1
|
Nakao H, Tsujii T, Saito H, Ikeda K, Nakano M. Synergistic effects of hydrophilic residues in the transmembrane region on lipid scrambling activity of dimeric helices. Colloids Surf B Biointerfaces 2025; 251:114612. [PMID: 40086211 DOI: 10.1016/j.colsurfb.2025.114612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/10/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Phospholipid scramblases promote lipid transbilayer movement (flip-flop) in the plasma membrane, which is involved in a wide range of cellular functions, such as phagocytosis and blood coagulation. One structural characteristic of scramblases and model lipid scrambling peptides is the presence of hydrophilic residues in their transmembrane domains. These hydrophilic regions are considered the active sites through which lipid polar headgroups pass during the translocation process. However, how the structural arrangement of hydrophilic residues results in strong lipid scrambling activities in scramblases needs to be investigated, because the effects of a single hydrophilic residue on lipid scrambling are much lower than the activity of natural scramblases. Here, we developed double-spanning transmembrane peptides containing varying numbers of Gln residues. A combination of lipid vesicle experiments and molecular dynamics simulations indicates that lipid scrambling activities are synergistically enhanced by the proximity between planes created by Gln residues aligned parallel to the helix and that interactions between Gln and Trp residues stabilize the strongly active structures. The contribution of Gln residues to lipid scrambling activity suggests that the alignment and proximity of hydrophilic residues in the transmembrane region is one of the mechanisms of lipid scrambling by natural scramblases. This study provides clues for the energetic and structural mechanisms of lipid scrambling and for the design of artificial phospholipid scramblases.
Collapse
Affiliation(s)
- Hiroyuki Nakao
- Department of Biointerface Chemistry, Faculty of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | - Toshiki Tsujii
- Department of Biointerface Chemistry, Faculty of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Hiroaki Saito
- Faculty of Pharmaceutical Sciences, Hokuriku University, 3 Kanagawamachi, Kanazawa, Ishikawa 920-1181, Japan
| | - Keisuke Ikeda
- Department of Biointerface Chemistry, Faculty of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Minoru Nakano
- Department of Biointerface Chemistry, Faculty of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| |
Collapse
|
2
|
Kostritskii AY, Kostritskaia Y, Dmitrieva N, Stauber T, Machtens JP. Calcium-activated chloride channel TMEM16A opens via pi-helical transition in transmembrane segment 4. Proc Natl Acad Sci U S A 2025; 122:e2421900122. [PMID: 40299692 DOI: 10.1073/pnas.2421900122] [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: 10/22/2024] [Accepted: 03/27/2025] [Indexed: 05/01/2025] Open
Abstract
TMEM16A is a Ca2+-activated Cl- channel that has crucial roles in various physiological and pathological processes. However, the structure of the open state of the channel and the mechanism of Ca2+-induced pore opening have remained elusive. Using extensive molecular dynamics simulations, protein structure prediction, and patch-clamp electrophysiology, we demonstrate that TMEM16A opens a hydrated Cl--conductive pore via a pi-helical transition in transmembrane segment 4 (TM4). We also describe a coupling mechanism that links pi-helical transition and pore opening to the Ca2+-induced conformational changes in TMEM16A. Furthermore, we designed a pi-helix-stabilizing mutation (I551P) that facilitates TMEM16A activation, revealing atomistic details of the ion-conduction mechanism. Finally, AlphaFold2 structure predictions revealed the importance of the pi helix in TM4 to structure-function relations in TMEM16 and the related OSCA/TMEM63 family, further highlighting the relevance of dynamic pi helices for gating in various ion channels.
Collapse
Affiliation(s)
- Andrei Y Kostritskii
- Institute of Biological Information Processing (IBI-1), Molekular- und Zellphysiologie, Forschungszentrum Jülich, Jülich 52428, Germany
| | - Yulia Kostritskaia
- Institute for Molecular Medicine, MSH Medical School Hamburg, Hamburg 20457, Germany
| | - Natalia Dmitrieva
- Institute of Biological Information Processing (IBI-1), Molekular- und Zellphysiologie, Forschungszentrum Jülich, Jülich 52428, Germany
| | - Tobias Stauber
- Institute for Molecular Medicine, MSH Medical School Hamburg, Hamburg 20457, Germany
| | - Jan-Philipp Machtens
- Institute of Biological Information Processing (IBI-1), Molekular- und Zellphysiologie, Forschungszentrum Jülich, Jülich 52428, Germany
- Institute of Neurophysiology, Hannover Medical School, Hannover 30625, Germany
| |
Collapse
|
3
|
Javanainen M, Šimek J, Tranter D, O'Keefe S, Karki S, Biriukov D, Šachl R, Paavilainen VO. Lipid Scrambling Pathways in the Sec61 Translocon Complex. J Am Chem Soc 2025. [PMID: 40325981 DOI: 10.1021/jacs.4c11142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Cellular homeostasis depends on the rapid, ATP-independent translocation of newly synthesized lipids across the endoplasmic reticulum (ER) membrane. Lipid translocation is facilitated by membrane proteins known as scramblases, a few of which have recently been identified in the ER. Our previous structure of the translocon-associated protein (TRAP) bound to the Sec61 translocation channel revealed local membrane thinning, suggesting that the Sec61/TRAP complex might be involved in lipid scrambling. Using complementary fluorescence spectroscopy assays, we detected nonselective scrambling by reconstituted translocon complexes. This activity was unaffected by Sec61 inhibitors that block its lateral gate, suggesting a second lipid scrambling pathway within the complex. Molecular dynamics simulations indicate that the trimeric TRAP subunit forms this alternative route, facilitating lipid translocation via a "credit card" mechanism, using a crevice lined with polar residues to shield lipid head groups from the hydrophobic membrane interior. Kinetic and thermodynamic analyses confirmed that local membrane thinning enhances scrambling efficiency and that both Sec61 and TRAP scramble phosphatidylcholine faster than phosphatidylethanolamine and phosphatidylserine, reflecting the intrinsic lipid flip-flop tendencies of these lipid species. As the Sec61 scrambling site lies in the lateral gate region, it is likely inaccessible during protein translocation, in line with our experiments on Sec61-inhibited samples. Hence, our findings suggest that the metazoan-specific trimeric TRAP bundle is a viable candidate for lipid scrambling activity that is insensitive to the functional state of the translocon.
Collapse
Affiliation(s)
- Matti Javanainen
- Unit of Physics, University of Tampere, FI-33720 Tampere, Finland
- Institute of Biotechnology, HiLIFE, University of Helsinki, FI-00790 Helsinki, Finland
| | - Jan Šimek
- J. Heyrovský Institute of Physical Chemistry, CZ-18223 Prague 8, Czech Republic
- Department of Physical and Macromolecular Chemistry, Charles University, Hlavova 8, CZ-12800 Prague 2, Czech Republic
| | - Dale Tranter
- Institute of Biotechnology, HiLIFE, University of Helsinki, FI-00790 Helsinki, Finland
| | - Sarah O'Keefe
- Institute of Biotechnology, HiLIFE, University of Helsinki, FI-00790 Helsinki, Finland
| | - Sudeep Karki
- Institute of Biotechnology, HiLIFE, University of Helsinki, FI-00790 Helsinki, Finland
- Onego Bio, Hämeentie 157, FI-00560 Helsinki, Finland
| | - Denys Biriukov
- Central European Institute of Technology, Masaryk University, Kamenice 5, CZ-62500 Brno, Czech Republic
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, CZ-62500 Brno, Czech Republic
| | - Radek Šachl
- J. Heyrovský Institute of Physical Chemistry, CZ-18223 Prague 8, Czech Republic
| | - Ville O Paavilainen
- Institute of Biotechnology, HiLIFE, University of Helsinki, FI-00790 Helsinki, Finland
| |
Collapse
|
4
|
Zhang S, Luo S, Zhang H, Xiao Q. Transmembrane protein 16A in the digestive diseases: A review of its physiology, pharmacology, and therapeutic opportunities. Int J Biol Macromol 2025; 310:143598. [PMID: 40300686 DOI: 10.1016/j.ijbiomac.2025.143598] [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: 01/20/2025] [Revised: 04/14/2025] [Accepted: 04/26/2025] [Indexed: 05/01/2025]
Abstract
Transmembrane protein 16A (TMEM16A) is a Ca2+-activated Cl- channel that is widely expressed in the digestive system, and numerous compounds have been developed for targeting TMEM16A. This review summarizes the current state of knowledge of physiological and pathological roles of TMEM16A in the digestive system, and discuss the potential therapeutic uses and challenges of TMEM16A modulators, with a focus on their selectivity, potency and molecular mechanisms as well as off-target tissue effects. We propose that TMEM16A exerts physiological and pathological roles in a tissue-specific or disease-specific way, and try to establish the idea that TMEM16A modulators are promising for therapeutic uses in digestive diseases such as secretory diarrhea, gastrointestinal motility disorders, and hepatobiliary and pancreatic diseases, as well as various cancers.
Collapse
Affiliation(s)
- Shen Zhang
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China; Department of Gastroenterology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110031, China
| | - Shuya Luo
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Hong Zhang
- Department of Colorectal Oncology/General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Qinghuan Xiao
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China.
| |
Collapse
|
5
|
Pant S, Tam SW, Long SB. The pentameric chloride channel BEST1 is activated by extracellular GABA. Proc Natl Acad Sci U S A 2025; 122:e2424474122. [PMID: 40249777 PMCID: PMC12037058 DOI: 10.1073/pnas.2424474122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/12/2025] [Indexed: 04/20/2025] Open
Abstract
Bestrophin-1 (BEST1) is a chloride channel expressed in the eye and other tissues of the body. A link between BEST1 and the principal inhibitory neurotransmitter γ-aminobutyric acid (GABA) has been proposed. The most appreciated receptors for extracellular GABA are the GABAB G-protein-coupled receptors and the pentameric GABAA chloride channels, both of which have fundamental roles in the central nervous system. Here, we demonstrate that BEST1 is directly activated by GABA. Through functional studies and atomic-resolution structures of human and chicken BEST1, we identify a GABA binding site on the channel's extracellular side and determine the mechanism by which GABA binding stabilizes opening of the channel's central gate. This same gate, "the neck," is activated by intracellular [Ca2+], indicating that BEST1 is controlled by ligands from both sides of the membrane. The studies demonstrate that BEST1, which shares no structural homology with GABAA receptors, is a GABA-activated chloride channel. The physiological implications of this finding remain to be studied.
Collapse
Affiliation(s)
- Swati Pant
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY10065
- Graduate Program in Biochemistry and Structural Biology, Cell and Developmental Biology, and Molecular Biology, Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY10065
| | - Stephanie W. Tam
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY10065
- Graduate Program in Physiology, Biophysics, and Systems Biology, Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY10065
| | - Stephen B. Long
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY10065
| |
Collapse
|
6
|
Hankins MTK, Parrag M, Garaeva AA, Earp JC, Seeger MA, Stansfeld PJ, Bublitz M. MprF from Pseudomonas aeruginosa is a promiscuous lipid scramblase with broad substrate specificity. SCIENCE ADVANCES 2025; 11:eads9135. [PMID: 40203087 PMCID: PMC11980842 DOI: 10.1126/sciadv.ads9135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 03/03/2025] [Indexed: 04/11/2025]
Abstract
The multiple peptide resistance factor (MprF) is a bifunctional membrane protein found in many bacteria, including Pseudomonas aeruginosa and Staphylococcus aureus. MprF modifies inner leaflet lipid headgroups through aminoacylation and translocates modified lipid to the outer leaflet. This activity provides increased resistance to antimicrobial agents. MprF presents a promising target in multiresistant pathogens, but structural information is limited and both substrate specificity and energization of MprF-mediated lipid transport are poorly understood. Here, we present the cryo-EM structure of MprF from P. aeruginosa (PaMprF) bound to a synthetic nanobody. PaMprF adopts an "open" conformation with a wide, lipid-exposed groove on the periplasmic side that induces a local membrane deformation in molecular dynamics simulations. Using an in vitro liposome transport assay, we demonstrate that PaMprF translocates a wide range of different lipids without an external energy source. This suggests that PaMprF is the first dedicated lipid scramblase to be characterized in bacteria.
Collapse
Affiliation(s)
- Matthew T. K. Hankins
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Matyas Parrag
- School of Life Sciences and Department of Chemistry, University of Warwick, Coventry CV4 7AL, UK
| | - Alisa A. Garaeva
- Institute of Medical Microbiology, University of Zurich, Gloriastrasse 28/30, 8006 Zurich, Switzerland
| | - Jennifer C. Earp
- Institute of Medical Microbiology, University of Zurich, Gloriastrasse 28/30, 8006 Zurich, Switzerland
| | - Markus A. Seeger
- Institute of Medical Microbiology, University of Zurich, Gloriastrasse 28/30, 8006 Zurich, Switzerland
| | - Phillip J. Stansfeld
- School of Life Sciences and Department of Chemistry, University of Warwick, Coventry CV4 7AL, UK
| | - Maike Bublitz
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
- Institute of Translational Medicine, Faculty of Medical Sciences, Private University in the Principality of Liechtenstein (UFL), Dorfstrasse 24, 9495 Triesen, Liechtenstein
| |
Collapse
|
7
|
Lee Y, Demes-Causse E, Yoo J, Jang SY, Jung S, Jaślan J, Hwang GS, Yoo J, De Angeli A, Lee S. Structural basis for malate-driven, pore lipid-regulated activation of the Arabidopsis vacuolar anion channel ALMT9. Nat Commun 2025; 16:1817. [PMID: 39979303 PMCID: PMC11842843 DOI: 10.1038/s41467-025-56940-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 02/04/2025] [Indexed: 02/22/2025] Open
Abstract
In plant cells, ALMTs are key plasma and vacuolar membrane-localized anion channels regulating plant responses to the environment. Vacuolar ALMTs control anion accumulation in plant cells and, in guard cells, they regulate stomata aperture. The activation of vacuolar ALMTs depends on voltage and cytosolic malate, but the underlying molecular mechanisms remain elusive. Here we report the cryo-EM structures of ALMT9 from Arabidopsis thaliana (AtALMT9), a malate-activated vacuolar anion channel, in plugged and unplugged lipid-bound states. In all these states, membrane lipids interact with the ion conduction pathway of AtALMT9. We identify two unplugged states presenting two distinct pore width profiles. Combining structural and functional analysis we identified conserved residues involved in ion conduction and in the pore lipid interaction. Molecular dynamics simulations revealed a peculiar anion conduction mechanism in AtALMT9. We propose a voltage-dependent activation mechanism based on the competition between pore lipids and malate at the cytosolic entrance of the channel.
Collapse
Affiliation(s)
- Yeongmok Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Republic of Korea
| | - Elsa Demes-Causse
- IPSiM, CNRS, INRAE, Institut Agro, Université Montpellier, Montpellier, France
| | - Jaemin Yoo
- Department of Physics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Seo Young Jang
- Integrated Metabolomics Research Group, Metropolitan Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
| | - Seoyeon Jung
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Republic of Korea
| | - Justyna Jaślan
- IPSiM, CNRS, INRAE, Institut Agro, Université Montpellier, Montpellier, France
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Metropolitan Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Jejoong Yoo
- Department of Physics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Alexis De Angeli
- IPSiM, CNRS, INRAE, Institut Agro, Université Montpellier, Montpellier, France
| | - Sangho Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Republic of Korea.
- Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Metabiohealth, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
8
|
Jan LY, Jan YN. Wide-ranging cellular functions of ion channels and lipid scramblases in the structurally related TMC, TMEM16 and TMEM63 families. Nat Struct Mol Biol 2025; 32:222-236. [PMID: 39715905 DOI: 10.1038/s41594-024-01444-x] [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: 01/18/2024] [Accepted: 10/31/2024] [Indexed: 12/25/2024]
Abstract
Calcium (Ca2+)-activated ion channels and lipid scramblases in the transmembrane protein 16 (TMEM16) family are structurally related to mechanosensitive ion channels in the TMEM63 and transmembrane channel-like (TMC) families. Members of this structurally related superfamily share similarities in gating transitions and serve a wide range of physiological functions, which is evident from their disease associations. The TMEM16, TMEM63 and TMC families include members with important functions in the cell membrane and/or intracellular organelles such as the endoplasmic reticulum, membrane contact sites, endosomes and lysosomes. Moreover, some members of the TMEM16 family and the TMC family perform dual functions of ion channel and lipid scramblase, leading to intriguing physiological implications. In addition to their physiological functions such as mediating phosphatidylserine exposure and facilitation of extracellular vesicle generation and cell fusion, scramblases are involved in the entry and replication of enveloped viruses. Comparisons of structurally diverse scramblases may uncover features in the lipid-scrambling mechanisms that are likely shared by scramblases.
Collapse
Affiliation(s)
- Lily Yeh Jan
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA.
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA.
| | - Yuh Nung Jan
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA.
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
9
|
Sebinelli HG, Syska C, Čopič A, Lenoir G. Established and emerging players in phospholipid scrambling: A structural perspective. Biochimie 2024; 227:111-122. [PMID: 39304020 DOI: 10.1016/j.biochi.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
The maintenance of a diverse and non-homogeneous lipid composition in cell membranes is crucial for a multitude of cellular processes. One important example is transbilayer lipid asymmetry, which refers to a difference in lipid composition between the two leaflets of a cellular membrane. Transbilayer asymmetry is especially pronounced at the plasma membrane, where at resting state, negatively-charged phospholipids such as phosphatidylserine (PS) are almost exclusively restricted to the cytosolic leaflet, whereas sphingolipids are mostly found in the exoplasmic leaflet. Transbilayer movement of lipids is inherently slow, and for a fast cellular response, for example during apoptosis, transmembrane proteins termed scramblases facilitate the movement of polar/charged lipid headgroups through the membrane interior. In recent years, an expanding number of proteins from diverse families have been suggested to possess a lipid scramblase activity. Members of TMEM16 and XKR proteins have been implicated in blood clotting and apoptosis, whereas the scrambling activity of ATG9 and TMEM41B/VMP1 proteins contributes to the synthesis of autophagosomal membrane during autophagy. Structural studies, in vitro reconstitution of lipid scrambling, and molecular dynamics simulations have significantly advanced our understanding of the molecular mechanisms of lipid scrambling and helped delineate potential lipid transport pathways through the membrane. A number of examples also suggest that lipid scrambling activity can be combined with another activity, as is the case for TMEM16 proteins, which also function as ion channels, rhodopsin in the photoreceptor membrane, and possibly other G-protein coupled receptors.
Collapse
Affiliation(s)
- Heitor Gobbi Sebinelli
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell, Gif-sur-Yvette, France
| | - Camille Syska
- Centre de Recherche en Biologie Cellulaire de Montpellier (CRBM), Université de Montpellier, CNRS, 34293, Montpellier, Cedex 05, France
| | - Alenka Čopič
- Centre de Recherche en Biologie Cellulaire de Montpellier (CRBM), Université de Montpellier, CNRS, 34293, Montpellier, Cedex 05, France
| | - Guillaume Lenoir
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell, Gif-sur-Yvette, France.
| |
Collapse
|
10
|
Pant S, Tam SW, Long SB. The pentameric chloride channel BEST1 is activated by extracellular GABA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624909. [PMID: 39605608 PMCID: PMC11601618 DOI: 10.1101/2024.11.22.624909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Bestrophin 1 (BEST1) is chloride channel expressed in the eye, central nervous system (CNS), and other tissues in the body. A link between BEST1 and the principal inhibitory neurotransmitter γ-aminobutyric acid (GABA) has been proposed. The most appreciated receptors for extracellular GABA are the GABAB G-protein coupled receptors and the pentameric GABAA chloride channels, both of which have fundamental roles in the CNS. Here, we demonstrate that BEST1 is directly activated by GABA. Through functional studies and atomic-resolution structures of human and chicken BEST1, we identify a GABA binding site on the channel's extracellular side and determine the mechanism by which GABA binding induces opening of the channel's central gate. This same gate is activated by intracellular [Ca2+], indicating that BEST1 is controlled by ligands from both sides of the membrane. The studies demonstrate that BEST1, which shares no structural homology with GABAA, is a GABA-activated chloride channel. The physiological implications of this finding remain to be studied.
Collapse
Affiliation(s)
- Swati Pant
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
- Graduate Program in Biochemistry and Structural Biology, Cell and Developmental Biology, and Molecular Biology, Weill Cornell Medicine Graduate School of Medical Sciences, New York, USA
| | - Stephanie W. Tam
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
- Graduate Program in Physiology, Biophysics, and Systems Biology, Weill Cornell Medicine Graduate School of Medical Sciences, New York, USA
| | - Stephen B. Long
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
11
|
Lowry AJ, Liang P, Song M, Wan Y, Pei ZM, Yang H, Zhang Y. TMEM16 and OSCA/TMEM63 proteins share a conserved potential to permeate ions and phospholipids. eLife 2024; 13:RP96957. [PMID: 39495104 PMCID: PMC11534332 DOI: 10.7554/elife.96957] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024] Open
Abstract
The calcium-activated TMEM16 proteins and the mechanosensitive/osmolarity-activated OSCA/TMEM63 proteins belong to the Transmembrane Channel/Scramblase (TCS) superfamily. Within the superfamily, OSCA/TMEM63 proteins, as well as TMEM16A and TMEM16B, are thought to function solely as ion channels. However, most TMEM16 members, including TMEM16F, maintain an additional function as scramblases, rapidly exchanging phospholipids between leaflets of the membrane. Although recent studies have advanced our understanding of TCS structure-function relationships, the molecular determinants of TCS ion and lipid permeation remain unclear. Here, we show that single mutations along the transmembrane helix (TM) 4/6 interface allow non-scrambling TCS members to permeate phospholipids. In particular, this study highlights the key role of TM 4 in controlling TCS ion and lipid permeation and offers novel insights into the evolution of the TCS superfamily, suggesting that, like TMEM16s, the OSCA/TMEM63 family maintains a conserved potential to permeate ions and phospholipids.
Collapse
Affiliation(s)
- Augustus J Lowry
- Department of Biochemistry, Duke University School of MedicineDurhamUnited States
| | - Pengfei Liang
- Department of Biochemistry, Duke University School of MedicineDurhamUnited States
| | - Mo Song
- Institute of Molecular Physiology, Shenzhen Bay LaboratoryGuangdongChina
| | - Yuichun Wan
- Department of Biochemistry, Duke University School of MedicineDurhamUnited States
| | - Zhen-Ming Pei
- Department of Biology, Duke UniversityDurhamUnited States
| | - Huanghe Yang
- Department of Biochemistry, Duke University School of MedicineDurhamUnited States
- Department of Neurobiology, Duke University School of MedicineDurhamUnited States
| | - Yang Zhang
- Department of Biochemistry, Duke University School of MedicineDurhamUnited States
- Institute of Molecular Physiology, Shenzhen Bay LaboratoryGuangdongChina
| |
Collapse
|
12
|
Jagtap P, Meena VK, Sambhare S, Basu A, Abraham P, Cherian S. Exploring Niclosamide as a Multi-target Drug Against SARS-CoV-2: Molecular Dynamics Simulation Studies on Host and Viral Proteins. Mol Biotechnol 2024:10.1007/s12033-024-01296-2. [PMID: 39373955 DOI: 10.1007/s12033-024-01296-2] [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: 08/29/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024]
Abstract
Niclosamide has emerged as a promising repurposed drug against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In vitro studies suggested that niclosamide inhibits the host transmembrane protein 16F (hTMEM16F), crucial for lipid scramblase activity, which consequently reduces syncytia formation that aids viral spread. Based on other in vitro reports, niclosamide may also target viral proteases such as papain-like protease (PLpro) and main protease (Mpro), essential for viral replication and maturation. However, the precise interactions by which niclosamide interacts with these multiple targets remain largely unclear. Docking and molecular dynamics (MD) simulation studies were undertaken based on a homology model of the hTMEM16F and available crystal structures of SARS-CoV-2 PLpro and Mpro. Niclosamide was observed to bind stably throughout a 400 ns MD simulation at the extracellular exit gate of the hTMEM16F tunnel, forming crucial interactions with residues spanning the TM1-TM2 loop (Gln350), TM3 (Phe481), and TM5-TM6 loop (Lys573, Glu594, and Asp596). Among the SARS-CoV-2 proteases, niclosamide was found to interact effectively with conserved active site residues of PLpro (Tyr268), exhibiting better stability in comparison to the control inhibitor, GRL0617. In conclusion, our in silico analyses support niclosamide as a multi-targeted drug inhibiting viral and host proteins involved in SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Prachi Jagtap
- Bioinformatics & Data Management Group, ICMR National Institute of Virology, 20A Dr. Ambedkar Road, Pune, Maharashtra, 411 001, India
| | - Virendra Kumar Meena
- ICMR National Institute of Virology, 20A Dr. Ambedkar Road, Pune, Maharashtra, 411 001, India
| | - Susmit Sambhare
- ICMR National Institute of Virology, 20A Dr. Ambedkar Road, Pune, Maharashtra, 411 001, India
| | - Atanu Basu
- ICMR National Institute of Virology, 20A Dr. Ambedkar Road, Pune, Maharashtra, 411 001, India
| | - Priya Abraham
- Christian Medical College, Vellore, Tamil Nadu, India
| | - Sarah Cherian
- Bioinformatics & Data Management Group, ICMR National Institute of Virology, 20A Dr. Ambedkar Road, Pune, Maharashtra, 411 001, India.
| |
Collapse
|
13
|
Guo X, Geng R, Li C, Ma Z, Chen Y, Liu Y, Li S, Kang X, Guo S. Structural and theoretical basis for drug development targeting TMEM16A: Inhibition mechanism of tracheloside analogs. Int J Biol Macromol 2024; 277:134057. [PMID: 39038568 DOI: 10.1016/j.ijbiomac.2024.134057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/12/2024] [Accepted: 07/19/2024] [Indexed: 07/24/2024]
Abstract
Ion channels play a crucial role in the electrophysiological activities of organisms. The calcium-activated chloride channel TMEM16A is involved in various physiological processes. Therefore, inhibitors of TMEM16A are used to treat diseases caused by TMEM16A dysfunction. However, the unclear inhibition mechanism hinders the progress of drug development. Based on our previous study, we found that the molecular structures of TMEM16A inhibitors tracheloside, matairesinoside and arctigenin are similar. In this study, we conducted a structure-based virtual screening of tracheloside analogs from the PubChem database. The six tracheloside analogs with the highest affinity to TMEM16A were selected, and their inhibitory effects were detected by fluorescence and electrophysiological experiments. Subsequently, the interaction between the tracheloside analogs and TMEM16A was investigated through molecular docking and site-directed mutagenesis. Based on the above results, the mechanism of inhibition of TMEM16A gated conformation by tracheloside analogs was proposed. These findings provide a structural and theoretical basis for drug development targeting TMEM16A.
Collapse
Affiliation(s)
- Xiaomeng Guo
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Ruili Geng
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Chao Li
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Zhouye Ma
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Yue Chen
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Yinuo Liu
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Shaochun Li
- School of Basic Medical Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Xianjiang Kang
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China; Collaborative Innovation Center for Baiyangdian Basin Ecological Protection and Beijing-Tianjin-Hebei Sustainable Development, Hebei University, Baoding 071002, Hebei, China; Institute of Life Sciences and Green Development, Hebei University, Baoding 071002, Hebei, China; Hebei Basic Science Center for Biotic Interaction, Hebei University, Baoding 071002, Hebei, China.
| | - Shuai Guo
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China; Collaborative Innovation Center for Baiyangdian Basin Ecological Protection and Beijing-Tianjin-Hebei Sustainable Development, Hebei University, Baoding 071002, Hebei, China; Institute of Life Sciences and Green Development, Hebei University, Baoding 071002, Hebei, China; Hebei Basic Science Center for Biotic Interaction, Hebei University, Baoding 071002, Hebei, China.
| |
Collapse
|
14
|
Feng Z, Alvarenga OE, Accardi A. Structural basis of closed groove scrambling by a TMEM16 protein. Nat Struct Mol Biol 2024; 31:1468-1481. [PMID: 38684930 DOI: 10.1038/s41594-024-01284-9] [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: 08/11/2023] [Accepted: 03/21/2024] [Indexed: 05/02/2024]
Abstract
Activation of Ca2+-dependent TMEM16 scramblases induces phosphatidylserine externalization, a key step in multiple signaling processes. Current models suggest that the TMEM16s scramble lipids by deforming the membrane near a hydrophilic groove and that Ca2+ dependence arises from the different association of lipids with an open or closed groove. However, the molecular rearrangements underlying groove opening and how lipids reorganize outside the closed groove remain unknown. Here we directly visualize how lipids associate at the closed groove of Ca2+-bound fungal nhTMEM16 in nanodiscs using cryo-EM. Functional experiments pinpoint lipid-protein interaction sites critical for closed groove scrambling. Structural and functional analyses suggest groove opening entails the sequential appearance of two π-helical turns in the groove-lining TM6 helix and identify critical rearrangements. Finally, we show that the choice of scaffold protein and lipids affects the conformations of nhTMEM16 and their distribution, highlighting a key role of these factors in cryo-EM structure determination.
Collapse
Affiliation(s)
- Zhang Feng
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY, USA
| | - Omar E Alvarenga
- Physiology, Biophysics and Systems Biology Graduate Program, Weill Cornell Medical College, New York, NY, USA
| | - Alessio Accardi
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY, USA.
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA.
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
15
|
Lowry AJ, Liang P, Song M, Serena Wan YC, Pei ZM, Yang H, Zhang Y. TMEM16 and OSCA/TMEM63 proteins share a conserved potential to permeate ions and phospholipids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.04.578431. [PMID: 38370744 PMCID: PMC10871192 DOI: 10.1101/2024.02.04.578431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
The calcium-activated TMEM16 proteins and the mechanosensitive/osmolarity-activated OSCA/TMEM63 proteins belong to the Transmembrane Channel/Scramblase (TCS) superfamily. Within the superfamily, OSCA/TMEM63 proteins, as well as TMEM16A and TMEM16B, are thought to function solely as ion channels. However, most TMEM16 members, including TMEM16F, maintain an additional function as scramblases, rapidly exchanging phospholipids between leaflets of the membrane. Although recent studies have advanced our understanding of TCS structure-function relationships, the molecular determinants of TCS ion and lipid permeation remain unclear. Here we show that single mutations along the transmembrane helix (TM) 4/6 interface allow non-scrambling TCS members to permeate phospholipids. In particular, this study highlights the key role of TM 4 in controlling TCS ion and lipid permeation and offers novel insights into the evolution of the TCS superfamily, suggesting that, like TMEM16s, the OSCA/TMEM63 family maintains a conserved potential to permeate ions and phospholipids.
Collapse
Affiliation(s)
- Augustus J Lowry
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA
| | - Pengfei Liang
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mo Song
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Guangdong 518106, China
| | - Y C Serena Wan
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA
| | - Zhen-Ming Pei
- Department of Biology, Duke University, Durham, NC 27710, USA
| | - Huanghe Yang
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Yang Zhang
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Guangdong 518106, China
| |
Collapse
|
16
|
Niu H, Maruoka M, Noguchi Y, Kosako H, Suzuki J. Phospholipid scrambling induced by an ion channel/metabolite transporter complex. Nat Commun 2024; 15:7566. [PMID: 39217145 PMCID: PMC11366033 DOI: 10.1038/s41467-024-51939-w] [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: 12/15/2023] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
Cells establish the asymmetrical distribution of phospholipids and alter their distribution by phospholipid scrambling (PLS) to adapt to environmental changes. Here, we demonstrate that a protein complex, consisting of the ion channel Tmem63b and the thiamine transporter Slc19a2, induces PLS upon calcium (Ca2+) stimulation. Through revival screening using a CRISPR sgRNA library on high PLS cells, we identify Tmem63b as a PLS-inducing factor. Ca2+ stimulation-mediated PLS is suppressed by deletion of Tmem63b, while human disease-related Tmem63b mutants induce constitutive PLS. To search for a molecular link between Ca2+ stimulation and PLS, we perform revival screening on Tmem63b-overexpressing cells, and identify Slc19a2 and the Ca2+-activated K+ channel Kcnn4 as PLS-regulating factors. Deletion of either of these genes decreases PLS activity. Biochemical screening indicates that Tmem63b and Slc19a2 form a heterodimer. These results demonstrate that a Tmem63b/Slc19a2 heterodimer induces PLS upon Ca2+ stimulation, along with Kcnn4 activation.
Collapse
Affiliation(s)
- Han Niu
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto, Japan
- Graduate School of Biostudies, Kyoto University, Konoe-cho, Yoshida, Sakyoku, Kyoto, Japan
| | - Masahiro Maruoka
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto, Japan
- Center for Integrated Biosystems, Institute for Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yuki Noguchi
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto, Japan
| | - Hidetaka Kosako
- Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Jun Suzuki
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto, Japan.
- Graduate School of Biostudies, Kyoto University, Konoe-cho, Yoshida, Sakyoku, Kyoto, Japan.
- Center for Integrated Biosystems, Institute for Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan.
| |
Collapse
|
17
|
Ebrahim S, Ballesteros A, Zheng WS, Mukherjee S, Hu G, Weng WH, Montgomery JS, Agyemang Y, Cui R, Sun W, Krystofiak E, Foster MP, Sotomayor M, Kachar B. Transmembrane channel-like 4 and 5 proteins at microvillar tips are potential ion channels and lipid scramblases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.22.609173. [PMID: 39229161 PMCID: PMC11370596 DOI: 10.1101/2024.08.22.609173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Microvilli-membrane bound actin protrusions on the surface of epithelial cells-are sites of critical processes including absorption, secretion, and adhesion. Increasing evidence suggests microvilli are mechanosensitive, but underlying molecules and mechanisms remain unknown. Here, we localize transmembrane channel-like proteins 4 and 5 (TMC4 and 5) and calcium and integrin binding protein 3 (CIB3) to microvillar tips in intestinal epithelial cells, near glycocalyx insertion sites. We find that TMC5 colocalizes with CIB3 in cultured cells and that a TMC5 fragment forms a complex with CIB3 in vitro. Homology and AlphaFold2 models reveal a putative ion permeation pathway in TMC4 and 5, and molecular dynamics simulations predict both proteins can conduct ions and perform lipid scrambling. These findings raise the possibility that TMC4 and 5 interact with CIB3 at microvillar tips to form a mechanosensitive complex, akin to TMC1 and 2, and CIB2 and 3, within the mechanotransduction channel complex at the tips of inner ear stereocilia.
Collapse
Affiliation(s)
- Seham Ebrahim
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA
| | - Angela Ballesteros
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
- Section on Sensory Physiology and Biophysics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - W. Sharon Zheng
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA
| | - Shounak Mukherjee
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Gaizun Hu
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA
| | - Wei-Hsiang Weng
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Jonathan S. Montgomery
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210, USA
| | - Yaw Agyemang
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Runjia Cui
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Willy Sun
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Evan Krystofiak
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark P. Foster
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210, USA
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210, USA
| | - Bechara Kachar
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Chakraborty S, Feng Z, Lee S, Alvarenga OE, Panda A, Bruni R, Khelashvili G, Gupta K, Accardi A. Structure and function of the human apoptotic scramblase Xkr4. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.07.607004. [PMID: 39149361 PMCID: PMC11326236 DOI: 10.1101/2024.08.07.607004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Phosphatidylserine externalization on the surface of dying cells is a key signal for their recognition and clearance by macrophages and is mediated by members of the X-Kell related (Xkr) protein family. Defective Xkr-mediated scrambling impairs clearance, leading to inflammation. It was proposed that activation of the Xkr4 apoptotic scramblase requires caspase cleavage, followed by dimerization and ligand binding. Here, using a combination of biochemical approaches we show that purified monomeric, full-length human Xkr4 (hXkr4) scrambles lipids. CryoEM imaging shows that hXkr4 adopts a novel conformation, where three conserved acidic residues create an electronegative surface embedded in the membrane. Molecular dynamics simulations show this conformation induces membrane thinning, which could promote scrambling. Thinning is ablated or reduced in conditions where scrambling is abolished or reduced. Our work provides insights into the molecular mechanisms of hXkr4 scrambling and suggests the ability to thin membranes might be a general property of active scramblases.
Collapse
Affiliation(s)
| | - Zhang Feng
- Department of Anesthesiology, Weill Cornell Medical College
| | - Sangyun Lee
- Department of Anesthesiology, Weill Cornell Medical College
| | - Omar E. Alvarenga
- Physiology, Biophysics and Systems Biology Graduate Program, Weill Cornell Medical College
| | - Aniruddha Panda
- Nanobiology Institute, Yale University, West Haven, Connecticut 06516, United States
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Renato Bruni
- Center on Membrane Protein Production and Analysis (COMPPÅ), New York Structural Biology Center, New York, NY 10027, USA
| | | | - Kallol Gupta
- Nanobiology Institute, Yale University, West Haven, Connecticut 06516, United States
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Alessio Accardi
- Department of Anesthesiology, Weill Cornell Medical College
- Department of Physiology and Biophysics, Weill Cornell Medical College
- Department of Biochemistry, Weill Cornell Medical College
| |
Collapse
|
19
|
Lipids act as structural components of the pore of an ion-channel family. Nature 2024:10.1038/d41586-024-02477-4. [PMID: 39075316 DOI: 10.1038/d41586-024-02477-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
|
20
|
Feng Z, Di Zanni E, Alvarenga O, Chakraborty S, Rychlik N, Accardi A. In or out of the groove? Mechanisms of lipid scrambling by TMEM16 proteins. Cell Calcium 2024; 121:102896. [PMID: 38749289 PMCID: PMC11178363 DOI: 10.1016/j.ceca.2024.102896] [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] [Revised: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 06/13/2024]
Abstract
Phospholipid scramblases mediate the rapid movement of lipids between membrane leaflets, a key step in establishing and maintaining membrane homeostasis of the membranes of all eukaryotic cells and their organelles. Thus, impairment of lipid scrambling can lead to a variety of pathologies. How scramblases catalyzed the transbilayer movement of lipids remains poorly understood. Despite the availability of direct structural information on three unrelated families of scramblases, the TMEM16s, the Xkrs, and ATG-9, a unifying mechanism has failed to emerge thus far. Among these, the most extensively studied and best understood are the Ca2+ activated TMEM16s, which comprise ion channels and/or scramblases. Early work supported the view that these proteins provided a hydrophilic, membrane-exposed groove through which the lipid headgroups could permeate. However, structural, and functional experiments have since challenged this mechanism, leading to the proposal that the TMEM16s distort and thin the membrane near the groove to facilitate lipid scrambling. Here, we review our understanding of the structural and mechanistic underpinnings of lipid scrambling by the TMEM16s and discuss how the different proposals account for the various experimental observations.
Collapse
Affiliation(s)
- Zhang Feng
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States
| | - Eleonora Di Zanni
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States
| | - Omar Alvarenga
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States
| | - Sayan Chakraborty
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States
| | - Nicole Rychlik
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States; Institute of Physiology I, University of Münster, Robert-Koch-Str. 27a, D-48149 Münster, Germany
| | - Alessio Accardi
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States; Department of Biochemistry, Weill Cornell Medicine, New York, NY, United States.
| |
Collapse
|
21
|
Arreola J, López-Romero AE, Huerta M, Guzmán-Hernández ML, Pérez-Cornejo P. Insights into the function and regulation of the calcium-activated chloride channel TMEM16A. Cell Calcium 2024; 121:102891. [PMID: 38772195 DOI: 10.1016/j.ceca.2024.102891] [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: 02/27/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/23/2024]
Abstract
The TMEM16A channel, a member of the TMEM16 protein family comprising chloride (Cl-) channels and lipid scramblases, is activated by the free intracellular Ca2+ increments produced by inositol 1,4,5-trisphosphate (IP3)-induced Ca2+ release after GqPCRs or Ca2+ entry through cationic channels. It is a ubiquitous transmembrane protein that participates in multiple physiological functions essential to mammals' lives. TMEM16A structure contains two identical 10-segment monomers joined at their transmembrane segment 10. Each monomer harbours one independent hourglass-shaped pore gated by Ca2+ ligation to an orthosteric site adjacent to the pore and controlled by two gates. The orthosteric site is created by assembling negatively charged glutamate side chains near the pore´s cytosolic end. When empty, this site generates an electrostatic barrier that controls channel rectification. In addition, an isoleucine-triad forms a hydrophobic gate at the boundary of the cytosolic vestibule and the inner side of the neck. When the cytosolic Ca2+ rises, one or two Ca2+ ions bind to the orthosteric site in a voltage (V)-dependent manner, thus neutralising the electrostatic barrier and triggering an allosteric gating mechanism propagating via transmembrane segment 6 to the hydrophobic gate. These coordinated events lead to pore opening, allowing the Cl- flux to ensure the physiological response. The Ca2+-dependent function of TMEM16A is highly regulated. Anions with higher permeability than Cl- facilitate V dependence by increasing the Ca2+ sensitivity, intracellular protons can replace Ca2+ and induce channel opening, and phosphatidylinositol 4,5-bisphosphate bound to four cytosolic sites likely maintains Ca2+ sensitivity. Additional regulation is afforded by cytosolic proteins, most likely by phosphorylation and protein-protein interaction mechanisms.
Collapse
Affiliation(s)
- Jorge Arreola
- Jorge Arreola, Physics Institute of Universidad Autónoma de San Luis Potosí. Av. Parque Chapultepec 1570, Privadas del Pedregal, 78295 San Luis Potosí, SLP., Mexico.
| | - Ana Elena López-Romero
- Jorge Arreola, Physics Institute of Universidad Autónoma de San Luis Potosí. Av. Parque Chapultepec 1570, Privadas del Pedregal, 78295 San Luis Potosí, SLP., Mexico
| | - Miriam Huerta
- Jorge Arreola, Physics Institute of Universidad Autónoma de San Luis Potosí. Av. Parque Chapultepec 1570, Privadas del Pedregal, 78295 San Luis Potosí, SLP., Mexico
| | - María Luisa Guzmán-Hernández
- Catedrática CONAHCYT, Department of Physiology and Biophysics, School of Medicine, Universidad Autónoma de San Luis Potosí. Ave. V. Carranza 2905, Los Filtros, San Luis Potosí, SLP 78210, Mexico
| | - Patricia Pérez-Cornejo
- Department of Physiology and Biophysics, School of Medicine, Universidad Autónoma de San Luis Potosí. Ave. V. Carranza 2905, Los Filtros, San Luis Potosí, SLP 78210, Mexico
| |
Collapse
|
22
|
Yang F, Begemann A, Reichhart N, Haeckel A, Steindl K, Schellenberger E, Sturm RF, Barth M, Bassani S, Boonsawat P, Courtin T, Delobel B, Gunning B, Hardies K, Jennesson M, Legoff L, Linnankivi T, Prouteau C, Smal N, Spodenkiewicz M, Toelle SP, Van Gassen K, Van Paesschen W, Verbeek N, Ziegler A, Zweier M, Horn AHC, Sticht H, Lerche H, Weckhuysen S, Strauß O, Rauch A. Missense variants in ANO4 cause sporadic encephalopathic or familial epilepsy with evidence for a dominant-negative effect. Am J Hum Genet 2024; 111:1184-1205. [PMID: 38744284 PMCID: PMC11179416 DOI: 10.1016/j.ajhg.2024.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 05/16/2024] Open
Abstract
Anoctamins are a family of Ca2+-activated proteins that may act as ion channels and/or phospholipid scramblases with limited understanding of function and disease association. Here, we identified five de novo and two inherited missense variants in ANO4 (alias TMEM16D) as a cause of fever-sensitive developmental and epileptic or epileptic encephalopathy (DEE/EE) and generalized epilepsy with febrile seizures plus (GEFS+) or temporal lobe epilepsy. In silico modeling of the ANO4 structure predicted that all identified variants lead to destabilization of the ANO4 structure. Four variants are localized close to the Ca2+ binding sites of ANO4, suggesting impaired protein function. Variant mapping to the protein topology suggests a preliminary genotype-phenotype correlation. Moreover, the observation of a heterozygous ANO4 deletion in a healthy individual suggests a dysfunctional protein as disease mechanism rather than haploinsufficiency. To test this hypothesis, we examined mutant ANO4 functional properties in a heterologous expression system by patch-clamp recordings, immunocytochemistry, and surface expression of annexin A5 as a measure of phosphatidylserine scramblase activity. All ANO4 variants showed severe loss of ion channel function and DEE/EE associated variants presented mild loss of surface expression due to impaired plasma membrane trafficking. Increased levels of Ca2+-independent annexin A5 at the cell surface suggested an increased apoptosis rate in DEE-mutant expressing cells, but no changes in Ca2+-dependent scramblase activity were observed. Co-transfection with ANO4 wild-type suggested a dominant-negative effect. In summary, we expand the genetic base for both encephalopathic sporadic and inherited fever-sensitive epilepsies and link germline variants in ANO4 to a hereditary disease.
Collapse
Affiliation(s)
- Fang Yang
- Experimental Ophthalmology, Department of Ophthalmology, Charité - Universitätsmedizin Berlin, a Corporate Member of Freie Universität, Humboldt-University, the Berlin Institute of Health, Berlin, Germany
| | - Anais Begemann
- Institute of Medical Genetics, University of Zurich, Schlieren-Zurich, Switzerland
| | - Nadine Reichhart
- Experimental Ophthalmology, Department of Ophthalmology, Charité - Universitätsmedizin Berlin, a Corporate Member of Freie Universität, Humboldt-University, the Berlin Institute of Health, Berlin, Germany
| | - Akvile Haeckel
- Institute for Radiology and Children's Radiology, Charité-Universitätsmedizin Berlin, a Corporate Member of Freie Universität, Humboldt-University, the Berlin Institute of Health, Berlin, Germany
| | - Katharina Steindl
- Institute of Medical Genetics, University of Zurich, Schlieren-Zurich, Switzerland
| | - Eyk Schellenberger
- Institute for Radiology and Children's Radiology, Charité-Universitätsmedizin Berlin, a Corporate Member of Freie Universität, Humboldt-University, the Berlin Institute of Health, Berlin, Germany
| | - Ronja Fini Sturm
- Experimental Ophthalmology, Department of Ophthalmology, Charité - Universitätsmedizin Berlin, a Corporate Member of Freie Universität, Humboldt-University, the Berlin Institute of Health, Berlin, Germany
| | - Magalie Barth
- University Hospital of Angers, Department of Genetics, Angers, France
| | - Sissy Bassani
- Institute of Medical Genetics, University of Zurich, Schlieren-Zurich, Switzerland
| | - Paranchai Boonsawat
- Institute of Medical Genetics, University of Zurich, Schlieren-Zurich, Switzerland
| | - Thomas Courtin
- Sorbonne Université, INSERM, CNRS, Institut du Cerveau - Paris Brain Institute - ICM, 75013 Paris, France; Hôpital Pitié-Salpêtrière, DMU BioGe'M, AP-HP, 75013 Paris, France
| | - Bruno Delobel
- Service de Cytogénétique, GH de l'Institut Catholique de Lille, Hopital Saint Vincent de Paul, Lille, France
| | | | - Katia Hardies
- Applied & Translational Neurogenomics Group, VIB Center for Molecular Neurology, VIB, University of Antwerp, 2610 Antwerp, Belgium
| | | | - Louis Legoff
- University Hospital of Angers, Department of Genetics, Angers, France
| | - Tarja Linnankivi
- Epilepsia Helsinki, University of Helsinki and Helsinki University Hospital, 00029 HUS Helsinki, Finland; Department of Pediatric Neurology and Pediatric Research Center, New Children's Hospital, Helsinki University Hospital and University of Helsinki, 00029 HUS Helsinki, Finland
| | - Clément Prouteau
- University Hospital of Angers, Department of Genetics, Angers, France
| | - Noor Smal
- Applied & Translational Neurogenomics Group, VIB Center for Molecular Neurology, VIB, University of Antwerp, 2610 Antwerp, Belgium
| | - Marta Spodenkiewicz
- Department of Genetics, La Réunion University Hospital, Saint-Pierre, France
| | - Sandra P Toelle
- Department of Pediatric Neurology, Children's University Hospital Zurich, Zurich, Switzerland
| | - Koen Van Gassen
- University Medical Center Utrecht, Department of Genetics, Utrecht, the Netherlands
| | - Wim Van Paesschen
- Laboratory for Epilepsy Research, KU Leuven, and Neurology Department, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Nienke Verbeek
- University Medical Center Utrecht, Department of Genetics, Utrecht, the Netherlands
| | - Alban Ziegler
- University Hospital of Angers, Department of Genetics, Angers, France
| | - Markus Zweier
- Institute of Medical Genetics, University of Zurich, Schlieren-Zurich, Switzerland
| | - Anselm H C Horn
- Institute of Medical Genetics, University of Zurich, Schlieren-Zurich, Switzerland; Division of Bioinformatics, Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Heinrich Sticht
- Division of Bioinformatics, Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Sarah Weckhuysen
- Applied & Translational Neurogenomics Group, VIB Center for Molecular Neurology, VIB, University of Antwerp, 2610 Antwerp, Belgium; Department of Neurology, Antwerp University Hospital, Antwerp, Belgium; Translational Neurosciences, Faculty of Medicine and Health Science, University of Antwerp, 2610 Antwerp, Belgium
| | - Olaf Strauß
- Experimental Ophthalmology, Department of Ophthalmology, Charité - Universitätsmedizin Berlin, a Corporate Member of Freie Universität, Humboldt-University, the Berlin Institute of Health, Berlin, Germany
| | - Anita Rauch
- Institute of Medical Genetics, University of Zurich, Schlieren-Zurich, Switzerland; Children's University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
23
|
Shaydakov ME, Diaz JA, Eklöf B, Lurie F. Venous valve hypoxia as a possible mechanism of deep vein thrombosis: a scoping review. INT ANGIOL 2024; 43:309-322. [PMID: 38864688 DOI: 10.23736/s0392-9590.24.05170-8] [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: 06/13/2024]
Abstract
INTRODUCTION The pathogenesis of deep vein thrombosis (DVT) has been explained by an interplay between a changed blood composition, vein wall alteration, and blood flow abnormalities. A comprehensive investigation of these components of DVT pathogenesis has substantially promoted our understanding of thrombogenesis in the venous system. Meanwhile, the process of DVT initiation remains obscure. This systematic review aims to collect, analyze, and synthesize the published evidence to propose hypoxia as a possible trigger of DVT. EVIDENCE ACQUISITION An exhaustive literature search was conducted across multiple electronic databased including PubMed, EMBASE, Scopus, and Web of Science to identify studies pertinent to the research hypothesis. The search was aimed at exploring the connection between hypoxia, reoxygenation, and the initiation of deep vein thrombosis (DVT). The following key words were used: "deep vein thrombosis," "venous thrombosis," "venous thromboembolism," "hypoxia," "reoxygenation," "venous valve," and "venous endothelium." Reviews, case reports, editorials, and letters were excluded. EVIDENCE SYNTHESIS Based on the systematic search outcome, 156 original papers relevant to the issue were selected for detailed review. These studies encompassed a range of experimental and observational clinical research, focusing on various aspects of DVT, including the anatomical, physiological, and cellular bases of the disease. A number of studies suggested limitations in the traditional understanding of Virchow's triad as an acceptable explanation for DVT initiation. Emerging evidence points to more complex interactions and additional factors that may be critical in the early stages of thrombogenesis. The role of venous valves has been recognized but remains underappreciated, with several studies indicating that these sites may act as primary loci for thrombus formation. A collection of studies describes the effects of hypoxia on venous endothelial cells at the cellular and molecular levels. Hypoxia influences several pathways that regulate endothelial cell permeability, inflammatory response, and procoagulation activity, underpinning the endothelial dysfunction noted in DVT. CONCLUSIONS Hypoxia of the venous valve may serve as an independent hypothesis to outline the DVT triggering process. Further research projects in this field may discover new molecular pathways responsible for the disease and suggest new therapeutic targets.
Collapse
Affiliation(s)
- Maxim E Shaydakov
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburg, PA, USA -
| | - Jose A Diaz
- Division of Surgical Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Fedor Lurie
- Jobst Vascular Institute, ProMedica Health System, Toledo, OH, USA
| |
Collapse
|
24
|
Moran O, Tammaro P. Identification of determinants of lipid and ion transport in TMEM16/anoctamin proteins through a Bayesian statistical analysis. Biophys Chem 2024; 308:107194. [PMID: 38401241 DOI: 10.1016/j.bpc.2024.107194] [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: 12/16/2023] [Revised: 01/17/2024] [Accepted: 01/28/2024] [Indexed: 02/26/2024]
Abstract
The TMEM16/Anoctamin protein family (TMEM16x) is composed of members with different functions; some members form Ca2+-activated chloride channels, while others are lipid scramblases or combine the two functions. TMEM16x proteins are typically activated in response to agonist-induced rises of intracellular Ca2+; thus, they couple Ca2+-signalling with cell electrical activity or plasmalemmal lipid homeostasis. The structural domains underlying these functions are not fully defined. We used a Naïve Bayes classifier to gain insights into these domains. The method enabled identification of regions involved in either ion or lipid transport, and suggested domains for possible pharmacological exploitation. The method allowed the prediction of the transport property of any given TMEM16x. We envisage this strategy could be exploited to illuminate the structure-function relationship of any protein family composed of members playing different molecular roles.
Collapse
Affiliation(s)
- Oscar Moran
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche (CNR), Via De Marini 6, 16149 Genova, Italy
| | - Paolo Tammaro
- Department Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.
| |
Collapse
|
25
|
Li X, Wang Y, Zhang L, Yao S, Liu Q, Jin H, Tuo B. The role of anoctamin 1 in liver disease. J Cell Mol Med 2024; 28:e18320. [PMID: 38685684 PMCID: PMC11058335 DOI: 10.1111/jcmm.18320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/21/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024] Open
Abstract
Liver diseases include all types of viral hepatitis, alcoholic liver disease (ALD), nonalcoholic fatty liver disease (NAFLD), cirrhosis, liver failure (LF) and hepatocellular carcinoma (HCC). Liver disease is now one of the leading causes of disease and death worldwide, which compels us to better understand the mechanisms involved in the development of liver diseases. Anoctamin 1 (ANO1), a calcium-activated chloride channel (CaCC), plays an important role in epithelial cell secretion, proliferation and migration. ANO1 plays a key role in transcriptional regulation as well as in many signalling pathways. It is involved in the genesis, development, progression and/or metastasis of several tumours and other diseases including liver diseases. This paper reviews the role and molecular mechanisms of ANO1 in the development of various liver diseases, aiming to provide a reference for further research on the role of ANO1 in liver diseases and to contribute to the improvement of therapeutic strategies for liver diseases by regulating ANO1.
Collapse
Affiliation(s)
- Xin Li
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Yongfeng Wang
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Li Zhang
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Shun Yao
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Qian Liu
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Hai Jin
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine of Zunyi Medical UniversityZunyiChina
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine of Zunyi Medical UniversityZunyiChina
| |
Collapse
|
26
|
Li D, Rocha-Roa C, Schilling MA, Reinisch KM, Vanni S. Lipid scrambling is a general feature of protein insertases. Proc Natl Acad Sci U S A 2024; 121:e2319476121. [PMID: 38621120 PMCID: PMC11047089 DOI: 10.1073/pnas.2319476121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/13/2024] [Indexed: 04/17/2024] Open
Abstract
Glycerophospholipids are synthesized primarily in the cytosolic leaflet of the endoplasmic reticulum (ER) membrane and must be equilibrated between bilayer leaflets to allow the ER and membranes derived from it to grow. Lipid equilibration is facilitated by integral membrane proteins called "scramblases." These proteins feature a hydrophilic groove allowing the polar heads of lipids to traverse the hydrophobic membrane interior, similar to a credit card moving through a reader. Nevertheless, despite their fundamental role in membrane expansion and dynamics, the identity of most scramblases has remained elusive. Here, combining biochemical reconstitution and molecular dynamics simulations, we show that lipid scrambling is a general feature of protein insertases, integral membrane proteins which insert polypeptide chains into membranes of the ER and organelles disconnected from vesicle trafficking. Our data indicate that lipid scrambling occurs in the same hydrophilic channel through which protein insertion takes place and that scrambling is abolished in the presence of nascent polypeptide chains. We propose that protein insertases could have a so-far-overlooked role in membrane dynamics as scramblases.
Collapse
Affiliation(s)
- Dazhi Li
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT06520
| | - Cristian Rocha-Roa
- Department of Biology, University of Fribourg, FribourgCH-1700, Switzerland
| | - Matthew A. Schilling
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT06520
| | - Karin M. Reinisch
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT06520
| | - Stefano Vanni
- Department of Biology, University of Fribourg, FribourgCH-1700, Switzerland
- Swiss National Center for Competence in Research Bio-Inspired Materials, University of Fribourg, FribourgCH-1700, Switzerland
| |
Collapse
|
27
|
Bartoš L, Menon AK, Vácha R. Insertases scramble lipids: Molecular simulations of MTCH2. Structure 2024; 32:505-510.e4. [PMID: 38377988 PMCID: PMC11001264 DOI: 10.1016/j.str.2024.01.012] [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: 08/21/2023] [Revised: 11/30/2023] [Accepted: 01/24/2024] [Indexed: 02/22/2024]
Abstract
Scramblases play a pivotal role in facilitating bidirectional lipid transport across cell membranes, thereby influencing lipid metabolism, membrane homeostasis, and cellular signaling. MTCH2, a mitochondrial outer membrane protein insertase, has a membrane-spanning hydrophilic groove resembling those that form the lipid transit pathway in known scramblases. Employing both coarse-grained and atomistic molecular dynamics simulations, we show that MTCH2 significantly reduces the free energy barrier for lipid movement along the groove and therefore can indeed function as a scramblase. Notably, the scrambling rate of MTCH2 in silico is similar to that of voltage-dependent anion channel (VDAC), a recently discovered scramblase of the outer mitochondrial membrane, suggesting a potential complementary physiological role for these mitochondrial proteins. Finally, our findings suggest that other insertases which possess a hydrophilic path across the membrane like MTCH2, can also function as scramblases.
Collapse
Affiliation(s)
- Ladislav Bartoš
- CEITEC - Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Anant K Menon
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Robert Vácha
- CEITEC - Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; Department of Condensed Matter Physics, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic.
| |
Collapse
|
28
|
Zhou Y, Li R, Wang K, Lin F, Chen Y, Yang J, Han H, Li T, Jia Y, Yuan K, Zhang H, Li R, Li Z, Zhao Y, Hao Q, Chen X, Zhao Y. A retrospective study on the relationship between serum electrolyte disorder and delayed cerebral infarction after aneurysmal subarachnoid hemorrhage. J Stroke Cerebrovasc Dis 2024; 33:107579. [PMID: 38325032 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107579] [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: 07/12/2023] [Revised: 01/07/2024] [Accepted: 01/11/2024] [Indexed: 02/09/2024] Open
Abstract
OBJECTIVE Delayed cerebral ischemia (DCI)-induced cerebral infarction is a major cause of adverse neurological outcomes following aneurysmal subarachnoid hemorrhage (aSAH). This study aimed to investigate the relationship between postoperative serum electrolyte levels and DCI in patients with aSAH. MATERIALS AND METHODS We analyzed the data of patients with aSAH between 2015 and 2022. The patients were classified into two groups according to whether they experienced DCI. Electrolyte levels were categorized into three groups based on the normal ranges for electrolytes. Logistic regression models were used to study the relationship between electrolyte levels and DCI. Another logistic regression analysis was conducted to explore the relationship between the different severity levels of statistically significant indicators and DCI. A restrictive cubic spline model was adopted to assess the potential linear relationship between electrolytes and DCI. Subsequently, sensitivity analysis was performed to assess the impact of collinearity among ions. Finally, subgroup analysis was performed. RESULTS This study included 1,099 patients. Patients with hyperchloremia were more prone to DCI than those with normal chloride levels. Subsequently, excluding the population with hypochloremia, both mild and severe hyperchloremia were found to be associated with an increased risk of DCI compared with normal chloride levels. Within the framework of a restrictive cubic spline, our findings revealed an increased incidence of DCI (P for nonlinear = 0.735) as chloride levels increased. Sensitivity analysis revealed that patients with severe hyperchloremia were more susceptible to DCI. CONCLUSIONS This study found that patients with aSAH and postoperative hyperchloremia are more prone to developing DCI.
Collapse
Affiliation(s)
- Yunfan Zhou
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Runting Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ke Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fa Lin
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yu Chen
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jun Yang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Heze Han
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tu Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yitong Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kexin Yuan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Haibin Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ruinan Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhipeng Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yahui Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qiang Hao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Stroke Center, Beijing Institute for Brain Disorders, Beijing, China; Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Xiaolin Chen
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Stroke Center, Beijing Institute for Brain Disorders, Beijing, China; Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China.
| | - Yuanli Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China; Stroke Center, Beijing Institute for Brain Disorders, Beijing, China; Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| |
Collapse
|
29
|
Liu X, Wang X, Ma X, Li H, Miao C, Tian Z, Hu Y. Genetic disruption of Ano5 leads to impaired osteoclastogenesis for gnathodiaphyseal dysplasia. Oral Dis 2024; 30:1403-1415. [PMID: 36989132 DOI: 10.1111/odi.14562] [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: 10/22/2022] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/19/2023]
Abstract
OBJECTIVES Gnathodiaphyseal dysplasia (GDD; OMIM#166260) is a rare skeletal genetic disorder characterized by sclerosis of tubular bones and cemento-osseous lesions in mandibles. TMEM16E/ANO5 gene mutations have been identified in patients with GDD. Here, Ano5 knockout (Ano5-/-) mice with enhanced osteoblastogenesis were used to investigate whether Ano5 disruption affects osteoclastogenesis. SUBJECTS AND METHODS The maturation of osteoclasts, formation of F-actin ring and bone resorption were detected by immunohistochemistry, TRAP, phalloidin staining and Coming Osteo assays. The expression of osteoclast-related factors was measured by qRT-PCR. Early signaling pathways were verified by western blot. RESULTS Ano5-/- mice exhibited inhibitory formation of multinucleated osteoclasts with a reduction of TRAP activity. The expression of Nfatc1, c-Fos, Trap, Ctsk, Mmp9, Rank and Dc-stamp was significantly decreased in bone tissues and bone marrow-derived macrophages (BMMs) of Ano5-/- mice. Ano5-/- osteoclasts manifested disrupted actin ring and less mineral resorption. RANKL-induced early signaling pathways were suppressed in Ano5-/- osteoclasts and Ano5 knockdown RAW264.7 cells. Moreover, the inhibitory effects of NF-κB signalling pathway on osteoclastogenesis were partially attenuated with NF-κB signalling activator. CONCLUSIONS Ano5 deficiency impairs osteoclastogenesis, which leads to enhanced osteogenic phenotypes mediated by bone homeostasis dysregulation.
Collapse
Affiliation(s)
- Xiu Liu
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
- Department of Oral Medicine, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Xiaoyu Wang
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Xinrong Ma
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Hongyu Li
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Congcong Miao
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Zhenchuan Tian
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Ying Hu
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| |
Collapse
|
30
|
Han Y, Zhou Z, Jin R, Dai F, Ge Y, Ju X, Ma X, He S, Yuan L, Wang Y, Yang W, Yue X, Chen Z, Sun Y, Corry B, Cox CD, Zhang Y. Mechanical activation opens a lipid-lined pore in OSCA ion channels. Nature 2024; 628:910-918. [PMID: 38570680 DOI: 10.1038/s41586-024-07256-9] [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: 06/26/2023] [Accepted: 02/28/2024] [Indexed: 04/05/2024]
Abstract
OSCA/TMEM63 channels are the largest known family of mechanosensitive channels1-3, playing critical roles in plant4-7 and mammalian8,9 mechanotransduction. Here we determined 44 cryogenic electron microscopy structures of OSCA/TMEM63 channels in different environments to investigate the molecular basis of OSCA/TMEM63 channel mechanosensitivity. In nanodiscs, we mimicked increased membrane tension and observed a dilated pore with membrane access in one of the OSCA1.2 subunits. In liposomes, we captured the fully open structure of OSCA1.2 in the inside-in orientation, in which the pore shows a large lateral opening to the membrane. Unusually for ion channels, structural, functional and computational evidence supports the existence of a 'proteo-lipidic pore' in which lipids act as a wall of the ion permeation pathway. In the less tension-sensitive homologue OSCA3.1, we identified an 'interlocking' lipid tightly bound in the central cleft, keeping the channel closed. Mutation of the lipid-coordinating residues induced OSCA3.1 activation, revealing a conserved open conformation of OSCA channels. Our structures provide a global picture of the OSCA channel gating cycle, uncover the importance of bound lipids and show that each subunit can open independently. This expands both our understanding of channel-mediated mechanotransduction and channel pore formation, with important mechanistic implications for the TMEM16 and TMC protein families.
Collapse
Affiliation(s)
- Yaoyao Han
- 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
| | - Zijing Zhou
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Ruitao Jin
- Research School of Biology, Australian National University, Acton, Australian Capital Territory, Australia
| | - Fei Dai
- 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
| | - Yifan Ge
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Xisan Ju
- 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
| | - Xiaonuo Ma
- 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
| | - Sitong He
- Research School of Biology, Australian National University, Acton, Australian Capital Territory, Australia
| | - Ling Yuan
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yingying Wang
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Yang
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaomin Yue
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhongwen Chen
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Yadong Sun
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Ben Corry
- Research School of Biology, Australian National University, Acton, Australian Capital Territory, Australia.
| | - Charles D Cox
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia.
- School of Biomedical Sciences, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia.
| | - Yixiao Zhang
- 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.
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
31
|
Clark S, Jeong H, Posert R, Goehring A, Gouaux E. The structure of the Caenorhabditis elegans TMC-2 complex suggests roles of lipid-mediated subunit contacts in mechanosensory transduction. Proc Natl Acad Sci U S A 2024; 121:e2314096121. [PMID: 38354260 PMCID: PMC10895266 DOI: 10.1073/pnas.2314096121] [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: 08/15/2023] [Accepted: 12/21/2023] [Indexed: 02/16/2024] Open
Abstract
Mechanotransduction is the process by which a mechanical force, such as touch, is converted into an electrical signal. Transmembrane channel-like (TMC) proteins are an evolutionarily conserved family of membrane proteins whose function has been linked to a variety of mechanosensory processes, including hearing and balance sensation in vertebrates and locomotion in Drosophila. TMC1 and TMC2 are components of ion channel complexes, but the molecular features that tune these complexes to diverse mechanical stimuli are unknown. Caenorhabditis elegans express two TMC homologs, TMC-1 and TMC-2, both of which are the likely pore-forming subunits of mechanosensitive ion channels but differ in their expression pattern and functional role in the worm. Here, we present the single-particle cryo-electron microscopy structure of the native TMC-2 complex isolated from C. elegans. The complex is composed of two copies of the pore-forming TMC-2 subunit, the calcium and integrin binding protein CALM-1 and the transmembrane inner ear protein TMIE. Comparison of the TMC-2 complex to the recently published cryo-EM structure of the C. elegans TMC-1 complex highlights conserved protein-lipid interactions, as well as a π-helical structural motif in the pore-forming helices, that together suggest a mechanism for TMC-mediated mechanosensory transduction.
Collapse
Affiliation(s)
- Sarah Clark
- Vollum Institute, Oregon Health and Science University, Portland, OR97239
| | - Hanbin Jeong
- Vollum Institute, Oregon Health and Science University, Portland, OR97239
| | - Rich Posert
- Vollum Institute, Oregon Health and Science University, Portland, OR97239
| | - April Goehring
- Vollum Institute, Oregon Health and Science University, Portland, OR97239
- HHMI, Oregon Health and Science University, Portland, OR97239
| | - Eric Gouaux
- Vollum Institute, Oregon Health and Science University, Portland, OR97239
- HHMI, Oregon Health and Science University, Portland, OR97239
| |
Collapse
|
32
|
Jojoa-Cruz S, Burendei B, Lee WH, Ward AB. Structure of mechanically activated ion channel OSCA2.3 reveals mobile elements in the transmembrane domain. Structure 2024; 32:157-167.e5. [PMID: 38103547 PMCID: PMC10872982 DOI: 10.1016/j.str.2023.11.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/29/2023] [Accepted: 11/21/2023] [Indexed: 12/19/2023]
Abstract
Members of the OSCA/TMEM63 family are mechanically activated ion channels and structures of some OSCA members have revealed the architecture of these channels and structural features that are potentially involved in mechanosensation. However, these structures are all in a similar state and information about the motion of different elements of the structure is limited, preventing a deeper understanding of how these channels work. Here, we used cryoelectron microscopy to determine high-resolution structures of Arabidopsis thaliana OSCA1.2 and OSCA2.3 in peptidiscs. The structure of OSCA1.2 matches previous structures of the same protein in different environments. Yet, in OSCA2.3, the TM6a-TM7 linker adopts a different conformation that constricts the pore on its cytoplasmic side. Furthermore, coevolutionary sequence analysis uncovered a conserved interaction between the TM6a-TM7 linker and the beam-like domain (BLD). Our results reveal conformational heterogeneity and differences in conserved interactions between the TMD and BLD among members of the OSCA family.
Collapse
Affiliation(s)
- Sebastian Jojoa-Cruz
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Batuujin Burendei
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Wen-Hsin Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
33
|
Feng Z, Alvarenga OE, Accardi A. Structural basis of closed groove scrambling by a TMEM16 protein. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.11.553029. [PMID: 37609346 PMCID: PMC10441378 DOI: 10.1101/2023.08.11.553029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Activation of Ca2+-dependent TMEM16 scramblases induces the externalization of phosphatidylserine, a key molecule in multiple signaling processes. Current models suggest that the TMEM16s scramble lipids by deforming the membrane near a hydrophilic groove, and that Ca2+ dependence arises from the different association of lipids with an open or closed groove. However, the molecular rearrangements involved in groove opening and of how lipids reorganize outside the closed groove remain unknown. Using cryogenic electron microscopy, we directly visualize how lipids associate at the closed groove of Ca2+-bound nhTMEM16 in nanodiscs. Functional experiments pinpoint the lipid-protein interaction sites critical for closed groove scrambling. Structural and functional analyses suggest groove opening entails the sequential appearance of two π-helical turns in the groove-lining TM6 helix and identify critical rearrangements. Finally, we show that the choice of scaffold protein and lipids affects the conformations of nhTMEM16 and their distribution, highlighting a key role of these factors in cryoEM structure determination.
Collapse
Affiliation(s)
- Zhang Feng
- Department of Anesthesiology, Weill Cornell Medical College
| | - Omar E. Alvarenga
- Physiology, Biophysics and Systems Biology Graduate Program, Weill Cornell Medical College
| | - Alessio Accardi
- Department of Anesthesiology, Weill Cornell Medical College
- Department of Physiology and Biophysics, Weill Cornell Medical College
- Department of Biochemistry, Weill Cornell Medical College
| |
Collapse
|
34
|
Ye Z, Galvanetto N, Puppulin L, Pifferi S, Flechsig H, Arndt M, Triviño CAS, Di Palma M, Guo S, Vogel H, Menini A, Franz CM, Torre V, Marchesi A. Structural heterogeneity of the ion and lipid channel TMEM16F. Nat Commun 2024; 15:110. [PMID: 38167485 PMCID: PMC10761740 DOI: 10.1038/s41467-023-44377-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 12/11/2023] [Indexed: 01/05/2024] Open
Abstract
Transmembrane protein 16 F (TMEM16F) is a Ca2+-activated homodimer which functions as an ion channel and a phospholipid scramblase. Despite the availability of several TMEM16F cryogenic electron microscopy (cryo-EM) structures, the mechanism of activation and substrate translocation remains controversial, possibly due to restrictions in the accessible protein conformational space. In this study, we use atomic force microscopy under physiological conditions to reveal a range of structurally and mechanically diverse TMEM16F assemblies, characterized by variable inter-subunit dimerization interfaces and protomer orientations, which have escaped prior cryo-EM studies. Furthermore, we find that Ca2+-induced activation is associated to stepwise changes in the pore region that affect the mechanical properties of transmembrane helices TM3, TM4 and TM6. Our direct observation of membrane remodelling in response to Ca2+ binding along with additional electrophysiological analysis, relate this structural multiplicity of TMEM16F to lipid and ion permeation processes. These results thus demonstrate how conformational heterogeneity of TMEM16F directly contributes to its diverse physiological functions.
Collapse
Affiliation(s)
- Zhongjie Ye
- International School for Advanced Studies (SISSA), 34136, Trieste, Italy
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Nicola Galvanetto
- Department of Physics, University of Zurich, 8057, Zurich, Switzerland
- Department of Biochemistry, University of Zurich, 8057, Zurich, Switzerland
| | - Leonardo Puppulin
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, I-30172 Mestre, Venice, Italy
- WPI Nano Life Science Institute, Kanazawa University, Kakuma-machi, 920-1192, Kanazawa, Japan
| | - Simone Pifferi
- International School for Advanced Studies (SISSA), 34136, Trieste, Italy
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy
| | - Holger Flechsig
- WPI Nano Life Science Institute, Kanazawa University, Kakuma-machi, 920-1192, Kanazawa, Japan
| | - Melanie Arndt
- Department of Biochemistry, University of Zurich, 8057, Zurich, Switzerland
| | | | - Michael Di Palma
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy
| | - Shifeng Guo
- Shenzhen Key Laboratory of Smart Sensing and Intelligent Systems, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Guangdong Provincial Key Lab of Robotics and Intelligent System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Horst Vogel
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
- Institut des Sciences et Ingénierie Chimiques (ISIC), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Anna Menini
- International School for Advanced Studies (SISSA), 34136, Trieste, Italy
| | - Clemens M Franz
- WPI Nano Life Science Institute, Kanazawa University, Kakuma-machi, 920-1192, Kanazawa, Japan
| | - Vincent Torre
- International School for Advanced Studies (SISSA), 34136, Trieste, Italy.
- Institute of Materials (ION-CNR), Area Science Park, Basovizza, 34149, Trieste, Italy.
- BIoValley Investments System and Solutions (BISS), 34148, Trieste, Italy.
| | - Arin Marchesi
- WPI Nano Life Science Institute, Kanazawa University, Kakuma-machi, 920-1192, Kanazawa, Japan.
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy.
| |
Collapse
|
35
|
Whitlock JM. Muscle Progenitor Cell Fusion in the Maintenance of Skeletal Muscle. Results Probl Cell Differ 2024; 71:257-279. [PMID: 37996682 DOI: 10.1007/978-3-031-37936-9_13] [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: 11/25/2023]
Abstract
Skeletal muscle possesses a resident, multipotent stem cell population that is essential for its repair and maintenance throughout life. Here I highlight the role of this stem cell population in muscle repair and regeneration and review the genetic control of the process; the mechanistic steps of activation, migration, recognition, adhesion, and fusion of these cells; and discuss the novel recognition of the membrane signaling that coordinates myogenic cell-cell fusion, as well as the identification of a two-part fusogen system that facilitates it.
Collapse
Affiliation(s)
- Jarred M Whitlock
- Section on Membrane Biology, Eunice Kennedy Shrive National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
36
|
Nguyen DM, Chen TY. Structure and Function of Calcium-Activated Chloride Channels and Phospholipid Scramblases in the TMEM16 Family. Handb Exp Pharmacol 2024; 283:153-180. [PMID: 35792944 DOI: 10.1007/164_2022_595] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The transmembrane protein 16 (TMEM16) family consists of Ca2+-activated chloride channels and phospholipid scramblases. Ten mammalian TMEM16 proteins, TMEM16A-K (with no TMEM16I), and several non-mammalian TMEM16 proteins, such as afTMEM16 and nhTMEM16, have been discovered. All known TMEM16 proteins are homodimeric proteins containing two subunits. Each subunit consists of ten transmembrane helices with Ca2+-binding sites and a single ion-permeation/phospholipid transport pathway. The ion-permeation pathway and the phospholipid transport pathway of TMEM16 proteins have a wide intracellular vestibule, a narrow neck, and a smaller extracellular vestibule. Interestingly, the lining wall of the ion-permeation/phospholipid transport pathway may be formed, at least partially, by membrane phospholipids, though the degree of pore-wall forming by phospholipids likely varies among TMEM16 proteins. Thus, the biophysical properties and activation mechanisms of TMEM16 proteins could differ from each other accordingly. Here we review the current understanding of the structure and function of TMEM16 molecules.
Collapse
Affiliation(s)
- Dung Manh Nguyen
- Center for Neuroscience, University of California, Davis, CA, USA.
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Tsung-Yu Chen
- Department of Neurology, Center for Neuroscience, University of California, Davis, CA, USA.
| |
Collapse
|
37
|
Bartoš L, Menon AK, Vácha R. Insertases Scramble Lipids: Molecular Simulations of MTCH2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.553169. [PMID: 37645813 PMCID: PMC10462046 DOI: 10.1101/2023.08.14.553169] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Scramblases play a pivotal role in facilitating bidirectional lipid transport across cell membranes, thereby influencing lipid metabolism, membrane homeostasis, and cellular signaling. MTCH2, a mitochondrial outer membrane protein insertase, has a membrane-spanning hydrophilic groove resembling those that form the lipid transit pathway in known scramblases. Employing both coarse-grained and atomistic molecular dynamics simulations, we show that MTCH2 significantly reduces the free energy barrier for lipid movement along the groove and therefore can indeed function as a scramblase. Notably, the scrambling rate of MTCH2 in silico is similar to that of VDAC, a recently discovered scramblase of the outer mitochondrial membrane, suggesting a potential complementary physiological role for these mitochondrial proteins. Finally, our findings suggest that other insertases which possess a hydrophilic path across the membrane like MTCH2, can also function as scramblases.
Collapse
Affiliation(s)
- Ladislav Bartoš
- CEITEC - Central European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic
| | - Anant K Menon
- Department of Biochemistry, Weill Cornell Medical College; New York, NY 10065, USA
| | - Robert Vácha
- CEITEC - Central European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic
- Department of Condensed Matter Physics, Faculty of Science, Masaryk University, Kotlarska 267/2, 611 37 Brno, Czech Republic
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic
| |
Collapse
|
38
|
Wang Y, Kinoshita T. The role of lipid scramblases in regulating lipid distributions at cellular membranes. Biochem Soc Trans 2023; 51:1857-1869. [PMID: 37767549 DOI: 10.1042/bst20221455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/12/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023]
Abstract
Glycerophospholipids, sphingolipids and cholesterol assemble into lipid bilayers that form the scaffold of cellular membranes, in which proteins are embedded. Membrane composition and membrane protein profiles differ between plasma and intracellular membranes and between the two leaflets of a membrane. Lipid distributions between two leaflets are mediated by lipid translocases, including flippases and scramblases. Flippases use ATP to catalyze the inward movement of specific lipids between leaflets. In contrast, bidirectional flip-flop movements of lipids across the membrane are mediated by scramblases in an ATP-independent manner. Scramblases have been implicated in disrupting the lipid asymmetry of the plasma membrane, protein glycosylation, autophagosome biogenesis, lipoprotein secretion, lipid droplet formation and communications between organelles. Although scramblases in plasma membranes were identified over 10 years ago, most progress about scramblases localized in intracellular membranes has been made in the last few years. Herein, we review the role of scramblases in regulating lipid distributions in cellular membranes, focusing primarily on intracellular membrane-localized scramblases.
Collapse
Affiliation(s)
- Yicheng Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China
| | - Taroh Kinoshita
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
39
|
Wang L, Bütikofer P. Lactose Permease Scrambles Phospholipids. BIOLOGY 2023; 12:1367. [PMID: 37997967 PMCID: PMC10669175 DOI: 10.3390/biology12111367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 09/30/2023] [Accepted: 10/24/2023] [Indexed: 11/25/2023]
Abstract
Lactose permease (LacY) from Escherichia coli belongs to the major facilitator superfamily. It facilitates the co-transport of β-galactosides, including lactose, into cells by using a proton gradient towards the cell. We now show that LacY is capable of scrambling glycerophospholipids across a membrane. We found that purified LacY reconstituted into liposomes at various protein to lipid ratios catalyzed the rapid translocation of fluorescently labeled and radiolabeled glycerophospholipids across the proteoliposome membrane bilayer. The use of LacY mutant proteins unable to transport lactose revealed that glycerophospholipid scrambling was independent of H+/lactose transport activity. Unexpectedly, in a LacY double mutant locked into an occluded conformation glycerophospholipid, scrambling activity was largely inhibited. The corresponding single mutants revealed the importance of amino acids G46 and G262 for glycerophospholipid scrambling of LacY.
Collapse
Affiliation(s)
| | - Peter Bütikofer
- Institute of Biochemistry and Molecular Medicine, University of Bern, 3012 Bern, Switzerland
| |
Collapse
|
40
|
Zheng W, Rawson S, Shen Z, Tamilselvan E, Smith HE, Halford J, Shen C, Murthy SE, Ulbrich MH, Sotomayor M, Fu TM, Holt JR. TMEM63 proteins function as monomeric high-threshold mechanosensitive ion channels. Neuron 2023; 111:3195-3210.e7. [PMID: 37543036 PMCID: PMC10592209 DOI: 10.1016/j.neuron.2023.07.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 05/12/2023] [Accepted: 07/08/2023] [Indexed: 08/07/2023]
Abstract
OSCA/TMEM63s form mechanically activated (MA) ion channels in plants and animals, respectively. OSCAs and related TMEM16s and transmembrane channel-like (TMC) proteins form homodimers with two pores. Here, we uncover an unanticipated monomeric configuration of TMEM63 proteins. Structures of TMEM63A and TMEM63B (referred to as TMEM63s) revealed a single highly restricted pore. Functional analyses demonstrated that TMEM63s are bona fide mechanosensitive ion channels, characterized by small conductance and high thresholds. TMEM63s possess evolutionary variations in the intracellular linker IL2, which mediates dimerization in OSCAs. Replacement of OSCA1.2 IL2 with TMEM63A IL2 or mutations to key variable residues resulted in monomeric OSCA1.2 and MA currents with significantly higher thresholds. Structural analyses revealed substantial conformational differences in the mechano-sensing domain IL2 and gating helix TM6 between TMEM63s and OSCA1.2. Our studies reveal that mechanosensitivity in OSCA/TMEM63 channels is affected by oligomerization and suggest gating mechanisms that may be shared by OSCA/TMEM63, TMEM16, and TMC channels.
Collapse
Affiliation(s)
- Wang Zheng
- Departments of Otolaryngology & Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Shaun Rawson
- Harvard Cryo-Electron Microscopy Center for Structural Biology, Harvard Medical School, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Zhangfei Shen
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA
| | - Elakkiya Tamilselvan
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
| | - Harper E Smith
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
| | - Julia Halford
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Chen Shen
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Swetha E Murthy
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Maximilian H Ulbrich
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany; Internal Medicine IV, University of Freiburg Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
| | - Tian-Min Fu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA.
| | - Jeffrey R Holt
- Departments of Otolaryngology & Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
41
|
Li S, Wang Z, Geng R, Zhang W, Wan H, Kang X, Guo S. TMEM16A ion channel: A novel target for cancer treatment. Life Sci 2023; 331:122034. [PMID: 37611692 DOI: 10.1016/j.lfs.2023.122034] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/13/2023] [Accepted: 08/18/2023] [Indexed: 08/25/2023]
Abstract
Cancer draws attention owing to the high morbidity and mortality. It is urgent to develop safe and effective cancer therapeutics. The calcium-activated chloride channel TMEM16A is widely distributed in various tissues and regulates physiological functions. TMEM16A is abnormally expressed in several cancers and associate with tumorigenesis, metastasis, and prognosis. Knockdown or inhibition of TMEM16A in cancer cells significantly inhibits cancer development. Therefore, TMEM16A is considered as a biomarker and therapeutic target for some cancers. This work reviews the cancers associated with TMEM16A. Then, the molecular mechanism of TMEM16A overexpression in cancer was analyzed, and the possible signal transduction mechanism of TMEM16A regulating cancer development was summarized. Finally, TMEM16A inhibitors with anticancer effect and their anticancer mechanism were concluded. We hope to provide new ideas for pharmacological studies on TMEM16A in cancer.
Collapse
Affiliation(s)
- Shuting Li
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Zhichen Wang
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Ruili Geng
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Weiwei Zhang
- School of Basic Medical Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Haifu Wan
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China; Institute of Life Sciences and Green Development, Hebei University, Baoding 071002, Hebei, China
| | - Xianjiang Kang
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China; Institute of Life Sciences and Green Development, Hebei University, Baoding 071002, Hebei, China.
| | - Shuai Guo
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China; Institute of Life Sciences and Green Development, Hebei University, Baoding 071002, Hebei, China.
| |
Collapse
|
42
|
Figueiredo IAD, Ferreira SRD, Fernandes JM, Silva BA, Vasconcelos LHC, Cavalcante FA. A review of the pathophysiology and the role of ion channels on bronchial asthma. Front Pharmacol 2023; 14:1236550. [PMID: 37841931 PMCID: PMC10568497 DOI: 10.3389/fphar.2023.1236550] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 09/08/2023] [Indexed: 10/17/2023] Open
Abstract
Asthma is one of the main non-communicable chronic diseases and affects a huge portion of the population. It is a multifactorial disease, classified into several phenotypes, being the allergic the most frequent. The pathophysiological mechanism of asthma involves a Th2-type immune response, with high concentrations of allergen-specific immunoglobulin E, eosinophilia, hyperreactivity and airway remodeling. These mechanisms are orchestrated by intracellular signaling from effector cells, such as lymphocytes and eosinophils. Ion channels play a fundamental role in maintaining the inflammatory response on asthma. In particular, transient receptor potential (TRP), stock-operated Ca2+ channels (SOCs), Ca2+-activated K+ channels (IKCa and BKCa), calcium-activated chloride channel (TMEM16A), cystic fibrosis transmembrane conductance regulator (CFTR), piezo-type mechanosensitive ion channel component 1 (PIEZO1) and purinergic P2X receptor (P2X). The recognition of the participation of these channels in the pathological process of asthma is important, as they become pharmacological targets for the discovery of new drugs and/or pharmacological tools that effectively help the pharmacotherapeutic follow-up of this disease, as well as the more specific mechanisms involved in worsening asthma.
Collapse
Affiliation(s)
- Indyra Alencar Duarte Figueiredo
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Sarah Rebeca Dantas Ferreira
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Jayne Muniz Fernandes
- Graduação em Farmácia, Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Bagnólia Araújo da Silva
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
- Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Luiz Henrique César Vasconcelos
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
- Departamento de Fisiologia e Patologia, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Fabiana de Andrade Cavalcante
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
- Departamento de Fisiologia e Patologia, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| |
Collapse
|
43
|
Zhang P, Maruoka M, Suzuki R, Katani H, Dou Y, Packwood DM, Kosako H, Tanaka M, Suzuki J. Extracellular calcium functions as a molecular glue for transmembrane helices to activate the scramblase Xkr4. Nat Commun 2023; 14:5592. [PMID: 37696806 PMCID: PMC10495444 DOI: 10.1038/s41467-023-40934-2] [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: 01/24/2023] [Accepted: 08/11/2023] [Indexed: 09/13/2023] Open
Abstract
The "eat me" signal, phosphatidylserine is exposed on the surface of dying cells by phospholipid scrambling. Previously, we showed that the Xkr family protein Xkr4 is activated by caspase-mediated cleavage and binding of the XRCC4 fragment. Here, we show that extracellular calcium is an additional factor needed to activate Xkr4. The constitutively active mutant of Xkr4 is found to induce phospholipid scrambling in an extracellular, but not intracellular, calcium-dependent manner. Importantly, other Xkr family members also require extracellular calcium for activation. Alanine scanning shows that D123 and D127 of TM1 and E310 of TM3 coordinate calcium binding. Moreover, lysine scanning demonstrates that the E310K mutation-mediated salt bridge between TM1 and TM3 bypasses the requirement of calcium. Cysteine scanning proves that disulfide bond formation between TM1 and TM3 also activates phospholipid scrambling without calcium. Collectively, this study shows that extracellular calcium functions as a molecular glue for TM1 and TM3 of Xkr proteins for activation, thus demonstrating a regulatory mechanism for multi-transmembrane region-containing proteins.
Collapse
Affiliation(s)
- Panpan Zhang
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto, 606-8501, Japan
- Graduate School of Biostudies, Kyoto University, Konoe-cho, Yoshida, Sakyoku, Kyoto, 606-8501, Japan
| | - Masahiro Maruoka
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto, 606-8501, Japan
- Center for Integrated Biosystems, Institute for Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ryo Suzuki
- Center for Integrative Medicine and Physics (CiMPhy), Institute for Advanced Study, Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto, 606-8501, Japan
| | - Hikaru Katani
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto, 606-8501, Japan
| | - Yu Dou
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto, 606-8501, Japan
- Graduate School of Biostudies, Kyoto University, Konoe-cho, Yoshida, Sakyoku, Kyoto, 606-8501, Japan
| | - Daniel M Packwood
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto, 606-8501, Japan
| | - Hidetaka Kosako
- Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Motomu Tanaka
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto, 606-8501, Japan
- Center for Integrative Medicine and Physics (CiMPhy), Institute for Advanced Study, Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto, 606-8501, Japan
- Physical Chemistry of Biosystems, Institute of Physical Chemistry, Heidelberg University, 69120, Heidelberg, Germany
| | - Jun Suzuki
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto, 606-8501, Japan.
- Graduate School of Biostudies, Kyoto University, Konoe-cho, Yoshida, Sakyoku, Kyoto, 606-8501, Japan.
- Center for Integrated Biosystems, Institute for Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, 332-0012, Japan.
| |
Collapse
|
44
|
Kreiter J, Škulj S, Brkljača Z, Bardakji S, Vazdar M, Pohl EE. FA Sliding as the Mechanism for the ANT1-Mediated Fatty Acid Anion Transport in Lipid Bilayers. Int J Mol Sci 2023; 24:13701. [PMID: 37762012 PMCID: PMC10531397 DOI: 10.3390/ijms241813701] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/27/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
Mitochondrial adenine nucleotide translocase (ANT) exchanges ADP for ATP to maintain energy production in the cell. Its protonophoric function in the presence of long-chain fatty acids (FA) is also recognized. Our previous results imply that proton/FA transport can be best described with the FA cycling model, in which protonated FA transports the proton to the mitochondrial matrix. The mechanism by which ANT1 transports FA anions back to the intermembrane space remains unclear. Using a combined approach involving measurements of the current through the planar lipid bilayers reconstituted with ANT1, site-directed mutagenesis and molecular dynamics simulations, we show that the FA anion is first attracted by positively charged arginines or lysines on the matrix side of ANT1 before moving along the positively charged protein-lipid interface and binding to R79, where it is protonated. We show that R79 is also critical for the competitive binding of ANT1 substrates (ADP and ATP) and inhibitors (carboxyatractyloside and bongkrekic acid). The binding sites are well conserved in mitochondrial SLC25 members, suggesting a general mechanism for transporting FA anions across the inner mitochondrial membrane.
Collapse
Affiliation(s)
- Jürgen Kreiter
- Institute of Physiology, Pathophysiology, and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, 1210 Vienna, Austria; (J.K.); (S.Š.); (S.B.)
| | - Sanja Škulj
- Institute of Physiology, Pathophysiology, and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, 1210 Vienna, Austria; (J.K.); (S.Š.); (S.B.)
| | - Zlatko Brkljača
- Division of Organic Chemistry and Biochemistry, Rudjer Bošković Institute, 10000 Zagreb, Croatia;
| | - Sarah Bardakji
- Institute of Physiology, Pathophysiology, and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, 1210 Vienna, Austria; (J.K.); (S.Š.); (S.B.)
| | - Mario Vazdar
- Department of Mathematics, Informatics, and Cybernetics, University of Chemistry and Technology, 166 28 Prague, Czech Republic
| | - Elena E. Pohl
- Institute of Physiology, Pathophysiology, and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, 1210 Vienna, Austria; (J.K.); (S.Š.); (S.B.)
| |
Collapse
|
45
|
Li D, Rocha-Roa C, Schilling MA, Reinisch KM, Vanni S. Lipid scrambling is a general feature of protein insertases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.01.555937. [PMID: 37693532 PMCID: PMC10491306 DOI: 10.1101/2023.09.01.555937] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Glycerophospholipids are synthesized primarily in the cytosolic leaflet of the endoplasmic reticulum (ER) membrane and must be equilibrated between bilayer leaflets to allow the ER and membranes derived from it to grow. Lipid equilibration is facilitated by integral membrane proteins called "scramblases". These proteins feature a hydrophilic groove allowing the polar heads of lipids to traverse the hydrophobic membrane interior, similar to a credit-card moving through a reader. Nevertheless, despite their fundamental role in membrane expansion and dynamics, the identity of most scramblases has remained elusive. Here, combining biochemical reconstitution and molecular dynamics simulations, we show that lipid scrambling is a general feature of protein insertases, integral membrane proteins which insert polypeptide chains into membranes of the ER and organelles disconnected from vesicle trafficking. Our data indicate that lipid scrambling occurs in the same hydrophilic channel through which protein insertion takes place, and that scrambling is abolished in the presence of nascent polypeptide chains. We propose that protein insertases could have a so-far overlooked role in membrane dynamics as scramblases.
Collapse
Affiliation(s)
- Dazhi Li
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Matthew A. Schilling
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| | - Karin M. Reinisch
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| | - Stefano Vanni
- Department of Biology, University of Fribourg, Switzerland
| |
Collapse
|
46
|
Ilcu L, Denkhaus L, Brausemann A, Zhang L, Einsle O. Architecture of the Heme-translocating CcmABCD/E complex required for Cytochrome c maturation. Nat Commun 2023; 14:5190. [PMID: 37626034 PMCID: PMC10457321 DOI: 10.1038/s41467-023-40881-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Mono- and multiheme cytochromes c are post-translationally matured by the covalent attachment of heme. For this, Escherichia coli employs the most complex type of maturation machineries, the Ccm-system (for cytochrome c maturation). It consists of two membrane protein complexes, one of which shuttles heme across the membrane to a mobile chaperone that then delivers the cofactor to the second complex, an apoprotein:heme lyase, for covalent attachment. Here we report cryo-electron microscopic structures of the heme translocation complex CcmABCD from E. coli, alone and bound to the heme chaperone CcmE. CcmABCD forms a heterooctameric complex centered around the ABC transporter CcmAB that does not by itself transport heme. Our data suggest that the complex flops a heme group from the inner to the outer leaflet at its CcmBC interfaces, driven by ATP hydrolysis at CcmA. A conserved heme-handling motif (WxWD) at the periplasmic side of CcmC rotates the heme by 90° for covalent attachment to the heme chaperone CcmE that we find interacting exclusively with the CcmB subunit.
Collapse
Affiliation(s)
- Lorena Ilcu
- Institut für Biochemie, Albert-Ludwigs-Universität Freiburg, 79104, Freiburg im Breisgau, Germany
| | - Lukas Denkhaus
- Institut für Biochemie, Albert-Ludwigs-Universität Freiburg, 79104, Freiburg im Breisgau, Germany
| | - Anton Brausemann
- Institut für Biochemie, Albert-Ludwigs-Universität Freiburg, 79104, Freiburg im Breisgau, Germany
| | - Lin Zhang
- Institut für Biochemie, Albert-Ludwigs-Universität Freiburg, 79104, Freiburg im Breisgau, Germany.
| | - Oliver Einsle
- Institut für Biochemie, Albert-Ludwigs-Universität Freiburg, 79104, Freiburg im Breisgau, Germany.
| |
Collapse
|
47
|
Clark S, Jeong H, Posert R, Goehring A, Gouaux E. Structure of C. elegans TMC-2 complex suggests roles of lipid-mediated subunit contacts in mechanosensory transduction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.16.553618. [PMID: 37645790 PMCID: PMC10462014 DOI: 10.1101/2023.08.16.553618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Mechanotransduction is the process by which a mechanical force, such as touch, is converted into an electrical signal. Transmembrane channel-like (TMC) proteins are an evolutionarily-conserved family of ion channels whose function has been linked to a variety of mechanosensory processes, including hearing and balance sensation in vertebrates and locomotion in Drosophila. The molecular features that tune homologous TMC ion channel complexes to diverse mechanical stimuli are unknown. Caenorhabditis elegans express two TMC homologs, TMC-1 and TMC-2, both of which are the likely pore-forming subunits of mechanosensitive ion channels but differ in their expression pattern and functional role in the worm. Here we present the single particle cryo-electron microscopy structure of the native TMC-2 complex isolated from C. elegans. The complex is composed of two copies each of the pore-forming TMC-2 subunit, the calcium and integrin binding protein CALM-1 and the transmembrane inner ear protein TMIE. Comparison of the TMC-2 complex to the recently published cryo-EM structure of the C. elegans TMC-1 complex reveals differences in subunit composition and highlights conserved protein-lipid interactions, as well as other structural features, that together suggest a mechanism for TMC-mediated mechanosensory transduction.
Collapse
Affiliation(s)
- Sarah Clark
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Hanbin Jeong
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Rich Posert
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - April Goehring
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
- Howard Hughes Medical Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Eric Gouaux
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
- Howard Hughes Medical Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| |
Collapse
|
48
|
Elverson K, Freeman S, Manson F, Warwicker J. Computational Investigation of Mechanisms for pH Modulation of Human Chloride Channels. Molecules 2023; 28:5753. [PMID: 37570721 PMCID: PMC10420675 DOI: 10.3390/molecules28155753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Many transmembrane proteins are modulated by intracellular or extracellular pH. Investigation of pH dependence generally proceeds by mutagenesis of a wide set of amino acids, guided by properties such as amino-acid conservation and structure. Prediction of pKas can streamline this process, allowing rapid and effective identification of amino acids of interest with respect to pH dependence. Commencing with the calcium-activated chloride channel bestrophin 1, the carboxylate ligand structure around calcium sites relaxes in the absence of calcium, consistent with a measured lack of pH dependence. By contrast, less relaxation in the absence of calcium in TMEM16A, and maintenance of elevated carboxylate sidechain pKas, is suggested to give rise to pH-dependent chloride channel activity. This hypothesis, modulation of calcium/proton coupling and pH-dependent activity through the extent of structural relaxation, is shown to apply to the well-characterised cytosolic proteins calmodulin (pH-independent) and calbindin D9k (pH-dependent). Further application of destabilised, ionisable charge sites, or electrostatic frustration, is made to other human chloride channels (that are not calcium-activated), ClC-2, GABAA, and GlyR. Experimentally determined sites of pH modulation are readily identified. Structure-based tools for pKa prediction are freely available, allowing users to focus on mutagenesis studies, construct hypothetical proton pathways, and derive hypotheses such as the model for control of pH-dependent calcium activation through structural flexibility. Predicting altered pH dependence for mutations in ion channel disorders can support experimentation and, ultimately, clinical intervention.
Collapse
Affiliation(s)
- Kathleen Elverson
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Sally Freeman
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Forbes Manson
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Jim Warwicker
- Division of Molecular and Cellular Function, Faculty of Biology, Medicine and Health, Manchester Institute of Biotechnology, The University of Manchester, Manchester M1 7DN, UK
| |
Collapse
|
49
|
Bergman S, Cater RJ, Plante A, Mancia F, Khelashvili G. Substrate binding-induced conformational transitions in the omega-3 fatty acid transporter MFSD2A. Nat Commun 2023; 14:3391. [PMID: 37296098 PMCID: PMC10250862 DOI: 10.1038/s41467-023-39088-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Major Facilitator Superfamily Domain containing 2 A (MFSD2A) is a transporter that is highly enriched at the blood-brain and blood-retinal barriers, where it mediates Na+-dependent uptake of ω-3 fatty acids in the form of lysolipids into the brain and eyes, respectively. Despite recent structural insights, it remains unclear how this process is initiated, and driven by Na+. Here, we perform Molecular Dynamics simulations which demonstrate that substrates enter outward facing MFSD2A from the outer leaflet of the membrane via lateral openings between transmembrane helices 5/8 and 2/11. The substrate headgroup enters first and engages in Na+ -bridged interactions with a conserved glutamic acid, while the tail is surrounded by hydrophobic residues. This binding mode is consistent with a "trap-and-flip" mechanism and triggers transition to an occluded conformation. Furthermore, using machine learning analysis, we identify key elements that enable these transitions. These results advance our molecular understanding of the MFSD2A transport cycle.
Collapse
Affiliation(s)
- Shana Bergman
- Department of Physiology and Biophysics, Weill Cornell Medical College, Cornell University, New York, NY, 10065, USA
| | - Rosemary J Cater
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, 10032, USA
| | - Ambrose Plante
- Department of Physiology and Biophysics, Weill Cornell Medical College, Cornell University, New York, NY, 10065, USA
| | - Filippo Mancia
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, 10032, USA
| | - George Khelashvili
- Department of Physiology and Biophysics, Weill Cornell Medical College, Cornell University, New York, NY, 10065, USA.
- Institute for Computational Biomedicine, Weill Cornell Medical College, Cornell University, New York, NY, 10065, USA.
| |
Collapse
|
50
|
Sakuragi T, Nagata S. Regulation of phospholipid distribution in the lipid bilayer by flippases and scramblases. Nat Rev Mol Cell Biol 2023:10.1038/s41580-023-00604-z. [PMID: 37106071 PMCID: PMC10134735 DOI: 10.1038/s41580-023-00604-z] [Citation(s) in RCA: 93] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2023] [Indexed: 04/29/2023]
Abstract
Cellular membranes function as permeability barriers that separate cells from the external environment or partition cells into distinct compartments. These membranes are lipid bilayers composed of glycerophospholipids, sphingolipids and cholesterol, in which proteins are embedded. Glycerophospholipids and sphingolipids freely move laterally, whereas transverse movement between lipid bilayers is limited. Phospholipids are asymmetrically distributed between membrane leaflets but change their location in biological processes, serving as signalling molecules or enzyme activators. Designated proteins - flippases and scramblases - mediate this lipid movement between the bilayers. Flippases mediate the confined localization of specific phospholipids (phosphatidylserine (PtdSer) and phosphatidylethanolamine) to the cytoplasmic leaflet. Scramblases randomly scramble phospholipids between leaflets and facilitate the exposure of PtdSer on the cell surface, which serves as an important signalling molecule and as an 'eat me' signal for phagocytes. Defects in flippases and scramblases cause various human diseases. We herein review the recent research on the structure of flippases and scramblases and their physiological roles. Although still poorly understood, we address the mechanisms by which they translocate phospholipids between lipid bilayers and how defects cause human diseases.
Collapse
Affiliation(s)
- Takaharu Sakuragi
- Biochemistry & Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Shigekazu Nagata
- Biochemistry & Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.
| |
Collapse
|