1
|
Wu H, Sun L, Huo T, Wensel TG, Horrigan FT, Wang Z. The identification of XPR1 as a voltage- and phosphate-activated phosphate-permeable ion channel. Nat Commun 2025; 16:4519. [PMID: 40374661 PMCID: PMC12081713 DOI: 10.1038/s41467-025-59678-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 04/29/2025] [Indexed: 05/17/2025] Open
Abstract
Maintaining a balance of inorganic phosphate (Pi) is vital for cellular functionality. Proper phosphate levels are managed through Pi import and export; and the processes governing Pi export remain the least understood. Xenotropic and Polytropic retrovirus Receptor 1 (XPR1) has been identified as the only known Pi export protein in mammals. In this study, we introduce the cryogenic electron microscopy structure of human XPR1 (hXPR1), unveiling a structural arrangement distinct from that of any known ion transporter. Our structural results suggest that hXPR1 may operate as an ion channel, a hypothesis supported by patch clamp recordings revealing hXPR1's voltage- and Pi-dependent activity and large unitary conductance. Further analyses, including the structure of hXPR1 in presence of Pi, and mutagenesis studies at one of the putative Pi binding sites, lead us to propose a plausible ion permeation pathway. Together, our results provide novel perspectives on the Pi transport mechanism of XPR1.
Collapse
Affiliation(s)
- Hongjiang Wu
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Liang Sun
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Tong Huo
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Theodore G Wensel
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Frank T Horrigan
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA.
| | - Zhao Wang
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- CryoEM Core (Advanced Technology Core), Baylor College of Medicine, Houston, TX, USA.
- Department of Materials Science and NanoEngineering, Rice University, Houston, TX, USA.
- Department of Molecular and Cellular Oncology, Division of Basic Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
2
|
Kariapper L, Marathe IA, Niesman AB, Suino-Powell K, Chook YM, Wysocki VH, Worden EJ. Setdb1 and Atf7IP form a hetero-trimeric complex that blocks Setdb1 nuclear export. J Biol Chem 2025:110171. [PMID: 40339988 DOI: 10.1016/j.jbc.2025.110171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/16/2025] [Accepted: 04/29/2025] [Indexed: 05/10/2025] Open
Abstract
Histone H3K9 methylation (H3K9me) by Setdb1 silences retrotransposons (rTE) by sequestering them in heterochromatin. Atf7IP is a constitutive binding partner of Setdb1 and is responsible for Setdb1 nuclear localization, activation and chromatin recruitment. However, structural details of the Setdb1/Atf7IP interaction have not been elucidated. We used Alphafold2 predictions and biochemical reconstitutions to show that one copy of Setdb1 and two copies of Atf7IP form a hetero-trimeric complex in vitro and in cells. We also find that Atf7IP self-associates, forming multimeric complexes that are resolved upon Setdb1 binding. Setdb1 binds to Atf7IP through coiled coil interactions that include both Setdb1 nuclear export signals (NES). Atf7IP directly competes with CRM1 to bind the Setdb1 NES motifs, explaining how Atf7IP prevents CRM1-mediated nuclear export of Setdb1. Setdb1 also forms hetero-trimeric complexes with the Atf7IP paralog Atf7IP2 and we show that Setdb1 can form mixed heterotrimers comprising one copy of each Setdb1, Atf7IP and Atf7IP2. Atf7IP and Atf7IP2 are co-expressed in many tissues suggesting that heterotrimers with different compositions of Atf7IP and Atf7IP2 may differentially regulate H3K9me by fine-tuning Setdb1 localization and activity.
Collapse
Affiliation(s)
- Leena Kariapper
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI, USA
| | - Ila A Marathe
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
| | - Ashley Brower Niesman
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kelly Suino-Powell
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI, USA
| | - Yuh Min Chook
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Vicki H Wysocki
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
| | - Evan J Worden
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI, USA
| |
Collapse
|
3
|
Zhang H, Yang X, Xue Y, Huang Y, Mo Y, Huang Y, Zhang H, Zhang X, Zhao W, Jia B, Li N, Gao N, Yang Y, Xiang D, Wang S, Qin Gao Y, Liao J. A basigin antibody modulates MCTs to impact tumor metabolism and immunity. Cell Discov 2025; 11:44. [PMID: 40324980 PMCID: PMC12053622 DOI: 10.1038/s41421-025-00777-1] [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: 09/24/2024] [Accepted: 01/20/2025] [Indexed: 05/07/2025] Open
Abstract
Lactate metabolism and signaling intricately intertwine in the context of cancer and immunity. Basigin, working alongside monocarboxylate transporters MCT1 and MCT4, orchestrates the movement of lactate across cell membranes. Despite their potential in treating formidable tumors, the mechanisms by which basigin antibodies affect basigin and MCTs remain unclear. Our research demonstrated that basigin positively modulates MCT activity. We subsequently developed a basigin antibody that converts basigin into a negative modulator, thereby suppressing lactate transport and enhancing anti-tumor immunity. Additionally, the antibody alters metabolic profiles in NSCLC-PDOs and T cells. Cryo-EM structural analysis and molecular dynamics simulations reveal that the extracellular Ig2 domain and transmembrane domain of basigin regulate MCT1 activity through an allosteric mechanism. The antibody decreases MCT1 transition rate by reducing the flexibility of basigin's Ig2 domain and diminishing interactions between basigin's transmembrane domain and MCT1. These findings underscore the promise of basigin antibodies in combating tumors by modulating metabolism and immunity, and the value of a common therapeutic subunit shared by multiple transporter targets.
Collapse
Affiliation(s)
- Heng Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xuemei Yang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yue Xue
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Yi Huang
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Yingxi Mo
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Yurun Huang
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Hong Zhang
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Xiaofei Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weixin Zhao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bin Jia
- Lung Cancer Department, Tianjin Cancer Hospital, Tianjin, China
| | - Ningning Li
- State Key Laboratory of Membrane Biology, Peking-Tsinghua Joint Center for Life Sciences, School of Life Sciences, Peking University, Beijing, China
| | - Ning Gao
- State Key Laboratory of Membrane Biology, Peking-Tsinghua Joint Center for Life Sciences, School of Life Sciences, Peking University, Beijing, China
| | - Yue Yang
- Institute of Toxicology, School of Public Health, Lanzhou University, Lanzhou, Gansu, China
| | - Dongxi Xiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, and Department of Biliary-Pancreatic Surgery, the Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Shan Wang
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China.
| | - Yi Qin Gao
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China.
| | - Jun Liao
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- Alphelix Biosciences, Foshan, Guangdong, China.
| |
Collapse
|
4
|
Juen Z, Lu Z, Yu R, Chang AN, Wang B, Fitzpatrick AWP, Zuker CS. The structure of human sweetness. Cell 2025:S0092-8674(25)00456-8. [PMID: 40339580 DOI: 10.1016/j.cell.2025.04.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/03/2025] [Accepted: 04/15/2025] [Indexed: 05/10/2025]
Abstract
In humans, the detection and ultimately the perception of sweetness begin in the oral cavity, where taste receptor cells (TRCs) dedicated to sweet-sensing interact with sugars, artificial sweeteners, and other sweet-tasting chemicals. Human sweet TRCs express on their cell surface a sweet receptor that initiates the cascade of signaling events responsible for our strong attraction to sweet stimuli. Here, we describe the cryo-electron microscopy (cryo-EM) structure of the human sweet receptor bound to two of the most widely used artificial sweeteners-sucralose and aspartame. Our results reveal the structural basis for sweet detection, provide insights into how a single receptor mediates all our responses to such a wide range of sweet-tasting compounds, and open up unique possibilities for designing a generation of taste modulators informed by the structure of the human receptor.
Collapse
Affiliation(s)
- Zhang Juen
- Zuckerman Mind Brain Behavior Institute and Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Howard Hughes Medical Institute, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Zhengyuan Lu
- Zuckerman Mind Brain Behavior Institute and Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Howard Hughes Medical Institute, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Ruihuan Yu
- Zuckerman Mind Brain Behavior Institute and Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Howard Hughes Medical Institute, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Andrew N Chang
- Zuckerman Mind Brain Behavior Institute and Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Brian Wang
- Zuckerman Mind Brain Behavior Institute and Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Howard Hughes Medical Institute, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Anthony W P Fitzpatrick
- Zuckerman Mind Brain Behavior Institute and Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Charles S Zuker
- Zuckerman Mind Brain Behavior Institute and Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Howard Hughes Medical Institute, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Department of Neuroscience, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
5
|
He Z, Zhang J, Xu Y, Fine EJ, Suomivuori CM, Dror RO, Feng L. Structure of mitochondrial pyruvate carrier and its inhibition mechanism. Nature 2025; 641:250-257. [PMID: 40044865 PMCID: PMC12043432 DOI: 10.1038/s41586-025-08667-y] [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: 05/18/2024] [Accepted: 01/17/2025] [Indexed: 04/13/2025]
Abstract
The mitochondrial pyruvate carrier (MPC) governs the entry of pyruvate-a central metabolite that bridges cytosolic glycolysis with mitochondrial oxidative phosphorylation-into the mitochondrial matrix1-5. It thus serves as a pivotal metabolic gatekeeper and has fundamental roles in cellular metabolism. Moreover, MPC is a key target for drugs aimed at managing diabetes, non-alcoholic steatohepatitis and neurodegenerative diseases4-6. However, despite MPC's critical roles in both physiology and medicine, the molecular mechanisms underlying its transport function and how it is inhibited by drugs have remained largely unclear. Here our structural findings on human MPC define the architecture of this vital transporter, delineate its substrate-binding site and translocation pathway, and reveal its major conformational states. Furthermore, we explain the binding and inhibition mechanisms of MPC inhibitors. Our findings provide the molecular basis for understanding MPC's function and pave the way for the development of more-effective therapeutic reagents that target MPC.
Collapse
Affiliation(s)
- Zheng He
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jianxiu Zhang
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yan Xu
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Eve J Fine
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Carl-Mikael Suomivuori
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Ron O Dror
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Liang Feng
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
6
|
Kumar Mondal A, Carrillo E, Jayaraman V, Twomey EC. Glutamate gating of AMPA-subtype iGluRs at physiological temperatures. Nature 2025; 641:788-796. [PMID: 40140570 PMCID: PMC12074995 DOI: 10.1038/s41586-025-08770-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/11/2025] [Indexed: 03/28/2025]
Abstract
Ionotropic glutamate receptors (iGluRs) are tetrameric ligand-gated ion channels that mediate most excitatory neurotransmission1. iGluRs are gated by glutamate, where on glutamate binding, they open their ion channels to enable cation influx into postsynaptic neurons, initiating signal transduction1,2. The structural mechanics of how glutamate gating occurs in full-length iGluRs is not well understood. Here, using the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid subtype iGluR (AMPAR), we identify the glutamate-gating mechanism. AMPAR activation by glutamate is augmented at physiological temperatures. By preparing AMPARs for cryogenic-electron microscopy at these temperatures, we captured the glutamate-gating mechanism. Activation by glutamate initiates ion channel opening that involves all ion channel helices hinging away from the pore axis in a motif that is conserved across all iGluRs. Desensitization occurs when the local dimer pairs decouple and enables closure of the ion channel below through restoring the channel hinges and refolding the channel gate. Our findings define how glutamate gates iGluRs, provide foundations for therapeutic design and demonstrate how physiological temperatures can alter iGluR function.
Collapse
Affiliation(s)
- Anish Kumar Mondal
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elisa Carrillo
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Vasanthi Jayaraman
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, USA.
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Edward C Twomey
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- The Beckman Center for Cryo-EM at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA.
| |
Collapse
|
7
|
Ma Q, Ma K, Dong Y, Meng Y, Zhao J, Li R, Bai Q, Wu D, Jiang D, Sun J, Zhao Y. Binding mechanism and antagonism of the vesicular acetylcholine transporter VAChT. Nat Struct Mol Biol 2025; 32:818-827. [PMID: 39806024 DOI: 10.1038/s41594-024-01462-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025]
Abstract
The vesicular acetylcholine transporter (VAChT) has a pivotal role in packaging and transporting acetylcholine for exocytotic release, serving as a vital component of cholinergic neurotransmission. Dysregulation of its function can result in neurological disorders. It also serves as a target for developing radiotracers to quantify cholinergic neuron deficits in neurodegenerative conditions. Here we unveil the cryo-electron microscopy structures of human VAChT in its apo state, the substrate acetylcholine-bound state and the inhibitor vesamicol-bound state. These structures assume a lumen-facing conformation, offering a clear depiction of architecture of VAChT. The acetylcholine-bound structure provides a detailed understanding of how VAChT recognizes its substrate, shedding light on the coupling mechanism of protonation and substrate binding. Meanwhile, the vesamicol-bound structure reveals the binding mode of vesamicol to VAChT, laying the structural foundation for the design of the next generation of radioligands targeting VAChT.
Collapse
Affiliation(s)
- Qiao Ma
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kunpeng Ma
- Interdisciplinary Center for Brain Information, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yanli Dong
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yufei Meng
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jun Zhao
- Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences at Weifang, Weifang, China
| | - Renjie Li
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qinru Bai
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Di Wu
- University of Chinese Academy of Sciences, Beijing, China
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
| | - Daohua Jiang
- University of Chinese Academy of Sciences, Beijing, China
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
| | - Jianyuan Sun
- University of Chinese Academy of Sciences, Beijing, China
- Interdisciplinary Center for Brain Information, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yan Zhao
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders and National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
8
|
Lin H, Huang J, Li T, Li W, Wu Y, Yang T, Nian Y, Lin X, Wang J, Wang R, Zhao X, Su N, Zhang J, Wu X, Fan M. Structure and mechanism of the plastid/parasite ATP/ADP translocator. Nature 2025; 641:797-804. [PMID: 40074904 DOI: 10.1038/s41586-025-08743-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025]
Abstract
Adenosine triphosphate (ATP) is the principal energy currency of all living cells1,2. Metabolically impaired obligate intracellular parasites, such as the human pathogens Chlamydia trachomatis and Rickettsia prowazekii, can acquire ATP from their host cells through a unique ATP/adenosine diphosphate (ADP) translocator, which mediates the import of ATP into and the export of ADP and phosphate out of the parasite cells, thus allowing the exploitation of the energy reserves of host cells (also known as energy parasitism). This type of ATP/ADP translocator also exists in the obligate intracellular endosymbionts of protists and the plastids of plants and algae and has been implicated to play an important role in endosymbiosis3-31. The plastid/parasite type of ATP/ADP translocator is phylogenetically and functionally distinct from the mitochondrial ATP/ADP translocator, and its structure and transport mechanism are still unknown. Here we report the cryo-electron microscopy structures of two plastid/parasite types of ATP/ADP translocators in the apo and substrate-bound states. The ATP/ADP-binding pocket is located at the interface between the N and C domains of the translocator, and a conserved asparagine residue within the pocket is critical for substrate specificity. The translocator operates through a rocker-switch alternating access mechanism involving the relative rotation of the two domains as rigid bodies. Our results provide critical insights for understanding ATP translocation across membranes in energy parasitism and endosymbiosis and offer a structural basis for developing drugs against obligate intracellular parasites.
Collapse
Affiliation(s)
- Huajian Lin
- CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Plant Carbon Capture, Chinese Academy of Sciences, Shanghai, China
| | - Jian Huang
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Tianming Li
- CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Plant Carbon Capture, Chinese Academy of Sciences, Shanghai, China
| | - Wenjuan Li
- CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Plant Carbon Capture, Chinese Academy of Sciences, Shanghai, China
| | - Yutong Wu
- CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Plant Carbon Capture, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Tianjiao Yang
- CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Plant Carbon Capture, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuwei Nian
- CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Plant Carbon Capture, Chinese Academy of Sciences, Shanghai, China
| | - Xiang Lin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Department of Biochemistry and Biophysics, School of Life Sciences, Fudan University, Shanghai, China
| | - Jiangqin Wang
- The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Ruiying Wang
- CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Plant Carbon Capture, Chinese Academy of Sciences, Shanghai, China
| | - Xiaohui Zhao
- CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Nannan Su
- The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China.
| | - Jinru Zhang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Department of Biochemistry and Biophysics, School of Life Sciences, Fudan University, Shanghai, China.
| | - Xudong Wu
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China.
| | - Minrui Fan
- CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China.
- Key Laboratory of Plant Carbon Capture, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
9
|
Yin Y, Park CG, Feng S, Guan Z, Lee HJ, Zhang F, Sharma K, Borgnia MJ, Im W, Lee SY. Molecular basis of neurosteroid and anticonvulsant regulation of TRPM3. Nat Struct Mol Biol 2025; 32:828-840. [PMID: 39809942 DOI: 10.1038/s41594-024-01463-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025]
Abstract
Transient receptor potential channel subfamily M member 3 (TRPM3) is a Ca2+-permeable cation channel activated by the neurosteroid pregnenolone sulfate (PregS) or heat, serving as a nociceptor in the peripheral sensory system. Recent discoveries of autosomal dominant neurodevelopmental disorders caused by gain-of-function mutations in TRPM3 highlight its role in the central nervous system. Notably, the TRPM3 inhibitor primidone, an anticonvulsant, has proven effective in treating patients with TRPM3-linked neurological disorders and in mouse models of thermal nociception. However, our understanding of neurosteroids, inhibitors and disease mutations on TRPM3 is limited. Here we present cryogenic electron microscopy structures of the mouse TRPM3 in complex with cholesteryl hemisuccinate, primidone and PregS with the synthetic agonist CIM 0216. Our studies identify the binding sites for the neurosteroid, synthetic agonist and inhibitor and offer insights into their effects and disease mutations on TRPM3 gating, aiding future drug development.
Collapse
Affiliation(s)
- Ying Yin
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Cheon-Gyu Park
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Shasha Feng
- Department of Biological Sciences, Lehigh University, Bethlehem, PA, USA
| | - Ziqiang Guan
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Hyuk-Joon Lee
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Feng Zhang
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Kedar Sharma
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Mario J Borgnia
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Wonpil Im
- Department of Biological Sciences, Lehigh University, Bethlehem, PA, USA
| | - Seok-Yong Lee
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
10
|
Wang Y, Hong S, Hudson H, Kory N, Kinch LN, Kozlitina J, Cohen JC, Hobbs HH. PNPLA3(148M) is a gain-of-function mutation that promotes hepatic steatosis by inhibiting ATGL-mediated triglyceride hydrolysis. J Hepatol 2025; 82:871-881. [PMID: 39550037 DOI: 10.1016/j.jhep.2024.10.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUND & AIMS PNPLA3(148M) (patatin-like phospholipase domain-containing protein 3) is the most impactful genetic risk factor for steatotic liver disease. A key unresolved issue is whether PNPLA3(148M) confers a loss- or gain-of-function. Here we test the hypothesis that PNPLA3 causes steatosis by sequestering ABHD5 (α/β hydrolase domain-containing protein 5), the cofactor of ATGL (adipose TG lipase), thus limiting mobilization of hepatic triglyceride (TG). METHODS We quantified and compared the physical interactions between ABHD5 and PNPLA3/ATGL in cultured hepatocytes using NanoBiT complementation assays and immunocytochemistry. Recombinant proteins purified from human cells were used to compare TG hydrolytic activities of PNPLA3 and ATGL in the presence or absence of ABHD5. Adenoviruses and adeno-associated viruses were used to express PNPLA3 in liver-specific Atgl-/- mice and to express ABHD5 in livers of Pnpla3M/M mice, respectively. RESULTS ABHD5 interacted preferentially with PNPLA3 relative to ATGL in cultured hepatocytes. No differences were seen in the strength of the interactions between ABHD5 with PNPLA3(WT) and PNPLA3(148M). In contrast to prior findings, we found that PNPLA3, like ATGL, is activated by ABHD5 in in vitro assays using purified proteins. PNPLA3(148M)-associated inhibition of TG hydrolysis required that ATGL be expressed and that PNPLA3 be located on lipid droplets. Finally, overexpression of ABHD5 reversed the hepatic steatosis in Pnpla3M/M mice. CONCLUSIONS These findings support the premise that PNPLA3(148M) is a gain-of-function mutation that promotes hepatic steatosis by accumulating on lipid droplets and inhibiting ATGL-mediated lipolysis in an ABHD5-dependent manner. Our results predict that reducing, rather than increasing, PNPLA3 expression will be the best strategy to treat PNPLA3(148M)-associated steatotic liver disease. IMPACT AND IMPLICATIONS Steatotic liver disease (SLD) is a common complex disorder associated with both environmental and genetic risk factors. PNPLA3(148M) is the most impactful genetic risk factor for SLD and yet its pathogenic mechanism remains controversial. Herein, we provide evidence that PNPLA3(148M) promotes triglyceride (TG) accumulation by sequestering ABHD5, thus limiting its availability to activate ATGL. Although the substitution of methionine for isoleucine reduces the TG hydrolase activity of PNPLA3, the loss of enzymatic function is not directly related to the steatotic effect of the variant. It is the resulting accumulation of PNPLA3 on LDs that confers a gain-of-function by interfering with ATGL-mediated TG hydrolysis. These findings have implications for the design of potential PNPLA3(148M)-based therapies. Reducing, rather than increasing, PNPLA3 levels is predicted to reverse steatosis in susceptible individuals.
Collapse
Affiliation(s)
- Yang Wang
- Department of Molecular Genetics, University of Texas Southwestern Medical Center (UTSW), Dallas, TX 75390-9046, USA.
| | - Sen Hong
- Department of Molecular Genetics, University of Texas Southwestern Medical Center (UTSW), Dallas, TX 75390-9046, USA; Howard Hughes Medical Institute, UTSW, Dallas, TX 75390, USA
| | - Hannah Hudson
- Department of Molecular Genetics, University of Texas Southwestern Medical Center (UTSW), Dallas, TX 75390-9046, USA
| | - Nora Kory
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Lisa N Kinch
- Department of Molecular Genetics, University of Texas Southwestern Medical Center (UTSW), Dallas, TX 75390-9046, USA; Howard Hughes Medical Institute, UTSW, Dallas, TX 75390, USA
| | - Julia Kozlitina
- The Eugene McDermott Center for Human Growth and Development, UTSW, Dallas, TX, 75390, USA
| | - Jonathan C Cohen
- Department of Molecular Genetics, University of Texas Southwestern Medical Center (UTSW), Dallas, TX 75390-9046, USA; Center for Human Nutrition, UTSW, Dallas, TX 75390, USA
| | - Helen H Hobbs
- Department of Molecular Genetics, University of Texas Southwestern Medical Center (UTSW), Dallas, TX 75390-9046, USA; Howard Hughes Medical Institute, UTSW, Dallas, TX 75390, USA; The Eugene McDermott Center for Human Growth and Development, UTSW, Dallas, TX, 75390, USA.
| |
Collapse
|
11
|
Yadav R, Han GW, Gati C. Molecular basis of human GABA transporter 3 inhibition. Nat Commun 2025; 16:3830. [PMID: 40268946 PMCID: PMC12019481 DOI: 10.1038/s41467-025-59066-w] [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: 08/08/2024] [Accepted: 04/10/2025] [Indexed: 04/25/2025] Open
Abstract
γ-Aminobutyric acid (GABA) transporters (GATs) are sodium- and chloride-dependent transporters that mediate the reuptake of the inhibitory neurotransmitter GABA after its release from synaptic vesicles. GAT3 transports GABA from the synaptic cleft into astrocytes and modulates synaptic signaling. GAT3 has been implicated in various neurological disorders and neurodegenerative diseases, rendering it a therapeutically important drug target. To understand the mechanism of transport and inhibition, here we determine cryo-electron microscopy structures of human GAT3 in its apo form and in complex with the selective inhibitor SNAP-5114. Unexpectedly, we have discovered that SNAP-5114 acts as a noncompetitive inhibitor at GAT3. SNAP-5114 binds at the orthosteric substrate binding pocket of GAT3 in its inward-open conformation, in agreement with its noncompetitive inhibition of GABA transport. In the apo state, GAT3 also adopts an inward-open conformation with the orthosteric substrate binding pocket exposed to cytoplasm, while an extensive network of interactions closes the extracellular gate. The structures, complemented with mutagenesis and radioligand uptake assays, show that the increased orthosteric substrate binding pocket volume and bulky moieties of SNAP-5114, drive the selective inhibition of GAT3 over GAT1. Our structural and functional studies reveal the mechanism of selective inhibition of GAT3 and provide a framework for GAT3-targeted rational drug design.
Collapse
Affiliation(s)
- Ravi Yadav
- The Bridge Institute, Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA
- Molecular and Computational Biology, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Gye Won Han
- The Bridge Institute, Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA
| | - Cornelius Gati
- The Bridge Institute, Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA.
- Molecular and Computational Biology, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA.
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
12
|
Matsushita D, Toyoda Y, Lee Y, Aoi M, Matsuo H, Takada T, Nishizawa T. Structural basis of urate transport by glucose transporter 9. Cell Rep 2025; 44:115514. [PMID: 40186864 DOI: 10.1016/j.celrep.2025.115514] [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: 09/04/2024] [Revised: 01/17/2025] [Accepted: 03/13/2025] [Indexed: 04/07/2025] Open
Abstract
Glucose transporter 9 (GLUT9) is a critical urate transporter involved in renal reabsorption, playing a pivotal role in regulating physiological urate levels and representing a potential therapeutic target for gout. Despite such clinical significance, the structural basis of urate recognition and transport by GLUT9 remains elusive. Here, we present the cryoelectron microscopy (cryo-EM) structures of GLUT9 in the inward-open conformation in both apo and urate-bound states. Urate binds in a cleft between the N-terminal and C-terminal domains, interacting via hydrogen bonds and hydrophobic interactions. Structural comparison with sugar-transporting GLUTs highlights unique amino acid compositions in the substrate recognition pocket of GLUT9. Functional and mutational studies directly measuring GLUT9-mediated urate uptake further demonstrate the cooperative roles of multiple residues in urate recognition. Our findings elucidate the structural basis of urate transport by GLUT9 and provide valuable insights for the development of uricosuric drugs targeting GLUT9.
Collapse
Affiliation(s)
- Daiki Matsushita
- Graduate School of Medical Life Science, Yokohama City University, Yokohama 230-0045, Japan
| | - Yu Toyoda
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo 113-8655, Japan; Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa 359-8513, Japan
| | - Yongchan Lee
- Graduate School of Medical Life Science, Yokohama City University, Yokohama 230-0045, Japan
| | - Maeda Aoi
- Graduate School of Medical Life Science, Yokohama City University, Yokohama 230-0045, Japan
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa 359-8513, Japan
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Tomohiro Nishizawa
- Graduate School of Medical Life Science, Yokohama City University, Yokohama 230-0045, Japan.
| |
Collapse
|
13
|
Nam YW, Im D, Garcia ASC, Tringides ML, Nguyen HM, Liu Y, Orfali R, Ramanishka A, Pintilie G, Su CC, Cui M, Logothetis DE, Yu EW, Wulff H, Chandy KG, Zhang M. Cryo-EM structures of the small-conductance Ca 2+-activated K Ca2.2 channel. Nat Commun 2025; 16:3690. [PMID: 40246884 PMCID: PMC12006403 DOI: 10.1038/s41467-025-59061-1] [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: 07/22/2024] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Abstract
Small-conductance Ca2+-activated K+ (KCa2.1-KCa2.3) channels modulate neuronal and cardiac excitability. We report cryo-electron microscopy structures of the KCa2.2 channel in complex with calmodulin and Ca2+, alone or bound to two small molecule inhibitors, at 3.18, 3.50, 2.99 and 2.97 angstrom resolution, respectively. Extracellular S3-S4 loops in β-hairpin configuration form an outer canopy over the pore with an aromatic box at the canopy's center. Each S3-S4 β-hairpin is tethered to the selectivity filter in the neighboring subunit by inter-subunit hydrogen bonds. This hydrogen bond network flips the aromatic residue (Tyr362) in the filter's GYG signature by 180°, causing the outer selectivity filter to widen and water to enter the filter. Disruption of the tether by a mutation narrows the outer selectivity filter, realigns Tyr362 to the position seen in other K+ channels, and significantly increases unitary conductance. UCL1684, a mimetic of the bee venom peptide apamin, sits atop the canopy and occludes the opening in the aromatic box. AP14145, an analogue of a therapeutic for atrial fibrillation, binds in the central cavity below the selectivity filter and induces closure of the inner gate. These structures provide a basis for understanding the small unitary conductance and pharmacology of KCa2.x channels.
Collapse
Affiliation(s)
- Young-Woo Nam
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Dohyun Im
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ana Santa Cruz Garcia
- Department of Pharmaceutical Sciences, Northeastern University School of Pharmacy and Pharmaceutical Sciences, Boston, MA, USA
| | - Marios L Tringides
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Hai Minh Nguyen
- Department of Pharmacology, School of Medicine, University of California Davis, Davis, CA, USA
| | - Yan Liu
- Division of CryoEM and Bioimaging, Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, CA, USA
| | - Razan Orfali
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Alena Ramanishka
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Grigore Pintilie
- Department of Bioengineering and Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Chih-Chia Su
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Meng Cui
- Department of Pharmaceutical Sciences, Northeastern University School of Pharmacy and Pharmaceutical Sciences, Boston, MA, USA
| | - Diomedes E Logothetis
- Department of Pharmaceutical Sciences, Northeastern University School of Pharmacy and Pharmaceutical Sciences, Boston, MA, USA
| | - Edward W Yu
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California Davis, Davis, CA, USA
| | - K George Chandy
- LKCMedicine-ICE Collaborative Platform, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Miao Zhang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA.
| |
Collapse
|
14
|
Riel EB, Bu W, Joseph TT, Khajoueinejad L, Eckenhoff RG, Riegelhaupt PM. The cryo-EM structure and physical basis for anesthetic inhibition of the THIK1 K2P channel. Proc Natl Acad Sci U S A 2025; 122:e2421654122. [PMID: 40178898 PMCID: PMC12002230 DOI: 10.1073/pnas.2421654122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 03/02/2025] [Indexed: 04/05/2025] Open
Abstract
THIK1 tandem pore domain (K2P) potassium channels regulate microglial surveillance of the central nervous system and responsiveness to inflammatory insults. With microglia recognized as critical to the pathogenesis of neurodegenerative diseases, THIK1 channels are putative therapeutic targets to control microglia dysfunction. While THIK channels can principally be distinguished from other K2Ps by their distinctive inhibitory response to volatile anesthetics (VAs), molecular details governing THIK channel gating remain largely unexplored. Here, we report a 3.2 Å cryo-electron microscopy structure of the THIK1 channel in a closed conformation. A central pore gate located directly below the THIK1 selectivity filter is formed by inward-facing TM4 helix tyrosine residues that occlude the ion conduction pathway. VA inhibition of THIK requires closure of this central pore gate. Using a combination of anesthetic photolabeling, electrophysiology, and molecular dynamics simulation, we identify a functionally critical THIK1 VA binding site positioned between the central gate and a structured section of the THIK1 TM2/TM3 loop. Our results demonstrate the molecular architecture of the THIK1 channel and elucidate critical structural features involved in regulation of THIK1 channel gating and anesthetic inhibition.
Collapse
Affiliation(s)
- Elena B. Riel
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY10065
| | - Weiming Bu
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
| | - Thomas T. Joseph
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
| | - Leila Khajoueinejad
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY10065
| | - Roderic G. Eckenhoff
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
| | - Paul M. Riegelhaupt
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY10065
| |
Collapse
|
15
|
Gruget C, Reddy BG, Moore JM. A structural and mechanistic model for BSEP dysfunction in PFIC2 cholestatic disease. Commun Biol 2025; 8:531. [PMID: 40195555 PMCID: PMC11977275 DOI: 10.1038/s42003-025-07908-0] [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: 08/22/2024] [Accepted: 03/10/2025] [Indexed: 04/09/2025] Open
Abstract
BSEP (ABCB11) transports bile salts across the canalicular membrane of hepatocytes, where they are incorporated into bile. Biallelic mutations in BSEP can cause Progressive Familial Intrahepatic Cholestasis Type 2 (PFIC2), a rare pediatric disease characterized by hepatic bile acid accumulation leading to hepatotoxicity and, ultimately, liver failure. The most frequently occurring PFIC2 disease-causing mutations are missense mutations, which often display a phenotype with decreased protein expression and impaired maturation and trafficking to the canalicular membrane. To characterize the mutational effects on protein thermodynamic stability, we carried out biophysical characterization of 13 distinct PFIC2-associated variants using in-cell thermal shift (CETSA) measurements. These experiments reveal a cluster of residues localized to the NBD2-ICL2 interface, which exhibit severe destabilization relative to wild-type BSEP. A high-resolution (2.8 Å) cryo-EM structure provides a framework for rationalizing the CETSA results, revealing a novel, NBD2-localized mechanism through which the most severe missense patient mutations drive cholestatic disease. These findings suggest potential strategies for identifying mechanism-based small molecule correctors to address BSEP trafficking defects and advance novel therapies for PFIC2 and other cholestatic diseases.
Collapse
MESH Headings
- Humans
- Cholestasis, Intrahepatic/genetics
- Cholestasis, Intrahepatic/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 11/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 11/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 11/chemistry
- Mutation, Missense
- Cryoelectron Microscopy
- Models, Molecular
- Bile Acids and Salts/metabolism
- Mutation
Collapse
Affiliation(s)
| | | | - Jonathan M Moore
- Massachusetts Institute of Technology, Cambridge, MA, USA.
- Rectify Pharmaceuticals, Cambridge, MA, USA.
| |
Collapse
|
16
|
Huang X, Sun X, Wang Q, Zhang J, Wen H, Chen WJ, Zhu S. Structural insights into the diverse actions of magnesium on NMDA receptors. Neuron 2025; 113:1006-1018.e4. [PMID: 40010346 DOI: 10.1016/j.neuron.2025.01.021] [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: 08/23/2024] [Revised: 12/09/2024] [Accepted: 01/27/2025] [Indexed: 02/28/2025]
Abstract
Magnesium (Mg2+) is a key regulatory ion of N-methyl-ᴅ-aspartate (NMDA) receptors, including conferring them to function as coincidence detectors for excitatory synaptic transmission. However, the structural basis underlying the Mg2+ action on NMDA receptors remains unclear. Here, we report the cryo-EM structures of GluN1-N2B receptors and identify three distinct Mg2+-binding pockets. Specifically, site Ⅰ is located at the selectivity filter where an asparagine ring forms coordination bonds with Mg2+ and is responsible for the voltage-dependent block. Sites Ⅱ and Ⅲ are located at the N-terminal domain (NTD) of the GluN2B subunit and involved in the allosteric potentiation and inhibition, respectively. Site Ⅱ consists of three acidic residues, and the combination of three mutations abolishes the GluN2B-specific Mg2+ potentiation, while site Ⅲ overlaps with the Zn2+ pocket, and mutations here significantly reduce the inhibition. Our study enhances the understanding of multifaceted roles of Mg2+ in NMDA receptors and synaptic plasticity.
Collapse
Affiliation(s)
- Xuejing Huang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Xiaole Sun
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Jilin Zhang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Han Wen
- DP Technology, Beijing 100089, China; AI for Science Institute, Beijing 100085, China; State Key Laboratory of Medical Proteomics, Beijing 102206, China
| | - Wan-Jin Chen
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian 350005, China.
| | - Shujia Zhu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
17
|
Gram M, Warren JM, Madsen EL, Nielsen JC, Loland CJ, Bols M. Is Cocaine Protonated When it Binds to the Dopamine Transporter? JACS AU 2025; 5:1157-1172. [PMID: 40151268 PMCID: PMC11937975 DOI: 10.1021/jacsau.4c00952] [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: 10/08/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 03/29/2025]
Abstract
There has been much controversy about whether the well-known alkaloid and tertiary amine base cocaine (pK a = 8.5) binds to the human dopamine transporter (DAT) in its protonated form. Most potent DAT inhibitors are also strong amines-yet there are some noteworthy examples where neutral cocaine analogues have high affinity, while the quaternary ammonium analog of cocaine, cocaine methiodide, is a comparatively poor inhibitor. In this paper, we show that a fluorescent cocaine analog, with a lower pK a than cocaine, becomes protonated in the DAT binding site and conclude that similar behavior must be expected from cocaine. By determining the pK a of the aspartate residue in DAT believed to interact with the amine of cocaine, we are able to explain the apparently contradictory structure-activity data of cocaine analogues.
Collapse
Affiliation(s)
- Marie
L. Gram
- Department
of Chemistry, Faculty of Science, University
of Copenhagen, Copenhagen DK-2100, Denmark
| | - Julia M. Warren
- Department
of Chemistry, Faculty of Science, University
of Copenhagen, Copenhagen DK-2100, Denmark
| | - Emilie L. Madsen
- Department
of Chemistry, Faculty of Science, University
of Copenhagen, Copenhagen DK-2100, Denmark
| | - Jeppe C. Nielsen
- Laboratory
for Membrane Protein Dynamics, Department of Neuroscience, Faculty
of Health and Medical Sciences, University
of Copenhagen, Copenhagen DK-2200, Denmark
| | - Claus J. Loland
- Laboratory
for Membrane Protein Dynamics, Department of Neuroscience, Faculty
of Health and Medical Sciences, University
of Copenhagen, Copenhagen DK-2200, Denmark
| | - Mikael Bols
- Department
of Chemistry, Faculty of Science, University
of Copenhagen, Copenhagen DK-2100, Denmark
| |
Collapse
|
18
|
Zhang J, Duan J, Li W, Wang X, Ren S, Ye L, Liu F, Tian X, Xie Y, Huang Y, Sun Y, Song N, Li T, Cai X, Liu Z, Zhou H, Huang C, Li Y, Zhu S, Guo F. An antidepressant mechanism underlying the allosteric inhibition of GluN2D-incorporated NMDA receptors at GABAergic interneurons. SCIENCE ADVANCES 2025; 11:eadq0444. [PMID: 40043126 PMCID: PMC11881904 DOI: 10.1126/sciadv.adq0444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 01/30/2025] [Indexed: 05/13/2025]
Abstract
N-methyl-d-aspartate receptors (NMDARs), key excitatory ion channels, have gained attention as anti-depression targets. NMDARs consist of two GluN1 and two GluN2 subunits (2A-2D), which determine their pharmacological properties. Few compounds selectively targeting GluN2 subunits with antidepressant effects have been identified. Here, we present YY-23, a compound that selectively inhibits GluN2C- or GluN2D-containing NMDARs. Cryo-EM analysis revealed that YY-23 binds to the transmembrane domain of the GluN2D subunit. YY-23 primarily affects GluN2D-containing NMDARs on GABAergic interneurons in the prefrontal cortex, suppressing GABAergic neurotransmission and enhancing excitatory transmission. Behavioral assays demonstrate YY-23's rapid antidepressant effects in both stress-naïve and stress-exposed models, which are lost in mice with global or selective knockout of the grin2d gene in parvalbumin-positive interneurons. These findings highlight GluN2D-containing NMDARs on GABAergic interneurons as potential depression treatment targets.
Collapse
Affiliation(s)
- Jilin Zhang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Jinjin Duan
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Wei Li
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Xian Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Shimin Ren
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Luyu Ye
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Fang Liu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaoting Tian
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yang Xie
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yiming Huang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yidi Sun
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Nan Song
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Tianyu Li
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiang Cai
- Oujiang Laboratory, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhiqiang Liu
- Gynecology Hospital of Fudan University, No. 128, Shenyang Rd, Yangpu District, Shanghai 200082, China
| | - Hu Zhou
- Gynecology Hospital of Fudan University, No. 128, Shenyang Rd, Yangpu District, Shanghai 200082, China
| | - Chenggang Huang
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yang Li
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Shujia Zhu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China
| | - Fei Guo
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Gynecology Hospital of Fudan University, No. 128, Shenyang Rd, Yangpu District, Shanghai 200082, China
| |
Collapse
|
19
|
Ning Y, Xu R, Yu J, Ge J. Structural basis for catalytic mechanism of human phosphatidylserine synthase 1. Cell Discov 2025; 11:20. [PMID: 40044636 PMCID: PMC11882778 DOI: 10.1038/s41421-025-00775-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 01/20/2025] [Indexed: 03/09/2025] Open
Affiliation(s)
- Yingjie Ning
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Lingang Laboratory, Shanghai, China
| | - Ruisheng Xu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jie Yu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
- Shanghai Key Laboratory of Aging Studies, Shanghai, China.
| | - Jingpeng Ge
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
20
|
Schrecker M, Son Y, Planells-Cases R, Kar S, Vorobeva V, Schulte U, Fakler B, Jentsch TJ, Hite RK. Structural basis of ClC-3 inhibition by TMEM9 and PI(3,5)P 2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640562. [PMID: 40093093 PMCID: PMC11908120 DOI: 10.1101/2025.02.28.640562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
The trafficking and activity of endosomes relies on the exchange of chloride ions and protons by members of the CLC family of chloride channels and transporters, whose mutations are associated with numerous diseases. Despite their critical roles, the mechanisms by which CLC transporters are regulated are poorly understood. Here, we show that two related accessory β-subunits, TMEM9 and TMEM9B, directly interact with ClC-3, -4 and -5. Cryo-EM structures reveal that TMEM9 inhibits ClC-3 by sealing the cytosolic entrance to the Cl- ion pathway. Unexpectedly, we find that PI(3,5)P2 stabilizes the interaction between TMEM9 and ClC-3 and is required for proper regulation of ClC-3 by TMEM9. Collectively, our findings reveal that TMEM9 and PI(3,5)P2 collaborate to regulate endosomal ion homeostasis by modulating the activity of ClC-3.
Collapse
Affiliation(s)
- Marina Schrecker
- Structural Biology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Yeeun Son
- Structural Biology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
- BCMB Allied Program, Weill Cornell Graduate School; New York, NY, USA
| | - Rosa Planells-Cases
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP); Berlin, Germany
| | - Sumanta Kar
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP); Berlin, Germany
| | - Viktoriia Vorobeva
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP); Berlin, Germany
- Graduate program of the Free University; Berlin, Germany
| | - Uwe Schulte
- Institute of Physiology, Faculty of Medicine, University of Freiburg; Freiburg, Germany
- Logopharm GmbH; March-Buchheim, Germany
| | - Bernd Fakler
- Institute of Physiology, Faculty of Medicine, University of Freiburg; Freiburg, Germany
- Signalling Research Centers BIOSS and CIBSS; Freiburg, Germany
| | - Thomas J. Jentsch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP); Berlin, Germany
- Neurocure Cluster of Excellence, Charité Universitätsmedizin; Berlin, Germany
| | - Richard K. Hite
- Structural Biology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| |
Collapse
|
21
|
Cao Q, Ammerman A, Saimi M, Lin Z, Shen G, Chen H, Sun J, Chai M, Liu S, Hsu FF, Krezel AM, Gross ML, Xu J, Garcia BA, Liu B, Li W. Molecular basis of vitamin-K-driven γ-carboxylation at the membrane interface. Nature 2025; 639:816-824. [PMID: 39880037 DOI: 10.1038/s41586-025-08648-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/14/2025] [Indexed: 01/31/2025]
Abstract
The γ-carboxylation of glutamate residues enables Ca2+-mediated membrane assembly of protein complexes that support broad physiological functions, including haemostasis, calcium homeostasis, immune response and endocrine regulation1-4. Modulating γ-carboxylation levels provides prevalent treatments for haemorrhagic and thromboembolic diseases5. This unique post-translational modification requires vitamin K hydroquinone (KH2) to drive highly demanding reactions6 catalysed by the membrane-integrated γ-carboxylase (VKGC). Here, to decipher the underlying mechanisms, we determined cryo-electron microscopy structures of human VKGC in unbound form, with KH2 and four haemostatic and non-haemostatic proteins possessing propeptides and glutamate-rich domains in different carboxylation states. VKGC recognizes substrate proteins through knob-and-hole interactions with propeptides, thereby bringing tethered glutamate-containing segments for processive carboxylation within a large chamber that provides steric control. Propeptide binding also triggers a global conformational change to signal VKGC activation. Through sequential deprotonation and KH2 epoxidation, VKGC generates a free hydroxide ion as an exceptionally strong base that is required to deprotonate the γ-carbon of glutamate for CO2 addition. The diffusion of this superbase-protected and guided by a sealed hydrophobic tunnel-elegantly resolves the challenge of coupling KH2 epoxidation to γ-carboxylation across the membrane interface. These structural insights and extensive functional experiments advance membrane enzymology and propel the development of treatments for γ-carboxylation disorders.
Collapse
Affiliation(s)
- Qing Cao
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO, USA
| | - Aaron Ammerman
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO, USA
| | - Mierxiati Saimi
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO, USA
| | - Zongtao Lin
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO, USA
| | - Guomin Shen
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO, USA
- Department of Cell Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, People's Republic of China
| | - Huaping Chen
- Department of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Jie Sun
- Department of Chemistry, Washington University, St Louis, MO, USA
| | - Mengqi Chai
- Department of Chemistry, Washington University, St Louis, MO, USA
| | - Shixuan Liu
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO, USA
| | - Fong-Fu Hsu
- Department of Internal Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Andrzej M Krezel
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO, USA
| | - Michael L Gross
- Department of Chemistry, Washington University, St Louis, MO, USA
| | - Jinbin Xu
- Department of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Benjamin A Garcia
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO, USA
| | - Bin Liu
- The Hormel Institute, University of Minnesota, Austin, MN, USA.
| | - Weikai Li
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
22
|
Park SB, Lee NY, Lee EY, Kim S, Lee N, Roh EC, Kim YG, Kim HJ, Jin MS, Park CS, Kim YC. Discovery of Diphenyl Ether Derivatives as Novel BK Ca Channel Activators: Structure-Activity Relationship, Cryo-EM Complex Structures, and In Vivo Animal Studies. J Med Chem 2025; 68:4259-4286. [PMID: 39947888 DOI: 10.1021/acs.jmedchem.4c02008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
The BKCa channel, a large-conductance calcium-activated potassium channel, plays a crucial role in maintaining the homeostasis of the micturition cycle and airway-related functions. In this study, we optimized a novel BKCa channel activator, 4d, with a diphenyl ether structure identified from library screening. This led to the discovery of potent activators, 10b (EC50 = 0.12 μM, cell-based assay) and 51b, an orally bioavailable derivative. Compound 10b demonstrated potent in vivo efficacy in a spontaneous hypertensive rat (SHR) of urinary incontinence model, while compound 51b showed dose-dependent cough suppression efficacy with an ED50 of 11.8 mg/kg in a citric acid-induced cough model. Furthermore, we reported the cryo-electron microscopy (cryo-EM) structures of the BKCa channel in complex with 10b and 51b at resolutions of 2.8 and 3.4 Å. Based on structural analyses, we determined the binding sites and key interaction residues of 51b, which were validated via mutation studies.
Collapse
Affiliation(s)
- Soo Bin Park
- School of Life Science, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Na Young Lee
- School of Life Science, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Eun-Young Lee
- School of Life Science, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Subin Kim
- School of Life Science, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Narasaem Lee
- School of Life Science, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Eun Chae Roh
- College of Pharmacy, Dankook University, 119, Dandae-ro, Dongnam-gu, Cheonan-si, Chungcheongnam-do 31116, Republic of Korea
| | - Yoon Gyoon Kim
- College of Pharmacy, Dankook University, 119, Dandae-ro, Dongnam-gu, Cheonan-si, Chungcheongnam-do 31116, Republic of Korea
| | - Hee Jin Kim
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, 815, Hwarang-ro, Nowon-gu, Seoul 01795, Republic of Korea
| | - Mi Sun Jin
- School of Life Science, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Chul-Seung Park
- School of Life Science, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Yong-Chul Kim
- School of Life Science, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| |
Collapse
|
23
|
Roy-Chowdhury S, Jang S, Abderemane-Ali F, Naughton F, Grabe M, Minor DL. Structure of the human K 2P13.1 channel reveals a hydrophilic pore restriction and lipid cofactor site. Nat Struct Mol Biol 2025:10.1038/s41594-024-01476-3. [PMID: 40011746 DOI: 10.1038/s41594-024-01476-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/17/2024] [Indexed: 02/28/2025]
Abstract
Polyunsaturated fatty acid (PUFA) lipids modulate the neuronal and microglial leak potassium channel K2P13.1 (THIK1) and other voltage-gated ion channel (VGIC) superfamily members through poorly understood mechanisms. Here we present cryo-electron microscopy structures of human THIK1 and mutants, revealing a unique two-chamber aqueous inner cavity obstructed by a hydrophilic barrier important for gating, the flow restrictor, and a P1-M4 intersubunit interface lipid at a site, the PUFA site, corresponding to the K2P small-molecule modulator pocket. This overlap, together with functional studies, indicates that PUFA site lipids are THIK1 cofactors. Comparison with a PUFA-responsive VGIC, Kv7.1, reveals a shared modulatory role for the pore domain intersubunit interface, providing a framework for understanding PUFA action on the VGIC superfamily. Our findings reveal the distinct THIK1 architecture, highlight the importance of the P1-M4 interface for K2P control by natural and synthetic ligands and should aid in the development of THIK subfamily modulators for neuroinflammation and autism.
Collapse
Affiliation(s)
| | - Seil Jang
- Cardiovascular Research Institute, UCSF Medical Center, San Francisco, CA, USA
| | - Fayal Abderemane-Ali
- Cardiovascular Research Institute, UCSF Medical Center, San Francisco, CA, USA
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Fiona Naughton
- Cardiovascular Research Institute, UCSF Medical Center, San Francisco, CA, USA
| | - Michael Grabe
- Cardiovascular Research Institute, UCSF Medical Center, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Daniel L Minor
- Cardiovascular Research Institute, UCSF Medical Center, San Francisco, CA, USA.
- Departments of Biochemistry and Biophysics and Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA.
- California Institute for Quantitative Biomedical Research, University of California, San Francisco, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA.
- Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| |
Collapse
|
24
|
An W, Gao Y, Liu L, Bai Q, Zhao J, Zhao Y, Zhang XC. Structural basis of urea transport by Arabidopsis thaliana DUR3. Nat Commun 2025; 16:1782. [PMID: 39972035 PMCID: PMC11840088 DOI: 10.1038/s41467-025-56943-2] [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: 08/11/2024] [Accepted: 02/03/2025] [Indexed: 02/21/2025] Open
Abstract
Urea is a primary nitrogen source used as fertilizer in agricultural plant production and a crucial nitrogen metabolite in plants, playing an essential role in modern agriculture. In plants, DUR3 is a proton-driven high-affinity urea transporter located on the plasma membrane. It not only absorbs external low-concentration urea as a nutrient but also facilitates nitrogen transfer by recovering urea from senescent leaves. Despite its importance, the high-affinity urea transport mechanism in plants remains insufficiently understood. In this study, we determine the structures of Arabidopsis thaliana DUR3 in two different conformations: the inward-facing open state of the apo structure and the occluded urea-bound state, with overall resolutions of 2.8 Å and 3.0 Å, respectively. By comparing these structures and analyzing their functional characteristics, we elucidated how urea molecules are specifically recognized. In the urea-bound structure, we identified key titratable amino acid residues and proposed a model for proton involvement in urea transport based on structural and functional data. This study enhances our understanding of proton-driven urea transport mechanisms in DUR3.
Collapse
Affiliation(s)
- Weidong An
- Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences at Weifang, Weifang, Shandong, China
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yiwei Gao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Laihua Liu
- Department of Plant Nutrition, Key Laboratory of Plant and Soil Interactions of MEoC, College of Resources and Environmental Sciences, China Agricultural University, Beijing, China
| | - Qinru Bai
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jun Zhao
- Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences at Weifang, Weifang, Shandong, China.
| | - Yan Zhao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.
| | - Xuejun C Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
25
|
Agarwal S, Kim ED, Lee S, Simon A, Accardi A, Nimigean CM. Ball-and-chain inactivation of a human large conductance calcium-activated potassium channel. Nat Commun 2025; 16:1769. [PMID: 39971906 PMCID: PMC11840039 DOI: 10.1038/s41467-025-56844-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 02/03/2025] [Indexed: 02/21/2025] Open
Abstract
BK channels are large-conductance calcium (Ca2+)-activated potassium channels crucial for neuronal excitability, muscle contraction, and neurotransmitter release. The pore-forming (α) subunits co-assemble with auxiliary (β and γ) subunits that modulate their function. Previous studies demonstrated that the N-termini of β2-subunits can inactivate BK channels, but with no structural correlate. Here, we investigate BK β2-subunit inactivation using cryo-electron microscopy, electrophysiology and molecular dynamics simulations. We find that the β2 N-terminus occludes the pore only in the Ca2+-bound open state, via a ball-and-chain mechanism. The first three hydrophobic residues of β2 are crucial for occlusion, while the remainder of the N-terminus remains flexible. Neither the closed channel conformation obtained in the absence of Ca2+ nor an intermediate conformation found in the presence of Ca2+ show density for the N-terminus of the β2 subunit in their pore, likely due to narrower side access portals preventing their entry into the channel pore.
Collapse
Affiliation(s)
- Shubhangi Agarwal
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Elizabeth D Kim
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Sangyun Lee
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Alexander Simon
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Alessio Accardi
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Crina M Nimigean
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA.
- Department of Physiology and Biophysics, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA.
| |
Collapse
|
26
|
Houser A, Baconguis I. Structural insights into subunit-dependent functional regulation in epithelial sodium channels. Structure 2025; 33:349-362.e4. [PMID: 39667931 PMCID: PMC11805665 DOI: 10.1016/j.str.2024.11.013] [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] [Revised: 10/04/2024] [Accepted: 11/18/2024] [Indexed: 12/14/2024]
Abstract
Epithelial sodium channels (ENaCs) play a crucial role in Na+ reabsorption in mammals. To date, four subunits have been identified-α, β, γ, and δ-believed to form different heteromeric complexes. Currently, only the structure of the αβγ complex is known. To investigate the formation of channels with different subunit compositions and to determine how each subunit contributes to distinct channel properties, we co-expressed human δ, β, and γ. Using single-particle cryoelectron microscopy, we observed three distinct ENaC complexes. The structures unveil a pattern in which β and γ positions are conserved among the different complexes while the α position in αβγ trimer is occupied by either δ or another β. The δ subunit induces structural rearrangements in the γ subunit, which may contribute to the differences in channel activity between αβγ and δβγ channels. These structural changes provide molecular insights into how ENaC subunit composition modulates channel function.
Collapse
Affiliation(s)
- Alexandra Houser
- Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR 97239, USA
| | - Isabelle Baconguis
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
27
|
Kumar S, Jin F, Park SJ, Choi W, Keuning SI, Massimino RP, Vu S, Lü W, Du J. Convergent Agonist and Heat Activation of Nociceptor TRPM3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.634542. [PMID: 39896661 PMCID: PMC11785169 DOI: 10.1101/2025.01.23.634542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Detecting noxious heat is vital for survival, triggering pain responses that protect against harm1,2. The TRPM3 channel is a key nociceptor for sensing noxious heat and a promising therapeutic target for pain treatment and neurological disorders such as epilepsy3-11. Here, we functionally and structurally characterized TRPM3 in response to diverse stimuli: the synthetic superagonist CIM0216 Ref12, the anticonvulsant antagonist primidone13,14, and heat1,10,15. Our findings reveal that TRPM3 is intrinsically dynamic, with its intracellular domain (ICD) sampling both resting and activated states, though strongly favoring the resting state without stimulation. CIM0216 binds to the S1-S4 domain, inducing conformational changes in the ICD and shifting the equilibrium toward activation. Remarkably, heat induces similar ICD rearrangements, revealing a converged activation mechanism driven by chemical compounds and temperature. This mechanism is supported by functional data showing that mutations facilitating the ICD movement markedly increase the sensitivity of TRPM3 to both chemical and thermal signals. These findings establish a critical role of the ICD in temperature sensing in TRPM3, a mechanism likely conserved across the TRPM family. Finally, we show that primidone binds to the same site as CIM0216 but acts as an antagonist. This study provides a framework for understanding the thermal sensing mechanisms of temperature-sensitive ion channels and offers a structural foundation for developing TRPM3-target therapeutics for pain and neurological disorders.
Collapse
Affiliation(s)
- Sushant Kumar
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | | | - Sung Jin Park
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | | | - Sarah I. Keuning
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | | | | | - Wei Lü
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
- Department of Pharmacology, Northwestern University
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Juan Du
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
- Department of Pharmacology, Northwestern University
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| |
Collapse
|
28
|
Kalenderoglou IE, Nygaard A, Vogt CD, Turaev A, Pape T, Adams NBP, Newman AH, Loland CJ. Structural basis of vilazodone dual binding mode to the serotonin transporter. RESEARCH SQUARE 2025:rs.3.rs-5671197. [PMID: 39975905 PMCID: PMC11838746 DOI: 10.21203/rs.3.rs-5671197/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The serotonin transporter (SERT) plays a pivotal role in regulating serotonin (5-HT) signaling and is a key target in treating psychiatric disorders. SERT has a binding site (S1) for 5-HT that also serves as a high-affinity binding site for antidepressants. The antidepressant vilazodone has been shown to inhibit SERT by binding to an allosteric site. Here, we present the cryo-EM structure of SERT with vilazodone bound to the S1 site and extending towards the allosteric site. We systematically dissect the vilazodone molecule into fragments and find that the terminal indole ring is the key determinant for its high affinity to SERT. Further, unlike typical Na+-dependent SERT-selective antidepressants, vilazodone exhibits a dissociation constant (K D) for purified SERT in the nanomolar range both in the presence or absence of Na+. We substantiate this binding mode by exploring the conformational impact of vilazodone binding to SERT using site-specific insertion of the fluorescent non-canonical amino acid Anap. Our results offer novel molecular insight into the distinct pharmacological profile of a clinically used polymodal antidepressant.
Collapse
Affiliation(s)
- Iris E Kalenderoglou
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Nygaard
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Caleb D Vogt
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - Anton Turaev
- NanoTemper Technologies, Floessegasse 4, 81369 Munich, Germany
| | - Tillmann Pape
- Structural Molecular Biology Group, Protein Structure & Function Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Core Facility for Integrated Microscopy (CFIM), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - Claus J Loland
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
29
|
Freitas MM, Gouaux E. The bile acid-sensitive ion channel is gated by Ca 2+-dependent conformational changes in the transmembrane domain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.10.632481. [PMID: 39829759 PMCID: PMC11741473 DOI: 10.1101/2025.01.10.632481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The bile acid-sensitive ion channel (BASIC) is the least understood member of the mammalian epithelial Na+ channel/degenerin (ENaC/DEG) superfamily of ion channels, which are involved in a variety of physiological processes. While some members of this superfamily, including BASIC, are inhibited by extracellular Ca2+ (Ca2+ o), the molecular mechanism underlying Ca2+ modulation remains unclear. Here, by determining the structure of human BASIC in the presence and absence of Ca2+ using single particle cryo-electron microscopy (cryo-EM), we reveal Ca2+-dependent conformational changes in the transmembrane domain and β-linkers. Electrophysiological experiments further show that a glutamate residue in the extracellular vestibule of the pore underpins the Ca2+-binding site, whose occupancy determines the conformation of the pore and therefore ion flow through the channel. These results reveal the molecular principles governing gating of BASIC and its regulation by Ca2+ ions, demonstrating that Ca2+ ions modulate BASIC function via changes in protein conformation rather than solely from pore-block, as proposed for other members of this superfamily.
Collapse
Affiliation(s)
- Makayla M. Freitas
- Vollum Institute, Oregon Health and Science University, 3232 SW Research Drive, Portland, OR, USA
| | - Eric Gouaux
- Vollum Institute, Oregon Health and Science University, 3232 SW Research Drive, Portland, OR, USA
- Howard Hughes Medical Institute, Oregon Health and Science University, 3232 SW Research Drive, Portland, OR, USA
| |
Collapse
|
30
|
Younus I, Ford RC, Prince SM. A Structural Bioinformatics-Guided Study of Adenosine Triphosphate-Binding Cassette (ABC) Transporters and Their Substrates. MEMBRANES 2025; 15:20. [PMID: 39852261 PMCID: PMC11766626 DOI: 10.3390/membranes15010020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/10/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025]
Abstract
Adenosine triphosphate-binding cassette (ABC) transporters form a ubiquitous superfamily of integral membrane proteins involved in the translocation of substrates across membranes. Human ABC transporters are closely linked to the pathogenesis of diseases such as cancer, metabolic diseases, and Alzheimer's disease. In this study, four ABC transporters were chosen based on (I) their importance in humans and (II) their score in a structural bioinformatics screen aimed at the prediction of crystallisation propensity. The top-scoring ABC transporters' orthologs (Mus musculus-mouse ABCB5, Ailuropoda melanoleuca-giant panda ABCB6, Myotis lucifugus-little brown bat ABCG1 and Mus musculus ABCG4) were then expressed in Saccharomyces cerevisiae with a combined green fluorescent protein and polyhistidine tag, enabling visualisation and purification. After partial purification and in the presence of the detergent (n-dodecyl-β-D-maltoside), the kinetic parameters of the ATP hydrolysis reactions of the orthologs were determined, as well as the extent of stimulation of their activity when presented with putative substrates. We discuss the efficiency of such bioinformatics approaches and make suggestions for their improvement and wider application in membrane protein-structure determination.
Collapse
|
31
|
Zhang F, Li S, Wu H, Chen S. Cryo-EM structure and oligomerization of the human planar cell polarity core protein Vangl1. Nat Commun 2025; 16:135. [PMID: 39753546 PMCID: PMC11698883 DOI: 10.1038/s41467-024-55397-2] [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: 06/22/2024] [Accepted: 12/04/2024] [Indexed: 01/06/2025] Open
Abstract
Vangl is a planar cell polarity (PCP) core protein essential for aligned cell orientation along the epithelial plane perpendicular to the apical-basal direction, which is important for tissue morphogenesis, development and collective cell behavior. Mutations in Vangl are associated with developmental defects, including neural tube defects (NTDs), according to human cohort studies of sporadic and familial cases. The complex mechanisms underlying Vangl-mediated PCP signaling or Vangl-associated human congenital diseases have been hampered by the lack of molecular characterizations of Vangl. Here, we show biochemical and structural evidence that human Vangl1 oligomerizes as dimers of trimers, and that the dimerization of trimers promotes binding to the PCP effector Prickle1 (Pk1) in vitro. Mapping of human disease-associated point mutations suggests potential pathological mechanisms and paves the way for future studies on the importance of lipid binding, central vestibule and oligomerization of Vangl, thereby providing insights into the molecular mechanisms of the PCP signaling pathway.
Collapse
Affiliation(s)
- Fan Zhang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaobai Li
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Wu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai, China.
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China.
| | - Shanshuang Chen
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai, China.
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China.
| |
Collapse
|
32
|
Neuberger A, Shalygin A, Trofimov YA, Veretenenko II, Nadezhdin KD, Krylov NA, Gudermann T, Efremov RG, Chubanov V, Sobolevsky AI. Structure-function analyses of human TRPV6 ancestral and derived haplotypes. Structure 2025; 33:91-103.e5. [PMID: 39500315 PMCID: PMC11698656 DOI: 10.1016/j.str.2024.10.018] [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: 08/17/2024] [Revised: 09/29/2024] [Accepted: 10/09/2024] [Indexed: 12/12/2024]
Abstract
TRPV6 is a Ca2+ selective channel that mediates calcium uptake in the gut and contributes to the development and progression of human cancers. TRPV6 is represented by the ancestral and derived haplotypes that differ by three non-synonymous polymorphisms, located in the N-terminal ankyrin repeat domain (C157R), S1-S2 extracellular loop (M378V), and C-terminus (M681T). The ancestral and derived haplotypes were proposed to serve as genomic factors causing a different outcome for cancer patients of African ancestry. We solved cryoelectron microscopy (cryo-EM) structures of ancestral and derived TRPV6 in the open and calmodulin (CaM)-bound inactivated states. Neither state shows substantial structural differences caused by the non-synonymous polymorphisms. Functional properties assessed by electrophysiological recordings and Ca2+ uptake measurements, and water and ion permeation evaluated by molecular modeling also appear similar between the haplotypes. Therefore, ancestral and derived TRPV6 have similar structure and function, implying that other factors are responsible for the differences in susceptibility to cancer.
Collapse
Affiliation(s)
- Arthur Neuberger
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - Alexey Shalygin
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, 80336 Munich, Germany
| | - Yury A Trofimov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Irina I Veretenenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Kirill D Nadezhdin
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - Nikolay A Krylov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Thomas Gudermann
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, 80336 Munich, Germany; Comprehensive Pneumology Center, German Center for Lung Research, 81377 Munich, Germany
| | - Roman G Efremov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; National Research University Higher School of Economics, 101000 Moscow, Russia
| | - Vladimir Chubanov
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, 80336 Munich, Germany
| | - Alexander I Sobolevsky
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
33
|
Szollosi A. Functional Characterization of Ion Channels in Planar Lipid Bilayers. Methods Mol Biol 2025; 2908:141-161. [PMID: 40304908 DOI: 10.1007/978-1-0716-4434-8_10] [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] [Indexed: 05/02/2025]
Abstract
Ion channels form transmembrane protein pores allowing ion fluxes across biological membranes. Their function is indispensable for normal homeostasis; therefore, channelopathies lead to a large variety of diseases affecting almost all tissues and organs. In the last decades, the structure of hundreds of ion channels has been solved. In the majority of the solved structures the pore is closed, and for most of the protein preparations used in these studies, currently little data is available to support that the purified protein forms a functional ion channel capable of gating. Planar lipid bilayer technique is a powerful tool to address this issue. Purified ion channel protein is incorporated into a membrane bilayer separating two compartments in an experimental chamber. Channel currents are recorded between the two sides. Such setup allows characterization of gating and permeation properties of ion channels unbiased by auxiliary components present in native membranes. The planar lipid bilayer technique is thus discussed herein together with the robust protein expressing BacMam system.
Collapse
Affiliation(s)
- Andras Szollosi
- Semmelweis University, Department of Biochemistry, Budapest, Hungary.
- HUN-REN-SE Ion Channel Research Group, Budapest, Hungary.
- HCEMM-SE Molecular Channelopathies Research Group, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
34
|
Miyata Y, Takahashi K, Lee Y, Sultan CS, Kuribayashi R, Takahashi M, Hata K, Bamba T, Izumi Y, Liu K, Uemura T, Nomura N, Iwata S, Nagata S, Nishizawa T, Segawa K. Membrane structure-responsive lipid scrambling by TMEM63B to control plasma membrane lipid distribution. Nat Struct Mol Biol 2025; 32:185-198. [PMID: 39424995 PMCID: PMC11753361 DOI: 10.1038/s41594-024-01411-6] [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: 12/03/2023] [Accepted: 09/27/2024] [Indexed: 10/21/2024]
Abstract
Phospholipids are asymmetrically distributed in the plasma membrane (PM), with phosphatidylcholine and sphingomyelin abundant in the outer leaflet. However, the mechanisms by which their distribution is regulated remain unclear. Here, we show that transmembrane protein 63B (TMEM63B) functions as a membrane structure-responsive lipid scramblase localized at the PM and lysosomes, activating bidirectional lipid translocation upon changes in membrane curvature and thickness. TMEM63B contains two intracellular loops with palmitoylated cysteine residue clusters essential for its scrambling function. TMEM63B deficiency alters phosphatidylcholine and sphingomyelin distributions in the PM. Persons with heterozygous mutations in TMEM63B are known to develop neurodevelopmental disorders. We show that V44M, the most frequent substitution, confers constitutive scramblase activity on TMEM63B, disrupting PM phospholipid asymmetry. We determined the cryo-electron microscopy structures of TMEM63B in its open and closed conformations, uncovering a lipid translocation pathway formed in response to changes in the membrane environment. Together, our results identify TMEM63B as a membrane structure-responsive scramblase that controls PM lipid distribution and we reveal the molecular basis for lipid scrambling and its biological importance.
Collapse
Affiliation(s)
- Yugo Miyata
- Department of Medical Chemistry, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
- Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Katsuya Takahashi
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Yongchan Lee
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Cheryl S Sultan
- Department of Medical Chemistry, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
- Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Risa Kuribayashi
- Department of Medical Chemistry, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
- Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masatomo Takahashi
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Kosuke Hata
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Takeshi Bamba
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Izumi
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Kehong Liu
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoko Uemura
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Norimichi Nomura
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - So Iwata
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shigekazu Nagata
- Biochemistry and Immunology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Tomohiro Nishizawa
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan.
| | - Katsumori Segawa
- Department of Medical Chemistry, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
- Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
| |
Collapse
|
35
|
Kariapper L, Marathe IA, Niesman AB, Suino-Powell K, Min Chook Y, Wysocki VH, Worden EJ. Setdb1 and Atf7IP form a hetero-trimeric complex that blocks Setdb1 nuclear export. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.23.630145. [PMID: 39764026 PMCID: PMC11703210 DOI: 10.1101/2024.12.23.630145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Histone H3K9 methylation (H3K9me) by Setdb1 silences retrotransposons (rTE) by sequestering them in constitutive heterochromatin. Atf7IP is a constitutive binding partner of Setdb1 and is responsible for Setdb1 nuclear localization, activation and chromatin recruitment. However, structural details of the Setdb1/Atf7IP interaction have not been evaluated. We used Alphafold2 predictions and biochemical reconstitutions to show that one copy of Setdb1 and two copies of Atf7IP form a hetero-trimeric complex in vitro and in cells. We also find that Atf7IP self-associates, forming multimeric complexes that are resolved upon Setdb1 binding. Setdb1 binds to Atf7IP through coiled coil interactions that include both Setdb1 nuclear export signals (NES). Atf7IP directly competes with CRM1 to bind the Setdb1 NES motifs, explaining how Atf7IP prevents CRM1-mediated nuclear export of Setdb1. Setdb1 also forms hetero-trimeric complexes with the Atf7IP paralog Atf7IP2 and we show that Setdb1 can form mixed heterotrimers comprising one copy of each Setdb1, Atf7IP and Atf7IP2. Atf7IP and Atf7IP2 are co-expressed in many tissues suggesting that heterotrimers with different compositions of Atf7IP and Atf7IP2 may differentially regulate H3K9me by fine-tuning Setdb1 localization and activity.
Collapse
Affiliation(s)
- Leena Kariapper
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI, USA
| | - Ila A. Marathe
- School Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, GA USA
| | - Ashley B. Niesman
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX USA
| | - Kelly Suino-Powell
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI, USA
| | - Yuh Min Chook
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX USA
| | - Vicki H. Wysocki
- School Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, GA USA
| | - Evan J. Worden
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI, USA
| |
Collapse
|
36
|
Koval A, Boudou C, Katanaev VL. Challenging Reported Frizzled-Targeting Compounds in Selective Assays Reveals Lack of Functional Inhibition and Claimed Profiles. ACS Pharmacol Transl Sci 2024; 7:4144-4154. [PMID: 39698282 PMCID: PMC11650735 DOI: 10.1021/acsptsci.4c00570] [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: 09/25/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/20/2024]
Abstract
Selective inhibitors of Frizzled (FZD) GPCRs are highly sought after as potentially highly efficacious and safe treatments for cancer as well as tools in regenerative medicine and fundamental science. In recent years, there have been several reports claiming the identification of small molecule agents that are selective toward certain FZD proteins using a variety of approaches. However, the majority of these studies lacked a selective functional assay to validate their functionality. In this study, we describe the development and application of a selective assay for individual FZD proteins. Our findings indicate that the majority of reported compounds lack the capacity to inhibit the functioning of the claimed FZD proteins when stimulated by a Wnt ligand in the canonical pathway. Instead, the compounds demonstrate a broad range of off-target effects, including inhibition of downstream pathway component(s) (3235-0367, SRI35959, carbamazepine, niclosamide), lack of activity (FzM1), and surprising antagonism of firefly luciferase (F7H). The only compound that fulfills the expected selectivity profile is peptide Fz7-21. These results highlight the necessity of implementing rigorous testing of the screening-derived compounds in selective functional assays and are important for the field of drug discovery and development targeting the highly demanded Wnt-FZD pathway.
Collapse
Affiliation(s)
- Alexey Koval
- Department of Cell Physiology
and Metabolism, Translational Research Centre in Oncohaematology,
Faculty of Medicine, University of Geneva,1206 Geneva, Switzerland
| | - Cédric Boudou
- Department of Cell Physiology
and Metabolism, Translational Research Centre in Oncohaematology,
Faculty of Medicine, University of Geneva,1206 Geneva, Switzerland
| | - Vladimir L. Katanaev
- Department of Cell Physiology
and Metabolism, Translational Research Centre in Oncohaematology,
Faculty of Medicine, University of Geneva,1206 Geneva, Switzerland
| |
Collapse
|
37
|
Mittal A, Martin MF, Levin EJ, Adams C, Yang M, Provins L, Hall A, Procter M, Ledecq M, Hillisch A, Wolff C, Gillard M, Horanyi PS, Coleman JA. Structures of synaptic vesicle protein 2A and 2B bound to anticonvulsants. Nat Struct Mol Biol 2024; 31:1964-1974. [PMID: 38898101 DOI: 10.1038/s41594-024-01335-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 05/14/2024] [Indexed: 06/21/2024]
Abstract
Epilepsy is a common neurological disorder characterized by abnormal activity of neuronal networks, leading to seizures. The racetam class of anti-seizure medications bind specifically to a membrane protein found in the synaptic vesicles of neurons called synaptic vesicle protein 2 (SV2) A (SV2A). SV2A belongs to an orphan subfamily of the solute carrier 22 organic ion transporter family that also includes SV2B and SV2C. The molecular basis for how anti-seizure medications act on SV2s remains unknown. Here we report cryo-electron microscopy structures of SV2A and SV2B captured in a luminal-occluded conformation complexed with anticonvulsant ligands. The conformation bound by anticonvulsants resembles an inhibited transporter with closed luminal and intracellular gates. Anticonvulsants bind to a highly conserved central site in SV2s. These structures provide blueprints for future drug design and will facilitate future investigations into the biological function of SV2s.
Collapse
Affiliation(s)
- Anshumali Mittal
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Matthew F Martin
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Jonathan A Coleman
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
38
|
Gangwar SP, Yelshanskaya MV, Aktolun M, Yen LY, Newton TP, Strømgaard K, Kurnikova MG, Sobolevsky AI. Trapping of spermine, Kukoamine A, and polyamine toxin blockers in GluK2 kainate receptor channels. Nat Commun 2024; 15:10257. [PMID: 39592599 PMCID: PMC11599716 DOI: 10.1038/s41467-024-54538-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Kainate receptors (KARs) are a subtype of ionotropic glutamate receptor (iGluR) channels, a superfamily of ligand-gated ion channels which mediate the majority of excitatory neurotransmission in the central nervous system. KARs modulate neuronal circuits and plasticity during development and are implicated in neurological disorders, including epilepsy, depression, schizophrenia, anxiety, and autism. Calcium-permeable KARs undergo ion channel block, but the therapeutic potential of channel blockers remains underdeveloped, mainly due to limited structural knowledge. Here, we present closed-state structures of GluK2 KAR homotetramers in complex with ion channel blockers NpTx-8, PhTx-74, Kukoamine A, and spermine. We find that blockers reside inside the GluK2 ion channel pore, intracellular to the closed M3 helix bundle-crossing gate, with their hydrophobic heads filling the central cavity and positively charged polyamine tails spanning the selectivity filter. Molecular dynamics (MD) simulations of our structures illuminate interactions responsible for different affinity and binding poses of the blockers. Our structures elucidate the trapping mechanism of KAR channel block and provide a template for designing new blockers that can selectively target calcium-permeable KARs in neuropathologies.
Collapse
Affiliation(s)
- Shanti Pal Gangwar
- Department of Biochemistry and Molecular Biophysics, Columbia University, 650 West 168th Street, New York, NY, 10032, USA
| | - Maria V Yelshanskaya
- Department of Biochemistry and Molecular Biophysics, Columbia University, 650 West 168th Street, New York, NY, 10032, USA
| | - Muhammed Aktolun
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Laura Y Yen
- Department of Biochemistry and Molecular Biophysics, Columbia University, 650 West 168th Street, New York, NY, 10032, USA
- Cellular and Molecular Physiology and Biophysics Graduate Program, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY, 10032, USA
| | - Thomas P Newton
- Department of Biochemistry and Molecular Biophysics, Columbia University, 650 West 168th Street, New York, NY, 10032, USA
- Integrated Program in Cellular, Molecular and Biomedical Studies, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY, 10032, USA
| | - Kristian Strømgaard
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, DK-2100, Copenhagen, Denmark
| | - Maria G Kurnikova
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Alexander I Sobolevsky
- Department of Biochemistry and Molecular Biophysics, Columbia University, 650 West 168th Street, New York, NY, 10032, USA.
| |
Collapse
|
39
|
Martinez Fiesco JA, Li N, Alvarez de la Cruz A, Metcalfe RD, Beilina A, Cookson MR, Zhang P. 14-3-3 binding maintains the Parkinson's associated kinase LRRK2 in an inactive state. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624879. [PMID: 39605327 PMCID: PMC11601620 DOI: 10.1101/2024.11.22.624879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a central player in cellular signaling and a significant contributor to Parkinson's disease (PD) pathogenesis. 14-3-3 proteins are essential regulators of LRRK2, modulating its activity. Here, we present the cryo- electron microscopy structure of the LRRK2:14-3-3 2 autoinhibitory complex, showing that a 14-3-3 dimer stabilizes an autoinhibited LRRK2 monomer by binding to key phosphorylation sites and the COR-A and COR-B subdomains within the Roc-COR GTPase domain of LRRK2. This interaction locks LRRK2 in an inactive conformation, restricting LRR domain mobility and preventing dimerization and oligomer formation. Our mutagenesis studies reveal that PD-associated mutations at the COR:14-3-3 interface and within the GTPase domain reduce 14-3-3 binding, diminishing its inhibitory effect on LRRK2. These findings provide a structural basis for understanding how LRRK2 likely remains dormant within cells, illuminate aspects of critical PD biomarkers, and suggest therapeutic strategies to enhance LRRK2-14-3-3 interactions to treat PD and related disorders.
Collapse
|
40
|
Fiedorczuk K, Iordanov I, Mihályi C, Szollosi A, Csanády L, Chen J. The structures of protein kinase A in complex with CFTR: Mechanisms of phosphorylation and noncatalytic activation. Proc Natl Acad Sci U S A 2024; 121:e2409049121. [PMID: 39495916 PMCID: PMC11573500 DOI: 10.1073/pnas.2409049121] [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: 05/06/2024] [Accepted: 10/03/2024] [Indexed: 11/06/2024] Open
Abstract
Protein kinase A (PKA) is a key regulator of cellular functions by selectively phosphorylating numerous substrates, including ion channels, enzymes, and transcription factors. It has long served as a model system for understanding the eukaryotic kinases. Using cryoelectron microscopy, we present complex structures of the PKA catalytic subunit (PKA-C) bound to a full-length protein substrate, the cystic fibrosis transmembrane conductance regulator (CFTR)-an ion channel vital to human health. CFTR gating requires phosphorylation of its regulatory (R) domain. Unphosphorylated CFTR engages PKA-C at two locations, establishing two "catalytic stations" near to, but not directly involving, the R domain. This configuration, coupled with the conformational flexibility of the R domain, permits transient interactions of the eleven spatially separated phosphorylation sites. Furthermore, we determined two structures of the open-pore CFTR stabilized by PKA-C, providing a molecular basis to understand how PKA-C stimulates CFTR currents even in the absence of phosphorylation.
Collapse
Affiliation(s)
- Karol Fiedorczuk
- Laboratory of Membrane Biology and Biophysics, The Rockefeller University, New York, NY 10065
| | - Iordan Iordanov
- Department of Biochemistry, Semmelweis University, Budapest H-1094, Hungary
- Ion Channel Research Group, Hungarian Research Network - Semmelweis University, Budapest H-1094, Hungary
| | - Csaba Mihályi
- Department of Biochemistry, Semmelweis University, Budapest H-1094, Hungary
- Ion Channel Research Group, Hungarian Research Network - Semmelweis University, Budapest H-1094, Hungary
| | - Andras Szollosi
- Department of Biochemistry, Semmelweis University, Budapest H-1094, Hungary
- Ion Channel Research Group, Hungarian Research Network - Semmelweis University, Budapest H-1094, Hungary
| | - László Csanády
- Department of Biochemistry, Semmelweis University, Budapest H-1094, Hungary
- Ion Channel Research Group, Hungarian Research Network - Semmelweis University, Budapest H-1094, Hungary
| | - Jue Chen
- Laboratory of Membrane Biology and Biophysics, The Rockefeller University, New York, NY 10065
- HHMI, Chevy Chase, MD 20815
| |
Collapse
|
41
|
Qian M, Xu Y, Shu HJ, Chen ZW, Wang L, Zorumski CF, Evers AS, Mennerick S, Covey DF. Synthesis and evaluation of photoaffinity labeling reagents for identifying binding sites of sulfated neurosteroids on NMDA and GABA A receptors. RSC Adv 2024; 14:36352-36369. [PMID: 39539530 PMCID: PMC11559520 DOI: 10.1039/d4ra07074g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
The endogenous neurosteroids dehydroepiandrosterone sulfate (DHEAS) and pregnenolone sulfate (PS) are allosteric modulators of γ-aminobutyric acid type A (GABAA) and N-methyl-d-aspartate (NMDA) type glutamate receptors. Analogues of these endogenous steroid sulfates can be either positive or negative allosteric modulators (PAMs or NAMs, respectively) of these receptors, but there is limited information about the steroid-protein binding interactions that mediate these effects and photoaffinity labeling reagents (PALs) of sulfated steroids have not been reported previously. The synthesis of a panel of ten sulfated steroid analogues containing a diazirine group, five of which also contain an alkyne group for click chemistry reactions, for use in photoaffinity labeling studies to identify binding sites for steroid sulfates that are either positive or negative allosteric modulators is reported. Electrophysiological measurements on cultured rat hippocampal neurons were made to determine the modes of allosteric modulation in comparison to those of PS on both receptors. PALs with the activity profile of PS (NMDA PAM, GABAA NAM) were identified. Unexpectedly, PALs with PAM activity at both receptors were also found. Photolabeling of both receptors by two of the PALs was performed to demonstrate their utility, and by inference those of the other PALs, for future studies to identify binding sites for endogenous steroid sulfates on both receptors.
Collapse
Affiliation(s)
- Mingxing Qian
- Department of Developmental Biology, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
| | - Yuanjian Xu
- Department of Developmental Biology, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
| | - Hong-Jin Shu
- Department of Psychiatry, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
| | - Zi-Wei Chen
- Department of Anesthesiology, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
| | - Lei Wang
- Department of Anesthesiology, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education China
| | - Charles F Zorumski
- Department of Psychiatry, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
| | - Alex S Evers
- Department of Developmental Biology, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Department of Anesthesiology, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
| | - Steven Mennerick
- Department of Psychiatry, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
| | - Douglas F Covey
- Department of Developmental Biology, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Department of Psychiatry, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Department of Anesthesiology, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
| |
Collapse
|
42
|
Ren Q, Wang J, Idikuda V, Zhang S, Shin J, Ludlam WG, Real Hernandez LM, Levental I, Martemyanov K, Chanda B, Bao H. DeFrND: detergent-free reconstitution into native nanodiscs with designer membrane scaffold peptides. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.07.622281. [PMID: 39574650 PMCID: PMC11580953 DOI: 10.1101/2024.11.07.622281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Membrane scaffold proteins-based nanodiscs (NDs) have facilitated unprecedented structural and biophysical analysis of membrane proteins in a near-native lipid environment. However, successful reconstitution of membrane proteins in NDs requires prior solubilization and purification in detergents, which may impact their physiological structure and function. Furthermore, the detergent-mediated reconstitution of NDs is unlikely to recapitulate the precise composition or asymmetry of native membranes. To circumvent this fundamental limitation of traditional ND technology, we herein describe the development of membrane-solubilizing peptides to directly extract membrane proteins from native cell membranes into nanoscale discoids. By systematically protein engineering and screening, we created a new class of chemically modified Apolipoprotein-A1 mimetic peptides to enable the formation of detergent-free NDs (DeFrNDs) with high efficiency. NDs generated with these engineered membrane scaffold peptides are suitable for obtaining high-resolution structures using single-particle cryo-EM with native lipids. To further highlight the versatility of DeFrNDs, we directly extract a sampling of membrane signaling proteins with their surrounding native membranes for biochemical and biophysical interrogations.
Collapse
|
43
|
Miyashita Y, Moriya T, Kato T, Kawasaki M, Yasuda S, Adachi N, Suzuki K, Ogasawara S, Saito T, Senda T, Murata T. Improved higher resolution cryo-EM structures reveal the binding modes of hERG channel inhibitors. Structure 2024; 32:1926-1935.e3. [PMID: 39321803 DOI: 10.1016/j.str.2024.08.021] [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: 07/05/2024] [Revised: 08/06/2024] [Accepted: 08/29/2024] [Indexed: 09/27/2024]
Abstract
During drug discovery, it is crucial to exclude compounds with toxic effects. The human ether-à-go-go-related gene (hERG) channel is essential for maintaining cardiac repolarization and is a critical target in drug safety evaluation due to its role in drug-induced arrhythmias. Inhibition of the hERG channel can lead to severe cardiac issues, including Torsades de Pointes tachycardia. Understanding hERG inhibition mechanisms is essential to avoid these toxicities. Several structural studies have elucidated the interactions between inhibitors and hERG. However, orientation and resolution issues have so far limited detailed insights. Here, we used digitonin to analyze the apo state of hERG, which resolved orientation issues and improved the resolution. We determined the structure of hERG bound to astemizole, showing a clear map in the pore pathway. Using this strategy, we also analyzed the binding modes of E-4031 and pimozide. These insights into inhibitor interactions with hERG may aid safer drug design and enhance cardiac safety.
Collapse
Affiliation(s)
- Yasuomi Miyashita
- Department of Developmental Biology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo, Chiba 260-8670, Japan; Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage, Chiba 263-8522, Japan
| | - Toshio Moriya
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), 1-1 Oho, Tsukuba 305-0801, Japan
| | - Takafumi Kato
- Department of Biochemistry, University of Oxford, South Parks Rd, Oxford OX13QC, UK
| | - Masato Kawasaki
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), 1-1 Oho, Tsukuba 305-0801, Japan
| | - Satoshi Yasuda
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage, Chiba 263-8522, Japan; Membrane Protein Research Center, Chiba University, 1-33 Yayoi-cho, Inage, Chiba 263-8522, Japan
| | - Naruhiko Adachi
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), 1-1 Oho, Tsukuba 305-0801, Japan; Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Kano Suzuki
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage, Chiba 263-8522, Japan; Membrane Protein Research Center, Chiba University, 1-33 Yayoi-cho, Inage, Chiba 263-8522, Japan
| | - Satoshi Ogasawara
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage, Chiba 263-8522, Japan; Membrane Protein Research Center, Chiba University, 1-33 Yayoi-cho, Inage, Chiba 263-8522, Japan
| | - Tetsuichiro Saito
- Department of Developmental Biology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo, Chiba 260-8670, Japan
| | - Toshiya Senda
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), 1-1 Oho, Tsukuba 305-0801, Japan
| | - Takeshi Murata
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage, Chiba 263-8522, Japan; Membrane Protein Research Center, Chiba University, 1-33 Yayoi-cho, Inage, Chiba 263-8522, Japan.
| |
Collapse
|
44
|
Dhingra S, Chopade PM, Vinnakota R, Kumar J. Functional implications of the exon 9 splice insert in GluK1 kainate receptors. eLife 2024; 12:RP89755. [PMID: 39504124 PMCID: PMC11540303 DOI: 10.7554/elife.89755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024] Open
Abstract
Kainate receptors are key modulators of synaptic transmission and plasticity in the central nervous system. Different kainate receptor isoforms with distinct spatiotemporal expressions have been identified in the brain. The GluK1-1 splice variant receptors, which are abundant in the adult brain, have an extra fifteen amino acids inserted in the amino-terminal domain (ATD) of the receptor resulting from alternative splicing of exon 9. However, the functional implications of this post-transcriptional modification are not yet clear. We employed a multi-pronged approach using cryogenic electron microscopy, electrophysiology, and other biophysical and biochemical tools to understand the structural and functional impact of this splice insert in the extracellular domain of GluK1 receptors. Our study reveals that the splice insert alters the key gating properties of GluK1 receptors and their modulation by the cognate auxiliary Neuropilin and tolloid-like (Neto) proteins 1 and 2. Mutational analysis identified the role of crucial splice residues that influence receptor properties and their modulation. Furthermore, the cryoEM structure of the variant shows that the presence of exon 9 in GluK1 does not affect the receptor architecture or domain arrangement in the desensitized state. Our study thus provides the first detailed structural and functional characterization of GluK1-1a receptors, highlighting the role of the splice insert in modulating receptor properties and their modulation.
Collapse
Affiliation(s)
- Surbhi Dhingra
- Laboratory of Membrane Protein Biology, National Centre for Cell SciencePuneIndia
| | - Prachi M Chopade
- Laboratory of Membrane Protein Biology, National Centre for Cell SciencePuneIndia
| | - Rajesh Vinnakota
- Laboratory of Membrane Protein Biology, National Centre for Cell SciencePuneIndia
| | - Janesh Kumar
- Laboratory of Membrane Protein Biology, National Centre for Cell SciencePuneIndia
- Laboratory of Membrane Protein Biology, CSIR-Centre for Cellular and Molecular BiologyHyderabadIndia
| |
Collapse
|
45
|
Mugo AN, Chou R, Qin F. Protein Dynamics Underlies Strong Temperature Dependence of Heat Receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621882. [PMID: 39574614 PMCID: PMC11580892 DOI: 10.1101/2024.11.04.621882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Ion channels are generally allosteric proteins, involving specialized stimulus sensor domains conformationally linked to the gate to drive channel opening. Temperature receptors are a group of ion channels from the transient receptor potential (TRP) family. They exhibit an unprecedentedly strong temperature dependence and are responsible for temperature sensing in mammals. Despite intensive studies, however, the nature of the temperature sensor domain in these channels remains elusive. By direct calorimetry of TRPV1 proteins, we have recently provided a proof of principle that temperature sensing by ion channels may diverge from the conventional allosterity theory; rather it is intimately linked to inherent thermal instability of channel proteins. Here we tackle the generality of the hypothesis and provide key molecular evidences on the coupling of thermal transitions in the channels. We show that while wild-type channels possess a single concerted thermal transition peak, the chimera, in which strong temperature dependence becomes disrupted, results in multi-transition peaks, and the activation enthalpies are accordingly reduced. The data show that the coupling with protein unfolding drives up the energy barrier of activation, leading to a strong temperature dependence of opening. Furthermore, we pinpoint the proximal N-terminus of the channels as a linchpin in coalescing different parts of the channels into concerted activation. Thus, we suggest that coupled interaction networks in proteins underlie the strong temperature dependence of temperature receptors.
Collapse
Affiliation(s)
- Andrew Njagi Mugo
- Department of Physiology and Biophysical Sciences, State University of New York at Buffalo, Buffalo, NY 14214
| | - Ryan Chou
- Trinity College of Arts and Sciences, Duke University
| | - Feng Qin
- Department of Physiology and Biophysical Sciences, State University of New York at Buffalo, Buffalo, NY 14214
| |
Collapse
|
46
|
Dai Y, Lee CH. Transport mechanism and structural pharmacology of human urate transporter URAT1. Cell Res 2024; 34:776-787. [PMID: 39245778 PMCID: PMC11528023 DOI: 10.1038/s41422-024-01023-1] [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: 07/02/2024] [Accepted: 08/20/2024] [Indexed: 09/10/2024] Open
Abstract
Urate is an endogenous product of purine metabolism in the liver. High urate levels in the blood lead to gout, a very common and painful inflammatory arthritis. Excreted urate is reabsorbed in the kidney mainly by URAT1 antiporter, a key target for anti-gout drugs. To uncover the mechanisms of urate transport and drug inhibition, we determined cryo-EM structures of human URAT1 with urate, counter anion pyrazinoate, or anti-gout drugs of different chemotypes - lesinurad, verinurad, and dotinurad. We captured the outward-to-inward transition of URAT1 during urate uptake, revealing that urate binds in a phenylalanine-rich pocket and engages with key gating residues to drive the transport cycle. In contrast to the single binding site for urate, pyrazinoate interacts with three distinct, functionally relevant sites within URAT1, a mechanism that has not yet been observed in other anion antiporters. In addition, we found that while all three drugs compete with substrates and halt the transport cycle, verinurad and dotinurad further hijack gating residues to achieve high potency. These insights advance our understanding of organic anion transport and provide a foundation for designing improved gout therapeutics.
Collapse
Affiliation(s)
- Yaxin Dai
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Chia-Hsueh Lee
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
47
|
Hale WD, Montaño Romero A, Gonzalez CU, Jayaraman V, Lau AY, Huganir RL, Twomey EC. Allosteric competition and inhibition in AMPA receptors. Nat Struct Mol Biol 2024; 31:1669-1679. [PMID: 38834914 PMCID: PMC11563869 DOI: 10.1038/s41594-024-01328-0] [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: 01/08/2024] [Accepted: 05/03/2024] [Indexed: 06/06/2024]
Abstract
Excitatory neurotransmission is principally mediated by α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-subtype ionotropic glutamate receptors (AMPARs). Negative allosteric modulators are therapeutic candidates that inhibit AMPAR activation and can compete with positive modulators to control AMPAR function through unresolved mechanisms. Here we show that allosteric inhibition pushes AMPARs into a distinct state that prevents both activation and positive allosteric modulation. We used cryo-electron microscopy to capture AMPARs bound to glutamate, while a negative allosteric modulator, GYKI-52466, and positive allosteric modulator, cyclothiazide, compete for control of the AMPARs. GYKI-52466 binds in the ion channel collar and inhibits AMPARs by decoupling the ligand-binding domains from the ion channel. The rearrangement of the ligand-binding domains ruptures the cyclothiazide site, preventing positive modulation. Our data provide a framework for understanding allostery of AMPARs and for rational design of therapeutics targeting AMPARs in neurological diseases.
Collapse
Affiliation(s)
- W Dylan Hale
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alejandra Montaño Romero
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cuauhtemoc U Gonzalez
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Vasanthi Jayaraman
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Albert Y Lau
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Richard L Huganir
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Edward C Twomey
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- The Beckman Center for Cryo-EM at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA.
| |
Collapse
|
48
|
Jin F, Lin YY, Wang RC, Xie TX, Zhao Y, Shen C, Sheng D, Ichikawa M, Yu Y, Wang J, Hattori M. Cryo-EM structure of the zinc-activated channel (ZAC) in the Cys-loop receptor superfamily. Proc Natl Acad Sci U S A 2024; 121:e2405659121. [PMID: 39441630 PMCID: PMC11536092 DOI: 10.1073/pnas.2405659121] [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: 03/20/2024] [Accepted: 08/28/2024] [Indexed: 10/25/2024] Open
Abstract
Cys-loop receptors are a large superfamily of pentameric ligand-gated ion channels with various physiological roles, especially in neurotransmission in the central nervous system. Among them, zinc-activated channel (ZAC) is a Zn2+-activated ion channel that is widely expressed in the human body and is conserved among eukaryotes. Due to its gating by extracellular Zn2+, ZAC has been considered a Zn2+ sensor, but it has undergone minimal structural and functional characterization since its molecular cloning. Among the families in the Cys-loop receptor superfamily, only the structure of ZAC has yet to be determined. Here, we determined the cryo-EM structure of ZAC in the apo state and performed structure-based mutation analyses. We identified a few residues in the extracellular domain whose mutations had a mild impact on Zn2+ sensitivity. The constriction site in the ion-conducting pore differs from the one in other Cys-loop receptor structures, and further mutational analysis identified a key residue that is important for ion selectivity. In summary, our work provides a structural framework for understanding the ion-conducting mechanism of ZAC.
Collapse
Affiliation(s)
- Fei Jin
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Bioactive Small Molecules, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai200438, China
| | - Yi-Yu Lin
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing200098, China
| | - Ru-Chun Wang
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing200098, China
| | - Tang-Xuan Xie
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing200098, China
| | - Yimeng Zhao
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Bioactive Small Molecules, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai200438, China
- Human Phenome Institute, Fudan University, Shanghai201203, China
| | - Cheng Shen
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Bioactive Small Molecules, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai200438, China
| | - Danqi Sheng
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Bioactive Small Molecules, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai200438, China
| | - Muneyoshi Ichikawa
- State Key Laboratory of Genetic Engineering, Department of Biochemistry and Biophysics, School of Life Sciences, Fudan University, Shanghai200438, China
| | - Ye Yu
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing200098, China
| | - Jin Wang
- Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing200098, China
| | - Motoyuki Hattori
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Bioactive Small Molecules, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai200438, China
| |
Collapse
|
49
|
Yu H, Wang W. Modulation of heteromeric glycine receptor function through high concentration clustering. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618879. [PMID: 39464082 PMCID: PMC11507885 DOI: 10.1101/2024.10.17.618879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Ion channels are targeted by many drugs for treating neurological, musculoskeletal, renal and other diseases. These drugs bind to and alter the function of individual channels to achieve desired therapeutic effects. However, many ion channels function in high concentration clusters in their native environment. It is unclear if and how clustering modulates ion channel function. Human heteromeric glycine receptors (GlyRs) are the major inhibitory neurotransmitter receptors in the spinal cord and are active targets for developing chronic pain medications. We show that the α2β heteromeric GlyR assembles with the master postsynaptic scaffolding gephyrin (GPHN) into micron-sized clustered at the plasma membrane after heterologous expression. The inhibitory trans- synaptic adhesion protein neuroligin-2 (NL2) further increases both the cluster sizes and GlyR concentration. The apparent glycine affinity increases monotonically as a function of GlyR concentration but not with cluster size. We also show that ligand re-binding to adjacent GlyRs alters kinetics but not chemical equilibrium. A positively charged N- terminus sequence of the GlyR β subunit was further identified essential for glycine affinity modulation through clustering. Taken together, we propose a mechanism where clustering enhances local electrostatic potential, which in turn concentrates ions and ligands, modulating the function of GlyR. This mechanism is likely universal across ion channel clusters found ubiquitously in biology and provides new perspectives in possible pharmaceutical development.
Collapse
|
50
|
Boyd R, Jaqaman K, Wang W. Weaker neuroligin 2 - neurexin 1β interaction tethers membranes and signal synaptogenesis through clustering. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.16.618688. [PMID: 39464163 PMCID: PMC11507839 DOI: 10.1101/2024.10.16.618688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Single-pass transmembrane proteins neuroligin (NL) and neurexin (NRX) constitute a pair of synaptic adhesion molecules (SAMs) that are essential for the formation of functional synapses. Binding affinities vary by ∼ 1000 folds between arrays of NL and NRX subtypes, which contribute to chemical and spatial specificities. Current structures are obtained with truncated extracellular domains of NL and NRX and are limited to the higher-affinity NL1/4-NRX complexes. How NL-NRX interaction leads to functional synapses remains unknown. Here we report structures of full-length NL2 alone, and in complex with NRX1β in several conformations, which has the lowest affinity among major NL-NRX subtypes. We show how conformational flexibilities may help in adapting local membrane geometry, and reveal mechanisms underlying variations in NL-NRX affinities modulation. We further show that, despite lower affinity, NL2-NRX1β interaction alone is capable of tethering different lipid membranes in total reconstitution, and that NL2 and NRX1β cluster at inter-cellular junctions without the need of other synaptic components. In addition, NL2 combines with the master post-synaptic scaffolding protein gephyrin and clusters neurotransmitter receptors at cellular membrane. These findings suggest dual roles of NL2 - NRX1β interaction - both as mechanical tether, and as signaling receptors, to ensure correct spatial and chemical coordination between two cells to generate function synapses.
Collapse
|