaya
- Institute for Drug Discovery, Medical Faculty, University of Leipzig, Brüderstr. 34, D-04103 Leipzig, Germany
| | - Georg Künze
- Institute for Drug Discovery, Medical Faculty, University of Leipzig, Brüderstr. 34, D-04103 Leipzig, Germany
- Interdisciplinary Center for Bioinformatics, University of Leipzig, Härtelstr. 16-18, D-04107 Leipzig, Germany
| |
Collapse
|
34
|
Patel OV, Partridge C, Plaut K. Space Environment Impacts Homeostasis: Exposure to Spaceflight Alters Mammary Gland Transportome Genes. Biomolecules 2023; 13:biom13050872. [PMID: 37238741 DOI: 10.3390/biom13050872] [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: 03/09/2023] [Revised: 04/22/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Membrane transporters and ion channels that play an indispensable role in metabolite trafficking have evolved to operate in Earth's gravity. Dysregulation of the transportome expression profile at normogravity not only affects homeostasis along with drug uptake and distribution but also plays a key role in the pathogenesis of diverse localized to systemic diseases including cancer. The profound physiological and biochemical perturbations experienced by astronauts during space expeditions are well-documented. However, there is a paucity of information on the effect of the space environment on the transportome profile at an organ level. Thus, the goal of this study was to analyze the effect of spaceflight on ion channels and membrane substrate transporter genes in the periparturient rat mammary gland. Comparative gene expression analysis revealed an upregulation (p < 0.01) of amino acid, Ca2+, K+, Na+, Zn2+, Cl-, PO43-, glucose, citrate, pyruvate, succinate, cholesterol, and water transporter genes in rats exposed to spaceflight. Genes associated with the trafficking of proton-coupled amino acids, Mg2+, Fe2+, voltage-gated K+-Na+, cation-coupled chloride, as well as Na+/Ca2+ and ATP-Mg/Pi exchangers were suppressed (p < 0.01) in these spaceflight-exposed rats. These findings suggest that an altered transportome profile contributes to the metabolic modulations observed in the rats exposed to the space environment.
Collapse
Affiliation(s)
- Osman V Patel
- Cell and Molecular Biology Department, Grand Valley State University, Allendale, MI 49401, USA
| | - Charlyn Partridge
- Annis Water Resources Institute, Grand Valley State University, Muskegon, MI 49441, USA
| | - Karen Plaut
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47906, USA
| |
Collapse
|
35
|
Shi S, Ma B, Ji Q, Guo S, An H, Ye S. Identification of a druggable pocket of the calcium-activated chloride channel TMEM16A in its open state. J Biol Chem 2023:104780. [PMID: 37142220 DOI: 10.1016/j.jbc.2023.104780] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/06/2023] Open
Abstract
The calcium-activated chloride channel TMEM16A is a potential drug target to treat hypertension, secretory diarrhea, and several cancers. However, all reported TMEM16A structures are either closed or desensitized, and direct inhibition of the open state by drug molecules lacks a reliable structural basis. Therefore, revealing the druggable pocket of TMEM16A exposed in the open state is important for understanding protein-ligand interactions and facilitating rational drug design. Here, we reconstructed the calcium-activated open conformation of TMEM16A using an enhanced sampling algorithm and segmental modeling. Furthermore, we identified an open state druggable pocket and screened a potent TMEM16A inhibitor, etoposide, which is a derivative of a traditional herbal monomer. Molecular simulations and site-directed mutagenesis showed that etoposide binds to the open state of TMEM16A, thereby blocking the ion conductance pore of the channel. Finally, we demonstrated that etoposide can target TMEM16A to inhibit the proliferation of prostate cancer PC-3 cells. Together, these findings provide a deep understanding of the TMEM16A open state at an atomic level and identify pockets for the design of novel inhibitors with broad applications in chloride channel biology, biophysics, and medicinal chemistry.
Collapse
Affiliation(s)
- Sai Shi
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Biao Ma
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Hebei University of Technology, Tianjin 300401, China
| | - Qiushuang Ji
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, China
| | - Shuai Guo
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China.
| | - Hailong An
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Hebei University of Technology, Tianjin 300401, China.
| | - Sheng Ye
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
36
|
Yazdani K, Jordan D, Yang M, Fullenkamp CR, Calabrese DR, Boer R, Hilimire T, Allen TEH, Khan RT, Schneekloth JS. Machine Learning Informs RNA-Binding Chemical Space. Angew Chem Int Ed Engl 2023; 62:e202211358. [PMID: 36584293 PMCID: PMC9992102 DOI: 10.1002/anie.202211358] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 01/01/2023]
Abstract
Small molecule targeting of RNA has emerged as a new frontier in medicinal chemistry, but compared to the protein targeting literature our understanding of chemical matter that binds to RNA is limited. In this study, we reported Repository Of BInders to Nucleic acids (ROBIN), a new library of nucleic acid binders identified by small molecule microarray (SMM) screening. The complete results of 36 individual nucleic acid SMM screens against a library of 24 572 small molecules were reported (including a total of 1 627 072 interactions assayed). A set of 2 003 RNA-binding small molecules was identified, representing the largest fully public, experimentally derived library of its kind to date. Machine learning was used to develop highly predictive and interpretable models to characterize RNA-binding molecules. This work demonstrates that machine learning algorithms applied to experimentally derived sets of RNA binders are a powerful method to inform RNA-targeted chemical space.
Collapse
Affiliation(s)
- Kamyar Yazdani
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201, USA
| | - Deondre Jordan
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201, USA
| | - Mo Yang
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201, USA
| | - Christopher R. Fullenkamp
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201, USA
| | - David R. Calabrese
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201, USA
| | - Robert Boer
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201, USA
| | - Thomas Hilimire
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201, USA
| | | | | | - John S. Schneekloth
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201, USA
| |
Collapse
|
37
|
Di Maio G, Villano I, Ilardi CR, Messina A, Monda V, Iodice AC, Porro C, Panaro MA, Chieffi S, Messina G, Monda M, La Marra M. Mechanisms of Transmission and Processing of Pain: A Narrative Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:3064. [PMID: 36833753 PMCID: PMC9964506 DOI: 10.3390/ijerph20043064] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/27/2023] [Accepted: 02/07/2023] [Indexed: 06/18/2023]
Abstract
Knowledge about the mechanisms of transmission and the processing of nociceptive information, both in healthy and pathological states, has greatly expanded in recent years. This rapid progress is due to a multidisciplinary approach involving the simultaneous use of different branches of study, such as systems neurobiology, behavioral analysis, genetics, and cell and molecular techniques. This narrative review aims to clarify the mechanisms of transmission and the processing of pain while also taking into account the characteristics and properties of nociceptors and how the immune system influences pain perception. Moreover, several important aspects of this crucial theme of human life will be discussed. Nociceptor neurons and the immune system play a key role in pain and inflammation. The interactions between the immune system and nociceptors occur within peripheral sites of injury and the central nervous system. The modulation of nociceptor activity or chemical mediators may provide promising novel approaches to the treatment of pain and chronic inflammatory disease. The sensory nervous system is fundamental in the modulation of the host's protective response, and understanding its interactions is pivotal in the process of revealing new strategies for the treatment of pain.
Collapse
Affiliation(s)
- Girolamo Di Maio
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Ines Villano
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Ciro Rosario Ilardi
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Department of Psychology, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy
| | - Antonietta Messina
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Vincenzo Monda
- Department of Movement Sciences and Wellbeing, University of Naples “Parthenope”, 80133 Naples, Italy
| | - Ashlei Clara Iodice
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Chiara Porro
- Department of Clinical and Experimental Medicine, University of Foggia, Viale Pinto, 71100 Foggia, Italy
| | - Maria Antonietta Panaro
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy
| | - Sergio Chieffi
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Giovanni Messina
- Department of Clinical and Experimental Medicine, University of Foggia, Viale Pinto, 71100 Foggia, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Marco La Marra
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| |
Collapse
|
38
|
He S, Lim GE. The Application of High-Throughput Approaches in Identifying Novel Therapeutic Targets and Agents to Treat Diabetes. Adv Biol (Weinh) 2023; 7:e2200151. [PMID: 36398493 DOI: 10.1002/adbi.202200151] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/04/2022] [Indexed: 11/19/2022]
Abstract
During the past decades, unprecedented progress in technologies has revolutionized traditional research methodologies. Among these, advances in high-throughput drug screening approaches have permitted the rapid identification of potential therapeutic agents from drug libraries that contain thousands or millions of molecules. Moreover, high-throughput-based therapeutic target discovery strategies can comprehensively interrogate relationships between biomolecules (e.g., gene, RNA, and protein) and diseases and significantly increase the authors' knowledge of disease mechanisms. Diabetes is a chronic disease primarily characterized by the incapacity of the body to maintain normoglycemia. The prevalence of diabetes in modern society has become a severe public health issue that threatens the well-being of millions of patients. Although a number of pharmacological treatments are available, there is no permanent cure for diabetes, and discovering novel therapeutic targets and agents continues to be an urgent need. The present review discusses the technical details of high-throughput screening approaches in drug discovery, followed by introducing the applications of such approaches to diabetes research. This review aims to provide an example of the applicability of high-throughput technologies in facilitating different aspects of disease research.
Collapse
Affiliation(s)
- Siyi He
- Department of Medicine, Université de Montréal, Pavillon Roger-Gaudry, 2900 Edouard Montpetit Blvd, Montreal, Québec, H3T 1J4, Canada.,Cardiometabolic Axis, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue St Denis, Montreal, Québec, H2X 0A9, Canada
| | - Gareth E Lim
- Department of Medicine, Université de Montréal, Pavillon Roger-Gaudry, 2900 Edouard Montpetit Blvd, Montreal, Québec, H3T 1J4, Canada.,Cardiometabolic Axis, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue St Denis, Montreal, Québec, H2X 0A9, Canada
| |
Collapse
|
39
|
Davies A, Tomas A. Appreciating the potential for GPCR crosstalk with ion channels. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 195:101-120. [PMID: 36707150 DOI: 10.1016/bs.pmbts.2022.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
G protein-coupled receptors (GPCRs) are expressed by most tissues in the body and are exploited pharmacologically in a variety of pathological conditions including diabetes, cardiovascular disease, neurological diseases, and cancers. Numerous cell signaling pathways can be regulated by GPCR activation, depending on the specific GPCR, ligand and cell type. Ion channels are among the many effector proteins downstream of these signaling pathways. Saliently, ion channels are also recognized as druggable targets, and there is evidence that their activity may regulate GPCR function via membrane potential and cytoplasmic ion concentration. Overall, there appears to be a large potential for crosstalk between ion channels and GPCRs. This might have implications not only for targeting GPCRs for drug development, but also opens the possibility of co-targeting them with ion channels to achieve improved therapeutic outcomes. In this review, we highlight the large variety of possible GPCR-ion channel crosstalk modes.
Collapse
Affiliation(s)
- Amy Davies
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Alejandra Tomas
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom.
| |
Collapse
|
40
|
Srinivasan SR. Targeting Circuit Abnormalities in Neurodegenerative Disease. Mol Pharmacol 2023; 103:38-44. [PMID: 36310030 DOI: 10.1124/molpharm.122.000563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 02/03/2023] Open
Abstract
Despite significant improvement in our ability to diagnose both common and rare neurodegenerative diseases and understand their underlying biologic mechanisms, there remains a disproportionate lack of effective treatments, reflecting the complexity of these disorders. Successfully advancing novel treatments for neurodegenerative disorders will require reconsideration of traditional approaches, which to date have focused largely on specific disease proteins or cells of origin. This article proposes reframing these diseases as conditions of dysfunctional circuitry as a complement to ongoing efforts. Specifically reviewed is how aberrant spiking is a common downstream mechanism in numerous neurodegenerative diseases, often driven by dysfunction in specific ion channels. Surgical modification of this electrical activity via deep brain stimulation is already an approved modality for many of these disorders. Therefore, restoring proper electrical activity by targeting these channels pharmacologically represents a viable strategy for intervention, not only for symptomatic management but also as a potential disease-modifying therapy. Such an approach is likely to be a promising route to treating these devastating disorders, either as monotherapy or in conjunction with current drugs. SIGNIFICANCE STATEMENT: Despite extensive research and improved understanding of the biology driving neurodegenerative disease, there has not been a concomitant increase in approved therapies. Accordingly, it is time to shift our perspective and recognize these diseases also as disorders of circuitry to further yield novel drug targets and new interventions. An approach focused on treating dysfunctional circuitry has the potential to reduce or reverse patient symptoms and potentially modify disease course.
Collapse
|
41
|
Nishiguchi R, Tanaka T, Hayashida J, Nakagita T, Zhou W, Takeda H. Evaluation of Cell-Free Synthesized Human Channel Proteins for In Vitro Channel Research. MEMBRANES 2022; 13:48. [PMID: 36676855 PMCID: PMC9861611 DOI: 10.3390/membranes13010048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 06/17/2023]
Abstract
Despite channel proteins being important drug targets, studies on channel proteins remain limited, as the proteins are difficult to express and require correct complex formation within membranes. Although several in vitro synthesized recombinant channels have been reported, considering the vast diversity of the structures and functions of channel proteins, it remains unclear which classes of channels cell-free synthesis can be applied to. In this study, we synthesized 250 clones of human channels, including ion channel pore-forming subunits, gap junction proteins, porins, and regulatory subunits, using a wheat cell-free membrane protein production system, and evaluated their synthetic efficiency and function. Western blotting confirmed that 95% of the channels were successfully synthesized, including very large channels with molecular weights of over 200 kDa. A subset of 47 voltage-gated potassium ion channels was further analyzed using a planar lipid bilayer assay, out of which 80% displayed a voltage-dependent opening in the assay. We co-synthesized KCNB1 and KCNS3, a known heteromeric complex pair, and demonstrated that these channels interact on a liposome. These results indicate that cell-free protein synthesis provides a promising solution for channel studies to overcome the bottleneck of in vitro protein production.
Collapse
Affiliation(s)
- Rei Nishiguchi
- Proteo-Science Center, Ehime University, Bunkyocho 3, Matsuyama 790-8577, Ehime, Japan
| | - Toyohisa Tanaka
- Proteo-Science Center, Ehime University, Bunkyocho 3, Matsuyama 790-8577, Ehime, Japan
| | - Jun Hayashida
- Nissan Chemical Corporation, Shiraoka 1470, Shiraoka 349-0294, Saitama, Japan
| | - Tomoya Nakagita
- Proteo-Science Center, Ehime University, Bunkyocho 3, Matsuyama 790-8577, Ehime, Japan
| | - Wei Zhou
- Proteo-Science Center, Ehime University, Bunkyocho 3, Matsuyama 790-8577, Ehime, Japan
| | - Hiroyuki Takeda
- Proteo-Science Center, Ehime University, Bunkyocho 3, Matsuyama 790-8577, Ehime, Japan
| |
Collapse
|
42
|
Paul D, Nedelcu AM. The underexplored links between cancer and the internal body climate: Implications for cancer prevention and treatment. Front Oncol 2022; 12:1040034. [PMID: 36620608 PMCID: PMC9815514 DOI: 10.3389/fonc.2022.1040034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/25/2022] [Indexed: 12/24/2022] Open
Abstract
In order to effectively manage and cure cancer we should move beyond the general view of cancer as a random process of genetic alterations leading to uncontrolled cell proliferation or simply a predictable evolutionary process involving selection for traits that increase cell fitness. In our view, cancer is a systemic disease that involves multiple interactions not only among cells within tumors or between tumors and surrounding tissues but also with the entire organism and its internal "milieu". We define the internal body climate as an emergent property resulting from spatial and temporal interactions among internal components themselves and with the external environment. The body climate itself can either prevent, promote or support cancer initiation and progression (top-down effect; i.e., body climate-induced effects on cancer), as well as be perturbed by cancer (bottom-up effect; i.e., cancer-induced body climate changes) to further favor cancer progression and spread. This positive feedback loop can move the system towards a "cancerized" organism and ultimately results in its demise. In our view, cancer not only affects the entire system; it is a reflection of an imbalance of the entire system. This model provides an integrated framework to study all aspects of cancer as a systemic disease, and also highlights unexplored links that can be altered to both prevent body climate changes that favor cancer initiation, progression and dissemination as well as manipulate or restore the body internal climate to hinder the success of cancer inception, progression and metastasis or improve therapy outcomes. To do so, we need to (i) identify cancer-relevant factors that affect specific climate components, (ii) develop 'body climate biomarkers', (iii) define 'body climate scores', and (iv) develop strategies to prevent climate changes, stop or slow the changes, or even revert the changes (climate restoration).
Collapse
Affiliation(s)
- Doru Paul
- Weill Cornell Medicine, New York, NY, United States
| | - Aurora M. Nedelcu
- Biology Department, University of New Brunswick, Fredericton, NB, Canada
| |
Collapse
|
43
|
Sabat M, Raveglia LF, Aldegheri L, Barilli A, Bianchi F, Brault L, Brodbeck D, Feriani A, Lingard I, Miura J, Myers R, Piccoli L, Tassini S, Tyhonas J, Ton-Nu T, Wang H, Virginio C. The discovery of (1R, 3R)-1-(3-chloro-5-fluorophenyl)-3-(hydroxymethyl)-1,2,3,4-tetrahydroisoquinoline-6-carbonitrile, a potent and selective agonist of human transient receptor potential cation channel subfamily m member 5 (TRPM5) and evaluation of as a potential gastrointestinal prokinetic agent. Bioorg Med Chem 2022; 76:117084. [PMID: 36402081 DOI: 10.1016/j.bmc.2022.117084] [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: 09/15/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/08/2022]
Abstract
This publication details the discovery of a series of selective transient receptor potential cation channel subfamily M member 5 (TRPM5) agonists culminating with the identification of the lead compound (1R, 3R)-1-(3-chloro-5-fluorophenyl)-3-(hydroxymethyl)-1,2,3,4-tetrahydroisoquinoline-6-carbonitrile (39). We describe herein our biological rationale for agonism of the target, the examination of the then current literature tool molecules, and finally the process of our discovery starting with a high throughput screening hit through lead development. We also detail the selectivity of the lead compound 39 versus related family members TRPA1, TRPV1, TRPV4, TRPM4 and TRPM8, the drug metabolism and pharmacokinetics (DMPK) profile and in vivo efficacy in a mouse model of gastrointestinal motility.
Collapse
Affiliation(s)
- M Sabat
- Turning Point Therapeutics, 10628 Science Center Drive, Suite 200, San Diego, CA 92121, United States.
| | - L F Raveglia
- Aptuit (Verona) Srl, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - L Aldegheri
- Aptuit (Verona) Srl, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - A Barilli
- Aptuit (Verona) Srl, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy.
| | - F Bianchi
- Aptuit (Verona) Srl, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - L Brault
- Aptuit (Verona) Srl, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - D Brodbeck
- Aptuit (Verona) Srl, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - A Feriani
- Aptuit (Verona) Srl, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - I Lingard
- Aptuit (Verona) Srl, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - J Miura
- Takeda California, 9625 Towne Centre Dr, San Diego, CA 92121, United States
| | - R Myers
- Takeda California, 9625 Towne Centre Dr, San Diego, CA 92121, United States
| | - L Piccoli
- Aptuit (Verona) Srl, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - S Tassini
- Aptuit (Verona) Srl, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - J Tyhonas
- Kinnate Biopharma Inc, 3611 Valley Centre Drive, Suite 175, San Diego, CA 92130, United States
| | - T Ton-Nu
- Takeda California, 9625 Towne Centre Dr, San Diego, CA 92121, United States
| | - H Wang
- Takeda California, 9625 Towne Centre Dr, San Diego, CA 92121, United States
| | - C Virginio
- Aptuit (Verona) Srl, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| |
Collapse
|
44
|
Magawa CT, Eaton-Fitch N, Balinas C, Sasso EM, Thapaliya K, Barnden L, Maksoud R, Weigel B, Rudd PA, Herrero LJ, Marshall-Gradisnik S. Identification of transient receptor potential melastatin 3 proteotypic peptides employing an efficient membrane protein extraction method for natural killer cells. Front Physiol 2022; 13:947723. [PMID: 36213251 PMCID: PMC9540229 DOI: 10.3389/fphys.2022.947723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction: Mutations and misfolding of membrane proteins are associated with various disorders, hence they make suitable targets in proteomic studies. However, extraction of membrane proteins is challenging due to their low abundance, stability, and susceptibility to protease degradation. Given the limitations in existing protocols for membrane protein extraction, the aim of this investigation was to develop a protocol for a high yield of membrane proteins for isolated Natural Killer (NK) cells. This will facilitate genetic analysis of membrane proteins known as transient receptor potential melastatin 3 (TRPM3) ion channels in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) research.Methods: Two protocols, internally identified as Protocol 1 and 2, were adapted and optimized for high yield protein extraction. Protocol 1 utilized ultrasonic and salt precipitation, while Protocol 2 implemented a detergent and chloroform/methanol approach. Protein concentrations were determined by the Pierce Bicinchoninic Acid (BCA) and the Bio-Rad DC (detergent compatible) protein assays according to manufacturer’s recommendation. Using Protocol 2, protein samples were extracted from NK cells of n = 6 healthy controls (HC) and n = 4 ME/CFS patients. In silico tryptic digest and enhanced signature peptide (ESP) predictor were used to predict high-responding TRPM3 tryptic peptides. Trypsin in-gel digestion was performed on protein samples loaded on SDS-PAGE gels (excised at 150–200 kDa). A liquid chromatography-multiple reaction monitoring (LC-MRM) method was optimized and used to evaluate the detectability of TRPM3 n = 5 proteotypic peptides in extracted protein samples.Results: The detergent-based protocol protein yield was significantly higher (p < 0.05) compared with the ultrasonic-based protocol. The Pierce BCA protein assay showed more reproducibility and compatibility compared to the Bio-Rad DC protein assay. Two high-responding tryptic peptides (GANASAPDQLSLALAWNR and QAILFPNEEPSWK) for TRPM3 were detectable in n = 10 extracted protein samples from NK cells isolated from HC and ME/CFS patients.Conclusion: A method was optimized for high yield protein extraction from human NK cells and for the first time TRPM3 proteotypic peptides were detected using LC-MRM. This new method provides for future research to assess membrane protein structural and functional relationships, particularly to facilitate proteomic investigation of TRPM3 ion channel isoforms in NK cells in both health and disease states, such as ME/CFS.
Collapse
Affiliation(s)
- Chandi T Magawa
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
- Consortium Health International for Myalgic Encephalomyelitis, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
| | - Natalie Eaton-Fitch
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
- Consortium Health International for Myalgic Encephalomyelitis, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
| | - Cassandra Balinas
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
- Consortium Health International for Myalgic Encephalomyelitis, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
| | - Etianne Martini Sasso
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
- Consortium Health International for Myalgic Encephalomyelitis, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
| | - Kiran Thapaliya
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
- Consortium Health International for Myalgic Encephalomyelitis, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
| | - Leighton Barnden
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
- Consortium Health International for Myalgic Encephalomyelitis, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
| | - Rebekah Maksoud
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
- Consortium Health International for Myalgic Encephalomyelitis, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
| | - Breanna Weigel
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
- Consortium Health International for Myalgic Encephalomyelitis, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
| | - Penny A Rudd
- Institute for Glycomics, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
| | - Lara J Herrero
- Institute for Glycomics, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
| | - Sonya Marshall-Gradisnik
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
- Consortium Health International for Myalgic Encephalomyelitis, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
| |
Collapse
|
45
|
Luo B, Ding L. Ion channels and ions as therapeutic targets and strategies for herpes simplex virus infection. Future Virol 2022. [DOI: 10.2217/fvl-2022-0052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Herpes simplex virus (HSV) is a highly contagious virus that cannot be completely cured currently. Existing treatment methods are mainly nucleoside antiviral drugs, and the emergence of drug-resistant strains severely limits their use. There is an urgent need to discover antiviral drugs that act on new targets. Ion channels, a class of cellular proteins with a wide range of functions, have become critical host factors for a wide variety of viral infections. Ion channel blockers have been shown to have antiviral activity. In this study, we discuss the role of ion channels and ions in the HSV life cycle, and the potential of targeting ion channels as a novel, pharmacologically safe and wide-range antiviral treatment option.
Collapse
Affiliation(s)
- Binhua Luo
- Department of Pharmaceutics, School of Pharmacy, Hubei University of Science & Technology, Xianning, 437100, China
- Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning, 437100, China
| | - Liqiong Ding
- Department of Pharmaceutics, School of Pharmacy, Hubei University of Science & Technology, Xianning, 437100, China
- Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning, 437100, China
| |
Collapse
|
46
|
Kageyama H, Ma T, Sato M, Komiya M, Tadaki D, Hirano-Iwata A. New Aspects of Bilayer Lipid Membranes for the Analysis of Ion Channel Functions. MEMBRANES 2022; 12:membranes12090863. [PMID: 36135882 PMCID: PMC9501126 DOI: 10.3390/membranes12090863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/26/2022] [Accepted: 08/31/2022] [Indexed: 05/30/2023]
Abstract
The bilayer lipid membrane (BLM) is the main structural component of cell membranes, in which various membrane proteins are embedded. Artificially formed BLMs have been used as a platform in studies of the functions of membrane proteins, including various ion channels. In this review, we summarize recent advances that have been made on artificial BLM systems for the analysis of ion channel functions. We focus on two BLM-based systems, cell-membrane mimicry and four-terminal BLM systems. As a cell-membrane-mimicking system, an efficient screening platform for the evaluation of drug side effects that act on a cell-free synthesized channel has been developed, and its prospects for use in personalized medicine will be discussed. In the four-terminal BLMs, we introduce "lateral voltage" to BLM systems as a novel input to regulate channel activities, in addition to the traditional transmembrane voltages. Such state-of-the-art technologies and new system setups are predicted to pave the way for a variety of applications, in both fundamental physiology and in drug discovery.
Collapse
Affiliation(s)
- Hironori Kageyama
- Graduate School of Biomedical Engineering, Tohoku University, 6-6 Aoba, Aramaki, Aoba-ku, Sendai 980-8579, Japan
- Research Institute of Electrical Communication, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan
| | - Teng Ma
- Advanced Institute for Materials Research (WPI-AIMR), Tohoku University, 2-2-1 Katahira, Aoba-ku, Sendai 980-8577, Japan
| | - Madoka Sato
- Graduate School of Biomedical Engineering, Tohoku University, 6-6 Aoba, Aramaki, Aoba-ku, Sendai 980-8579, Japan
- Research Institute of Electrical Communication, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan
| | - Maki Komiya
- Research Institute of Electrical Communication, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan
| | - Daisuke Tadaki
- Research Institute of Electrical Communication, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan
| | - Ayumi Hirano-Iwata
- Graduate School of Biomedical Engineering, Tohoku University, 6-6 Aoba, Aramaki, Aoba-ku, Sendai 980-8579, Japan
- Research Institute of Electrical Communication, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan
- Advanced Institute for Materials Research (WPI-AIMR), Tohoku University, 2-2-1 Katahira, Aoba-ku, Sendai 980-8577, Japan
| |
Collapse
|
47
|
Katsuta E, Takabe K, Vujcic M, Gottlieb PA, Dai T, Mercado-Perez A, Beyder A, Wang Q, Opyrchal M. Mechano-Sensing Channel PIEZO2 Enhances Invasive Phenotype in Triple-Negative Breast Cancer. Int J Mol Sci 2022; 23:9909. [PMID: 36077309 PMCID: PMC9455988 DOI: 10.3390/ijms23179909] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/25/2022] [Accepted: 08/27/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Mechanically gated PIEZO channels lead to an influx of cations, activation of additional Ca2+ channels, and cell depolarization. This study aimed to investigate PIEZO2's role in breast cancer. METHODS The clinical relevance of PIEZO2 expression in breast cancer patient was analyzed in a publicly available dataset. Utilizing PIEZO2 overexpressed breast cancer cells, and in vitro and in vivo experiments were conducted. RESULTS High expression of PIEZO2 was correlated with a worse survival in triple-negative breast cancer (TNBC) but not in other subtypes. Increased PEIZO2 channel function was confirmed in PIEZO2 overexpressed cells after mechanical stimulation. PIEZO2 overexpressed cells showed increased motility and invasive phenotypes as well as higher expression of SNAIL and Vimentin and lower expression of E-cadherin in TNBC cells. Correspondingly, high expression of PIEZO2 was correlated with the increased expression of epithelial-mesenchymal transition (EMT)-related genes in a TNBC patient. Activated Akt signaling was observed in PIEZO2 overexpressed TNBC cells. PIEZO2 overexpressed MDA-MB-231 cells formed a significantly higher number of lung metastases after orthotopic implantation. CONCLUSION PIEZO2 activation led to enhanced SNAIL stabilization through Akt activation. It enhanced Vimentin and repressed E-cadherin transcription, resulting in increased metastatic potential and poor clinical outcomes in TNBC patients.
Collapse
Affiliation(s)
- Eriko Katsuta
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, NY 14203, USA
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo 160-8402, Japan
- Department of Surgery, Yokohama City University, Yokohama 236-0004, Japan
- Department of Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
- Department of Breast Surgery, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Marija Vujcic
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Philip A. Gottlieb
- Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Tao Dai
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Arnaldo Mercado-Perez
- Enteric Neuroscience Program, Division of Gastroenterology & Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Arthur Beyder
- Enteric Neuroscience Program, Division of Gastroenterology & Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Qingfei Wang
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Mateusz Opyrchal
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
48
|
Tsukamoto S, Hlokoane O, Miyako K, Irie R, Sakai R, Oikawa M. Oxa-Michael-based divergent synthesis of artificial glutamate analogs. RSC Adv 2022; 12:22175-22179. [PMID: 36043066 PMCID: PMC9364357 DOI: 10.1039/d2ra03744k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/02/2022] [Indexed: 11/24/2022] Open
Abstract
Herein we report stereoselective generation of two skeletons, 1,3-dioxane and tetrahydropyranol, by oxa-Michael reaction as the key reaction from δ-hydroxyenone. The construction of the 1,3-dioxane skeleton, achieved through hemiacetal formation followed by oxa-Michael reaction from δ-hydroxyenone, was exploited to access structurally diverse heterotricyclic artificial glutamate analogs. On the other hand, formation of a novel tetrahydro-2H-pyranol skeleton was accomplished by the inverse reaction order: oxa-Michael reaction followed by hemiacetal formation. Thus, this study succeeded in showing that structural diversity in a compound collection can be acquired by interchanging the order of just two reactions. Among the skeletally diverse, heterotricyclic artificial glutamate analogs synthesized in this study, a neuronally active compound named TKM-50 was discovered in the mice in vivo assay. By interchanging the order of reactions, two types of skeletons were created and a neuroactive artificial glutamate analog was developed.![]()
Collapse
Affiliation(s)
- Shuntaro Tsukamoto
- Graduate School of Nanobioscience, Yokohama City University Seto 22-2, Kanazawa-ku Yokohama 236-0027 Japan
| | - Oriel Hlokoane
- Graduate School of Nanobioscience, Yokohama City University Seto 22-2, Kanazawa-ku Yokohama 236-0027 Japan .,Department of Pharmacy, National University of Lesotho P.O. Roma 180 Maseru Lesotho
| | - Kei Miyako
- Faculty of Fisheries Sciences, Hokkaido University Hakodate 041-8611 Japan
| | - Raku Irie
- Graduate School of Nanobioscience, Yokohama City University Seto 22-2, Kanazawa-ku Yokohama 236-0027 Japan
| | - Ryuichi Sakai
- Faculty of Fisheries Sciences, Hokkaido University Hakodate 041-8611 Japan
| | - Masato Oikawa
- Graduate School of Nanobioscience, Yokohama City University Seto 22-2, Kanazawa-ku Yokohama 236-0027 Japan
| |
Collapse
|
49
|
Identification of a Prognostic Microenvironment-Related Gene Signature in Glioblastoma Patients Treated with Carmustine Wafers. Cancers (Basel) 2022; 14:cancers14143413. [PMID: 35884475 PMCID: PMC9320240 DOI: 10.3390/cancers14143413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022] Open
Abstract
Despite the state-of-the-art treatment, patients diagnosed with glioblastoma (GBM) have a median overall survival (OS) of 14 months. The insertion of carmustine wafers (CWs) into the resection cavity as adjuvant treatment represents a promising option, although its use has been limited due to contrasting clinical results. Our retrospective evaluation of CW efficacy showed a significant improvement in terms of OS in a subgroup of patients. Given the crucial role of the tumor microenvironment (TME) in GBM progression and response to therapy, we hypothesized that the TME of patients who benefited from CW could have different properties compared to that of patients who did not show any advantage. Using an in vitro model of the glioma microenvironment, represented by glioma-associated-stem cells (GASC), we performed a transcriptomic analysis of GASC isolated from tumors of patients responsive and not responsive to CW to identify differentially expressed genes. We found different transcriptomic profiles, and we identified four genes, specifically down-regulated in GASC isolated from long-term survivors, correlated with clinical data deposited in the TCGA–GBM dataset. Our results highlight that studying the in vitro properties of patient-specific glioma microenvironments can help to identify molecular determinants potentially prognostic for patients treated with CW.
Collapse
|
50
|
Zhu Z, Deng Z, Wang Q, Wang Y, Zhang D, Xu R, Guo L, Wen H. Simulation and Machine Learning Methods for Ion-Channel Structure Determination, Mechanistic Studies and Drug Design. Front Pharmacol 2022; 13:939555. [PMID: 35837274 PMCID: PMC9275593 DOI: 10.3389/fphar.2022.939555] [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: 05/09/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Ion channels are expressed in almost all living cells, controlling the in-and-out communications, making them ideal drug targets, especially for central nervous system diseases. However, owing to their dynamic nature and the presence of a membrane environment, ion channels remain difficult targets for the past decades. Recent advancement in cryo-electron microscopy and computational methods has shed light on this issue. An explosion in high-resolution ion channel structures paved way for structure-based rational drug design and the state-of-the-art simulation and machine learning techniques dramatically improved the efficiency and effectiveness of computer-aided drug design. Here we present an overview of how simulation and machine learning-based methods fundamentally changed the ion channel-related drug design at different levels, as well as the emerging trends in the field.
Collapse
Affiliation(s)
- Zhengdan Zhu
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Beijing Institute of Big Data Research, Beijing, China
| | - Zhenfeng Deng
- DP Technology, Beijing, China
- School of Pharmaceutical Sciences, Peking University, Beijing, China
| | | | | | - Duo Zhang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- DP Technology, Beijing, China
| | - Ruihan Xu
- DP Technology, Beijing, China
- National Engineering Research Center of Visual Technology, Peking University, Beijing, China
| | | | - Han Wen
- DP Technology, Beijing, China
| |
Collapse
|