Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Cell Calcium ; 121: 102896, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38749289

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.


Subject(s)
Anoctamins , Phospholipid Transfer Proteins , Humans , Anoctamins/metabolism , Anoctamins/chemistry , Animals , Phospholipid Transfer Proteins/metabolism , Phospholipid Transfer Proteins/chemistry
2.
Cell Calcium ; 120: 102888, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38657371

ABSTRACT

Plasma membrane localized anoctamin 1, 2 and 6 (TMEM16A, B, F) have been examined in great detail with respect to structure and function, but much less is known about the other seven intracellular members of this exciting family of proteins. This is probably due to their limited accessibility in intracellular membranous compartments, such as the endoplasmic reticulum (ER) or endosomes. However, these so-called intracellular anoctamins are also found in the plasma membrane (PM) which adds to the confusion regarding their cellular role. Probably all intracellular anoctamins except of ANO8 operate as intracellular phospholipid (PL) scramblases, allowing for Ca2+-activated, passive transport of phospholipids like phosphatidylserine between both membrane leaflets. Probably all of them also conduct ions, which is probably part of their physiological function. In this brief overview, we summarize key findings on the biological functions of ANO3, 4, 5, 7, 8, 9 and 10 (TMEM16C, D, E, G, H, J, K) that are gradually coming to light. Compartmentalized regulation of intracellular Ca2+ signals, tethering of the ER to specific PM contact sites, and control of intracellular vesicular trafficking appear to be some of the functions of intracellular anoctamins, while loss of function and abnormal expression are the cause for various diseases.


Subject(s)
Anoctamins , Humans , Anoctamins/metabolism , Anoctamins/chemistry , Animals , Cell Membrane/metabolism , Structure-Activity Relationship
3.
Nature ; 628(8009): 910-918, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38570680

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.


Subject(s)
Calcium Channels , Cryoelectron Microscopy , Ion Channel Gating , Mechanotransduction, Cellular , Humans , Anoctamins/chemistry , Anoctamins/metabolism , Calcium Channels/chemistry , Calcium Channels/metabolism , Calcium Channels/ultrastructure , Lipids/chemistry , Liposomes/metabolism , Liposomes/chemistry , Models, Molecular , Nanostructures/chemistry
4.
Commun Biol ; 5(1): 990, 2022 09 19.
Article in English | MEDLINE | ID: mdl-36123525

ABSTRACT

TMEM16F is a Ca2+-activated phospholipid scramblase in the TMEM16 family of membrane proteins. Unlike other TMEM16s exhibiting a membrane-exposed hydrophilic groove that serves as a translocation pathway for lipids, the experimentally determined structures of TMEM16F shows the groove in a closed conformation even under conditions of maximal scramblase activity. It is currently unknown if/how TMEM16F groove can open for lipid scrambling. Here we describe the analysis of ~400 µs all-atom molecular dynamics (MD) simulations of the TMEM16F revealing an allosteric mechanism leading to an open-groove, lipid scrambling competent state of the protein. The groove opens into a continuous hydrophilic conduit that is highly similar in structure to that seen in other activated scramblases. The allosteric pathway connects this opening to an observed destabilization of the Ca2+ ion bound at the distal site near the dimer interface, to the dynamics of specific protein regions that produces the open-groove state to scramble phospholipids.


Subject(s)
Anoctamins , Phospholipid Transfer Proteins , Anoctamins/chemistry , Anoctamins/genetics , Anoctamins/metabolism , Cell Membrane/metabolism , Electric Conductivity , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/metabolism , Phospholipids/metabolism
5.
Biophys J ; 121(18): 3445-3457, 2022 09 20.
Article in English | MEDLINE | ID: mdl-35978550

ABSTRACT

Transmembrane protein 16F (TMEM16F) is a ubiquitously expressed Ca2+-activated phospholipid scramblase that also functions as a largely non-selective ion channel. Though recent structural studies have revealed the closed and intermediate conformations of mammalian TMEM16F (mTMEM16F), the open and conductive state remains elusive. Instead, it has been proposed that an open hydrophilic pathway may not be required for lipid scrambling. We previously identified an inner activation gate, consisting of F518, Y563, and I612, and showed that charged mutations of the inner gate residues led to constitutively active mTMEM16F scrambling. Herein, atomistic simulations show that lysine substitution of F518 and Y563 can indeed lead to spontaneous opening of the permeation pore in the Ca2+-bound state of mTMEM16F. Dilation of the pore exposes hydrophilic patches in the upper pore region, greatly increases the pore hydration level, and enables lipid scrambling. The putative open state of mTMEM16F resembles the active state of fungal scramblases and is a meta-stable state for the wild-type protein in the Ca2+-bound state. Therefore, mTMEM16F may be capable of supporting the canonical in-groove scrambling mechanism in addition to the out-of-groove one. Further analysis reveals that the in-groove phospholipid and ion transduction pathways of mTMEM16F overlap from the intracellular side up to the inner gate but diverge from each other with different exits to the extracellular side of membrane.


Subject(s)
Anoctamins , Phospholipid Transfer Proteins , Animals , Anoctamins/chemistry , Anoctamins/genetics , Anoctamins/metabolism , Ion Channels/metabolism , Lysine , Mammals/metabolism , Mutation , Phospholipid Transfer Proteins/metabolism , Phospholipids/chemistry
6.
Nat Commun ; 12(1): 6913, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824283

ABSTRACT

Tweety homologs (TTYHs) comprise a conserved family of transmembrane proteins found in eukaryotes with three members (TTYH1-3) in vertebrates. They are widely expressed in mammals including at high levels in the nervous system and have been implicated in cancers and other diseases including epilepsy, chronic pain, and viral infections. TTYHs have been reported to form Ca2+- and cell volume-regulated anion channels structurally distinct from any characterized protein family with potential roles in cell adhesion, migration, and developmental signaling. To provide insight into TTYH family structure and function, we determined cryo-EM structures of Mus musculus TTYH2 and TTYH3 in lipid nanodiscs. TTYH2 and TTYH3 adopt a previously unobserved fold which includes an extended extracellular domain with a partially solvent exposed pocket that may be an interaction site for hydrophobic molecules. In the presence of Ca2+, TTYH2 and TTYH3 form homomeric cis-dimers bridged by extracellularly coordinated Ca2+. Strikingly, in the absence of Ca2+, TTYH2 forms trans-dimers that span opposing membranes across a ~130 Å intermembrane space as well as a monomeric state. All TTYH structures lack ion conducting pathways and we do not observe TTYH2-dependent channel activity in cells. We conclude TTYHs are not pore forming subunits of anion channels and their function may involve Ca2+-dependent changes in quaternary structure, interactions with hydrophobic molecules near the extracellular membrane surface, and/or association with additional protein partners.


Subject(s)
Chloride Channels/chemistry , Chloride Channels/metabolism , Dimerization , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Animals , Anoctamins/chemistry , Biological Transport , Calcium/metabolism , Cell Adhesion , Cell Size , Chloride Channels/genetics , Chronic Pain , Cryoelectron Microscopy , Eukaryota , Hydrophobic and Hydrophilic Interactions , Membrane Proteins/genetics , Mice , Receptor, EphB2 , Signal Transduction
7.
Int J Mol Sci ; 22(12)2021 Jun 18.
Article in English | MEDLINE | ID: mdl-34207150

ABSTRACT

P2X7 receptors (P2X7) are cationic channels involved in many diseases. Following their activation by extracellular ATP, distinct signaling pathways are triggered, which lead to various physiological responses such as the secretion of pro-inflammatory cytokines or the modulation of cell death. P2X7 also exhibit unique behaviors, such as "macropore" formation, which corresponds to enhanced large molecule cell membrane permeability and current facilitation, which is caused by prolonged activation. These two phenomena have often been confounded but, thus far, no clear mechanisms have been resolved. Here, by combining different approaches including whole-cell and single-channel recordings, pharmacological and biochemical assays, CRISPR/Cas9 technology and cell imaging, we provide evidence that current facilitation and macropore formation involve functional complexes comprised of P2X7 and TMEM16, a family of Ca2+-activated ion channel/scramblases. We found that current facilitation results in an increase of functional complex-embedded P2X7 open probability, a result that is recapitulated by plasma membrane cholesterol depletion. We further show that macropore formation entails two distinct large molecule permeation components, one of which requires functional complexes featuring TMEM16F subtype, the other likely being direct permeation through the P2X7 pore itself. Such functional complexes can be considered to represent a regulatory hub that may orchestrate distinct P2X7 functionalities.


Subject(s)
Anoctamins/metabolism , Receptors, Purinergic P2X7/metabolism , Adenosine Triphosphate/metabolism , Algorithms , Animals , Anoctamins/chemistry , CRISPR-Cas Systems , Cell Membrane/metabolism , Cell Membrane Permeability , Cholesterol/metabolism , HEK293 Cells , Humans , Immunohistochemistry , Models, Biological , Oocytes , Receptors, Purinergic P2X7/chemistry
8.
Tissue Cell ; 72: 101546, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33940566

ABSTRACT

Anoctamin 7 (ANO7) is a member of the transmembrane protein TMEM16 family. It has a conservative topology similar to other members in this family, such as the typical eight-transmembrane domain, but it also has unique features. Although the ion channel role of ANO7 has been well accepted, evolutionary analyses and relevant studies suggest that ANO7 may be a multi-facet protein in function. Studies have shown that ANO7 may also function as a scramblase. ANO7 is highly expressed in prostate cancer as well as normal prostate tissues. A considerable amount of evidence has confirmed that ANO7 is associated with human physiology and pathology, particularly with the development of prostate cancer, which makes ANO7 a good candidate as a diagnostic and prognostic biomarker. In addition, ANO7 may be a potential target for prostate cancer immunotherapy. Antibody-based or T cell-mediated immunotherapies against prostate cancer by targeting ANO7 have been highly anticipated. ANO7 may also correlate with several other types of cancers or diseases, where further studies are warranted.


Subject(s)
Anoctamins/chemistry , Anoctamins/metabolism , Biomarkers/metabolism , Immunotherapy , Animals , Cells/metabolism , Humans , Ion Channels/metabolism , Models, Biological
9.
Nat Commun ; 12(1): 2826, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33990555

ABSTRACT

TMEM16 lipid scramblases transport lipids and also operate as ion channels with highly variable ion selectivities and various physiological functions. However, their molecular mechanisms of ion conduction and selectivity remain largely unknown. Using computational electrophysiology simulations at atomistic resolution, we identified the main ion-conductive state of TMEM16 lipid scramblases, in which an ion permeation pathway is lined by lipid headgroups that directly interact with permeating ions in a voltage polarity-dependent manner. We found that lipid headgroups modulate the ion-permeability state and regulate ion selectivity to varying degrees in different scramblase isoforms, depending on the amino-acid composition of the pores. Our work has defined the structural basis of ion conduction and selectivity in TMEM16 lipid scramblases and uncovered the mechanisms responsible for the direct effects of membrane lipids on the conduction properties of ion channels.


Subject(s)
Anoctamins/metabolism , Fungal Proteins/metabolism , Membrane Lipids/metabolism , Phospholipid Transfer Proteins/metabolism , Anoctamins/chemistry , Fungal Proteins/chemistry , Fusarium/metabolism , Ion Transport , Membrane Lipids/chemistry , Models, Molecular , Phospholipid Transfer Proteins/chemistry , Protein Conformation , Protein Structure, Quaternary , Static Electricity
10.
Int J Mol Sci ; 22(5)2021 Mar 08.
Article in English | MEDLINE | ID: mdl-33800471

ABSTRACT

The anoctamin (TMEM16) family of transmembrane protein consists of ten members in vertebrates, which act as Ca2+-dependent ion channels and/or Ca2+-dependent scramblases. ANO4 which is primarily expressed in the CNS and certain endocrine glands, has been associated with various neuronal disorders. Therefore, we focused our study on prioritizing missense mutations that are assumed to alter the structure and stability of ANO4 protein. We employed a wide array of evolution and structure based in silico prediction methods to identify potentially deleterious missense mutations in the ANO4 gene. Identified pathogenic mutations were then mapped to the modeled human ANO4 structure and the effects of missense mutations were studied on the atomic level using molecular dynamics simulations. Our data show that the G80A and A500T mutations significantly alter the stability of the mutant proteins, thus providing new perspective on the role of missense mutations in ANO4 gene. Results obtained in this study may help to identify disease associated mutations which affect ANO4 protein structure and function and might facilitate future functional characterization of ANO4.


Subject(s)
Amino Acid Substitution , Anoctamins , Mutation, Missense , Sequence Analysis, Protein , Anoctamins/chemistry , Anoctamins/genetics , Humans , Protein Stability
11.
Int J Mol Sci ; 22(4)2021 Feb 23.
Article in English | MEDLINE | ID: mdl-33672260

ABSTRACT

Intracellular divalent cations control the molecular function of transmembrane protein 16 (TMEM16) family members. Both anion channels (such as TMEM16A) and phospholipid scramblases (such as TMEM16F) in this family are activated by intracellular Ca2+ in the low µM range. In addition, intracellular Ca2+ or Co2+ at mM concentrations have been shown to further potentiate the saturated Ca2+-activated current of TMEM16A. In this study, we found that all alkaline earth divalent cations in mM concentrations can generate similar potentiation effects in TMEM16A when applied intracellularly, and that manipulations thought to deplete membrane phospholipids weaken the effect. In comparison, mM concentrations of divalent cations minimally potentiate the current of TMEM16F but significantly change its cation/anion selectivity. We suggest that divalent cations may increase local concentrations of permeant ions via a change in pore electrostatic potential, possibly acting through phospholipid head groups in or near the pore. Monovalent cations appear to exert a similar effect, although with a much lower affinity. Our findings resolve controversies regarding the ion selectivity of TMEM16 proteins. The physiological role of this mechanism, however, remains elusive because of the nearly constant high cation concentrations in cytosols.


Subject(s)
Anoctamins/metabolism , Cations, Divalent/metabolism , Anoctamin-1/chemistry , Anoctamin-1/genetics , Anoctamin-1/metabolism , Anoctamins/chemistry , Anoctamins/genetics , Calcium/metabolism , Cations, Divalent/pharmacology , Cobalt/metabolism , Electrophysiology/methods , HEK293 Cells , Humans , Magnesium/metabolism , Mannitol/metabolism , Mannitol/pharmacology , Mutation , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipids/metabolism , Polylysine/pharmacology
12.
Mol Cells ; 44(2): 88-100, 2021 Feb 28.
Article in English | MEDLINE | ID: mdl-33658434

ABSTRACT

Anoctamin 6/TMEM16F (ANO6) is a dual-function protein with Ca2+-activated ion channel and Ca2+-activated phospholipid scramblase activities, requiring a high intracellular Ca2+ concentration (e.g., half-maximal effective Ca2+ concentration [EC50] of [Ca2+]i > 10 µM), and strong and sustained depolarization above 0 mV. Structural comparison with Anoctamin 1/TMEM16A (ANO1), a canonical Ca2+- activated chloride channel exhibiting higher Ca2+ sensitivity (EC50 of 1 µM) than ANO6, suggested that a homologous Ca2+-transferring site in the N-terminal domain (Nt) might be responsible for the differential Ca2+ sensitivity and kinetics of activation between ANO6 and ANO1. To elucidate the role of the putative Ca2+-transferring reservoir in the Nt (Nt-CaRes), we constructed an ANO6-1-6 chimera in which Nt-CaRes was replaced with the corresponding domain of ANO1. ANO6- 1-6 showed higher sensitivity to Ca2+ than ANO6. However, neither the speed of activation nor the voltage-dependence differed between ANO6 and ANO6-1-6. Molecular dynamics simulation revealed a reduced Ca2+ interaction with Nt- CaRes in ANO6 than ANO6-1-6. Moreover, mutations on potentially Ca2+-interacting acidic amino acids in ANO6 Nt- CaRes resulted in reduced Ca2+ sensitivity, implying direct interactions of Ca2+ with these residues. Based on these results, we cautiously suggest that the net charge of Nt- CaRes is responsible for the difference in Ca2+ sensitivity between ANO1 and ANO6.


Subject(s)
Anoctamins/chemistry , Anoctamins/metabolism , Calcium/metabolism , Phospholipid Transfer Proteins/chemistry , Phospholipid Transfer Proteins/metabolism , Amino Acid Sequence , Anoctamins/genetics , EF Hand Motifs , HEK293 Cells , Humans , Models, Biological , Molecular Dynamics Simulation , Mutation/genetics , Phospholipid Transfer Proteins/genetics , Protein Domains , Structure-Activity Relationship
13.
J Mol Biol ; 433(16): 166941, 2021 08 06.
Article in English | MEDLINE | ID: mdl-33741412

ABSTRACT

The TMEM16 family of membrane proteins displays a remarkable functional dichotomy - while some family members function as Ca2+-activated anion channels, the majority of characterized TMEM16 homologs are Ca2+-activated lipid scramblases, which catalyze the exchange of phospholipids between the two membrane leaflets. Furthermore, some TMEM16 scramblases can also function as channels. Due to their involvement in important physiological processes, the family has been actively studied ever since their molecular identity was unraveled. In this review, we will summarize the recent advances in the field and how they influenced our view of TMEM16 family function and evolution. Structural, functional and computational studies reveal how relatively small rearrangements in the permeation pathway are responsible for the observed functional duality: while TMEM16 scramblases can adopt both ion- and lipid conductive conformations, TMEM16 channels can only populate the former. Recent data further provides the molecular details of a stepwise activation mechanism, which is initiated by Ca2+ binding and modulated by various cellular factors, including lipids. TMEM16 function and the surrounding membrane properties are inextricably intertwined, with the protein inducing bilayer deformations associated with scrambling, while the surrounding lipids modulate TMEM16 conformation and activity.


Subject(s)
Anoctamins/chemistry , Anoctamins/metabolism , Animals , Calcium/metabolism , Humans , Ion Transport , Lipid Metabolism , Models, Molecular , Protein Binding , Protein Conformation , Signal Transduction , Structure-Activity Relationship
14.
J Cell Biol ; 220(3)2021 03 01.
Article in English | MEDLINE | ID: mdl-33496727

ABSTRACT

Mutations in ANO5 (TMEM16E) cause limb-girdle muscular dystrophy R12. Defective plasma membrane repair is a likely mechanism. Using myofibers from Ano5 knockout mice, we show that trafficking of several annexin proteins, which together form a cap at the site of injury, is altered upon loss of ANO5. Annexin A2 accumulates at the wound to nearly twice the level observed in WT fibers, while annexin A6 accumulation is substantially inhibited in the absence of ANO5. Appearance of annexins A1 and A5 at the cap is likewise diminished in the Ano5 knockout. These changes are correlated with an alteration in annexin repair cap fine structure and shedding of annexin-positive vesicles. We conclude that loss of annexin coordination during repair is disrupted in Ano5 knockout mice and underlies the defective repair phenotype. Although ANO5 is a phospholipid scramblase, abnormal repair is rescued by overexpression of a scramblase-defective ANO5 mutant, suggesting a novel, scramblase-independent role of ANO5 in repair.


Subject(s)
Annexins/metabolism , Muscle Fibers, Skeletal/metabolism , Animals , Anoctamins/chemistry , Anoctamins/deficiency , Anoctamins/genetics , Anoctamins/metabolism , Calcium/metabolism , Cell Membrane/metabolism , Cells, Cultured , Humans , Kinetics , Mice, Knockout , Mutation/genetics , Phosphatidylethanolamines/metabolism , Phosphatidylserines/metabolism , Protein Domains , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism
15.
J Biol Chem ; 295(35): 12537-12544, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32709749

ABSTRACT

TMEM16 Ca2+-activated phospholipid scramblases (CaPLSases) mediate rapid transmembrane phospholipid flip-flop and as such play essential roles in various physiological and pathological processes such as blood coagulation, skeletal development, viral infection, cell-cell fusion, and ataxia. Pharmacological tools specifically targeting TMEM16 CaPLSases are urgently needed to understand these novel membrane transporters and their contributions to health and disease. Tannic acid (TA) and epigallocatechin gallate (EGCG) were recently reported as promising TMEM16F CaPLSase inhibitors. However, our present study shows that TA and EGCG do not inhibit the phospholipid-scrambling or ion conduction activities of the dual-functional TMEM16F. Instead, we found that TA and EGCG mainly acted as fluorescence quenchers that rapidly suppress the fluorophores conjugated to annexin V, a phosphatidylserine-binding probe commonly used to report on TMEM16 CaPLSase activity. These data demonstrate the false positive effects of TA and EGCG on inhibiting TMEM16F phospholipid scrambling and discourage the use of these polyphenols as CaPLSase inhibitors. Appropriate controls as well as a combination of both fluorescence imaging and electrophysiological validation are necessary in future endeavors to develop TMEM16 CaPLSase inhibitors.


Subject(s)
Anoctamins/chemistry , Phospholipid Transfer Proteins/chemistry , Phospholipids/chemistry , Animals , Anoctamins/antagonists & inhibitors , Anoctamins/metabolism , Catechin/analogs & derivatives , Catechin/chemistry , Catechin/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Mice , Phospholipid Transfer Proteins/antagonists & inhibitors , Phospholipid Transfer Proteins/metabolism , Phospholipids/metabolism , Polyphenols/chemistry , Polyphenols/pharmacology , Tannins/chemistry , Tannins/pharmacology
16.
Methods Mol Biol ; 2127: 207-225, 2020.
Article in English | MEDLINE | ID: mdl-32112325

ABSTRACT

Phospholipid scramblases catalyze the rapid trans-bilayer movement of lipids down their concentration gradients. This process is essential for numerous cellular signaling functions including cell fusion, blood coagulation, and apoptosis. The importance of scramblases is highlighted by the number of human diseases caused by mutations in these proteins. Because of their indispensable function, it is essential to understand and characterize the molecular function of phospholipid scramblases. Powerful tools to measure lipid transport in cells are available. However, these approaches provide limited mechanistic insights into the molecular bases of scrambling. Here we describe in detail an in vitro phospholipid scramblase assay and the accompanying analysis which allows for determination of the macroscopic rate constants associated with phospholipid scrambling. Notably, members of the TMEM16 family of scramblases also function as nonselective ion channels. To better understand the physiological relevance of this channel function as well as its relationship to the scrambling activity of the TMEM16s we also describe in detail an in vitro flux assay to measure nonselective channel activity. Together, these two assays can be used to investigate the dual activities of the TMEM16 scramblases/nonselective channels.


Subject(s)
Biological Assay/methods , Ion Channels/metabolism , Phospholipid Transfer Proteins/metabolism , Phospholipids/metabolism , Proteolipids/metabolism , Animals , Anoctamins/chemistry , Anoctamins/metabolism , Fluorescence , Humans , Ion Channels/chemistry , Ion Transport , Ions/metabolism , Liposomes/chemistry , Liposomes/metabolism , Models, Theoretical , Phospholipids/chemistry , Phospholipids/isolation & purification , Protein Renaturation , Proteolipids/chemistry , Proteolipids/isolation & purification
17.
Methods Mol Biol ; 2127: 245-273, 2020.
Article in English | MEDLINE | ID: mdl-32112327

ABSTRACT

Single-particle cryo-electron microscopy has become an indispensable technique in structural biology. In particular when studying membrane proteins, it allows the use of membrane-mimicking tools, which can be crucial for a comprehensive understanding of the structure-function relationship of the protein in its native environment. In this chapter we focus on the application of nanodiscs and use our recent studies on the TMEM16 family as an example.


Subject(s)
Cryoelectron Microscopy/methods , Lipid Bilayers/chemistry , Membrane Proteins/chemistry , Single Molecule Imaging/methods , Animals , Anoctamins/chemistry , Anoctamins/metabolism , Data Collection/methods , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Fusarium , Humans , Image Processing, Computer-Assisted/methods , Lipid Bilayers/metabolism , Membrane Proteins/metabolism , Models, Molecular , Nanostructures/chemistry , Protein Conformation
18.
J Comput Chem ; 41(6): 538-551, 2020 03 05.
Article in English | MEDLINE | ID: mdl-31750558

ABSTRACT

Recent discoveries about functional mechanisms of proteins in the TMEM16 family of phospholipid scramblases have illuminated the dual role of the membrane as both the substrate and a mechanistically responsive environment in the wide range of physiological processes and genetic disorders in which they are implicated. This is highlighted in the review of recent findings from our collaborative investigations of molecular mechanisms of TMEM16 scramblases that emerged from iterative functional, structural, and computational experimentation. In the context of this review, we present new MD simulations and trajectory analyses motivated by the fact that new structural information about the TMEM16 scramblases is emerging from cryo-EM determinations in lipid nanodiscs. Because the functional environment of these proteins in in vivo and in in vitro is closer to flat membranes, we studied comparatively the responses of the membrane to the TMEM16 proteins in flat membranes and nanodiscs. We find that bilayer shapes in the nanodiscs are very different from those observed in the flat membrane systems, but the function-related slanting of the membrane observed at the nhTMEM16 boundary with the protein is similar in the nanodiscs and in the flat bilayers. This changes, however, in the bilayer composed of longer-tail lipids, which is thicker near the phospholipid translocation pathway, which may reflect an enhanced tendency of the long tails to penetrate the pathway and create, as shown previously, a nonconductive environment. These findings support the correspondence between the mechanistic involvement of the lipid environment in the flat membranes, and the nanodiscs. © 2019 Wiley Periodicals, Inc.


Subject(s)
Anoctamins/chemistry , Membrane Lipids/chemistry , Phospholipid Transfer Proteins/chemistry , Anoctamins/metabolism , Membrane Lipids/metabolism , Molecular Dynamics Simulation , Phospholipid Transfer Proteins/metabolism
19.
Nat Commun ; 10(1): 3956, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31477691

ABSTRACT

Membranes in cells have defined distributions of lipids in each leaflet, controlled by lipid scramblases and flip/floppases. However, for some intracellular membranes such as the endoplasmic reticulum (ER) the scramblases have not been identified. Members of the TMEM16 family have either lipid scramblase or chloride channel activity. Although TMEM16K is widely distributed and associated with the neurological disorder autosomal recessive spinocerebellar ataxia type 10 (SCAR10), its location in cells, function and structure are largely uncharacterised. Here we show that TMEM16K is an ER-resident lipid scramblase with a requirement for short chain lipids and calcium for robust activity. Crystal structures of TMEM16K show a scramblase fold, with an open lipid transporting groove. Additional cryo-EM structures reveal extensive conformational changes from the cytoplasmic to the ER side of the membrane, giving a state with a closed lipid permeation pathway. Molecular dynamics simulations showed that the open-groove conformation is necessary for scramblase activity.


Subject(s)
Anoctamins/metabolism , Endoplasmic Reticulum/metabolism , Lipids/chemistry , Phospholipid Transfer Proteins/metabolism , Amino Acid Sequence , Animals , Anoctamins/chemistry , Anoctamins/genetics , COS Cells , Calcium/chemistry , Cell Line, Tumor , Chlorocebus aethiops , Crystallography, X-Ray , HEK293 Cells , Humans , Molecular Dynamics Simulation , Phospholipid Transfer Proteins/chemistry , Phospholipid Transfer Proteins/genetics , Sequence Homology, Amino Acid , Sf9 Cells , Spodoptera
20.
Elife ; 82019 07 18.
Article in English | MEDLINE | ID: mdl-31318330

ABSTRACT

TMEM16F is activated by elevated intracellular Ca2+, and functions as a small-conductance ion channel and as a phospholipid scramblase. In contrast to its paralogs, the TMEM16A/B calcium-activated chloride channels, mouse TMEM16F has been reported as a cation-, anion-, or non-selective ion channel, without a definite conclusion. Starting with the Q559K mutant that shows no current rundown and less outward rectification in excised patch, we found that the channel shifted its ion selectivity in response to the change of intracellular Ca2+ concentration, with an increased permeability ratio of Cl- to Na+ (PCl-/PNa+) at a higher Ca2+ level. The gradual shift of relative ion permeability did not correlate with the channel activation state. Instead, it was indicative of an alteration of electrostatic field in the permeation pathway. The dynamic change of ion selectivity suggests a charge-screening mechanism for TMEM16F ion conduction, and it provides hints to further studies of TMEM16F physiological functions.


Subject(s)
Anions/metabolism , Anoctamins/chemistry , Anoctamins/metabolism , Cations/metabolism , Phospholipid Transfer Proteins/chemistry , Phospholipid Transfer Proteins/metabolism , Animals , Anoctamins/genetics , DNA Mutational Analysis , Mice , Phospholipid Transfer Proteins/genetics , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...