scispace - formally typeset
Search or ask a question
Journal ArticleDOI

Mechanistic diversity in ATP-binding cassette (ABC) transporters

Kaspar P. Locher1
01 Jun 2016-Nature Structural & Molecular Biology (Nature Publishing Group)-Vol. 23, Iss: 6, pp 487-493
TL;DR: This review evaluates the differing mechanistic proposals and outlines future directions for the exploration of ABC-transporter-catalyzed reactions.
Abstract: ABC transporters catalyze transport reactions, such as the high-affinity uptake of micronutrients into bacteria and the export of cytotoxic compounds from mammalian cells. Crystal structures of ABC domains and full transporters have provided a framework for formulating reaction mechanisms of ATP-driven substrate transport, but recent studies have suggested remarkable mechanistic diversity within this protein family. This review evaluates the differing mechanistic proposals and outlines future directions for the exploration of ABC-transporter-catalyzed reactions.

Summary (2 min read)

Introduction

  • ABC transporters are a family of membrane proteins that mediate diverse, ATP-driven transport processes.
  • NBDs hydrolyze ATP and drive conformational changes in the attached transmembrane domains (TMDs), allowing substrates to cross the lipid bilayer of the membrane either into the cytoplasm or out .
  • Bacterial ABC exporters have diverse roles, including the extrusion of (often lipid-linked) building blocks required for cell wall assembly [5] [6] [7] .
  • Most are thought to be exporters, and some display a remarkable poly-specificity for substrates.
  • In addition to the fundamental question of how they translocate large substrates across lipid bilayers, a detailed structural and mechanistic understanding of ABC transporters may contribute to drug discovery, for example in the case of multidrug or liver ABC transporters [28] [29] [30] [31] .

Alternating access in ABC importers

  • Type I ABC importers facilitate medium-affinity uptake of diverse nutrients including ions, sugars, amino acids, short peptides, and oligosaccharides, into bacteria.
  • The transport cycle can be thought to start in an inward-facing conformation, with substrate-loaded binding protein approaching the external side of the transporter.
  • This generates a tunnel from the attached binding protein to a low-affinity substrate-binding pocket in the translocation pathway, halfway across the membrane.
  • The key difference to type 1 ABC importers is that in BtuCD-F, ATP binding and hydrolysis does not generate an inward-facing conformation by itself.
  • Upon substrate release, BtuCD relaxes into an asymmetric conformation (state 4) that does not feature a central cavity anymore, because both cyto-gate 1 and the peri-gate are now closed 51 .

Distinct mechanistic proposals for B-family ABC exporters

  • No common mechanism has been established for B-family ABC exporters.
  • Inward-facing conformations without NBD contact or substrate binding pockets are likely unphysiological.
  • Once a substrate is recruited, the transporter probably converts to an occluded conformation (state 1 to 2), with ATP bound between closed NBDs, but without access to the central cavity from either side of the membrane.
  • The mechanistic proposals shown in Fig. 4 are generic and there are multiple variations and complications.
  • EPR studies comparing a homodimeric ABC exporter (MsbA, containing two functional ATPase sites) with a heterodimeric one (BmrAB, containing one functional, one degenerate site) concluded that the presence of a degenerate ATPase site may prevent a wide opening of the NBDs in BmrAB, possibly indicating a fundamental difference between symmetrical vs. asymmetrical transporters 69 .

Mechanisms: Does one size fit all?

  • A number of attempts have been made to formulate generally applicable mechanisms for ABC transporters.
  • These include, among others, the "switch model", the "constant contact model", or the "reciprocating twin-channel model" 38, 75 .
  • Given the structural and biochemical differences discussed above, mechanistic similarities in the entire superfamily appear restricted to the events at the NBDs.
  • Because the different TMDs all feature coupling helices, they can interact with NBDs to harness the conformational power provided by ATP hydrolysis and facilitate import or export in a given ABC transporter.
  • This requires a combination of structural, biochemical, and biophysical (including single molecule spectroscopic 78 ) studies, and ultimately computational analysis, which, despite progress with select systems [79] [80] [81] [82] , has this far struggled with the fact that ABC transporters are very slow enzymes, with turnover rates approaching seconds in some cases.

ABC transporter research: Golden days ahead

  • Significant progress in understanding reaction mechanisms has been made since the first structures of ABC transporters were determined.
  • Architectures and conserved motifs of ABC transporters ABC transporters contain four core domains, two cytoplasmic NBDs and two TMDs, also known as Box 1.
  • While the coupling helix is not the only contact between TMDs and NBDs, it is the only architecturally conserved one among distinct TMD folds and provides the majority of contacts between domains.
  • Furthermore, the exchange of ADP for ATP following a hydrolysis cycle requires an apo-state to be populated, even if only briefly.
  • The two structures are very similar and revealed nucleotide-free, inward-facing conformations with substrates (GSH and GSSG) bound to an inwardfacing cavity at the level of the inner membrane leaflet.

Did you find this useful? Give us your feedback

Figures (4)

Content maybe subject to copyright    Report

ETH Library
Mechanistic diversity in
ATP-binding cassette (ABC)
transporters
Review Article
Author(s):
Locher, Kaspar P.
Publication date:
2016
Permanent link:
https://doi.org/10.3929/ethz-b-000117629
Rights / license:
In Copyright - Non-Commercial Use Permitted
Originally published in:
Nature Structural & Molecular Biology 23(6), https://doi.org/10.1038/nsmb.3216
This page was generated automatically upon download from the ETH Zurich Research Collection.
For more information, please consult the Terms of use.

"""""""""–"
1
Mechanistic diversity in ATP-binding cassette (ABC) transporters
Kaspar P. Locher
Institute of Molecular Biology and Biophysics, Department of Biology, ETH Zurich.
Abstract
ABC transporters catalyze transport reactions ranging from high-affinity uptake of micronutrients into
bacteria to the extrusion of cytotoxic compounds from mammalian cells. Crystal structures of ABC
domains and full transporters have provided a framework for formulating reaction mechanisms of
ATP-driven substrate transport, but recent studies suggest a remarkable mechanistic diversity within
the protein family. This review evaluates the differing mechanistic proposals and outlines future
directions to explore ABC transporter-catalyzed reactions.
Introduction
ABC transporters are a family of membrane proteins that mediate diverse, ATP-driven transport
processes. They contain a pair of conserved, cytoplasmic domains termed ATP-binding cassettes
(ABC) or nucleotide-binding domains (NBD). NBDs hydrolyze ATP and drive conformational changes
in the attached transmembrane domains (TMDs), allowing substrates to cross the lipid bilayer of the
membrane either into the cytoplasm (import) or out (export). The domain arrangement and conserved
sequence motifs are summarized in box 1, and representative structures of the different sub-families
of ABC transporters are shown in Fig. 1.
ABC transporters are found in all domains of life. In bacteria, ABC importers are involved in the
uptake of nutrients and micro-nutrients through medium- and high-affinity pathways
1,2
. These
reactions are highly specific and require a periplasmic (or lipid-anchored external) substrate binding
protein
3,4
. Bacterial ABC exporters have diverse roles, including the extrusion of (often lipid-linked)
building blocks required for cell wall assembly
5-7
. Other ABC exporters are poly-specific and
contribute to drug resistance by exporting certain toxic substances
8,9
.
There are 48 distinct human ABC transporters that belong to various sub-families
10
. Most are thought
to be exporters, and some display a remarkable poly-specificity for substrates. Some human ABC
transporters have functions other than substrate translocation, e.g. in regulating potassium channels
(SUR1 and SUR2)
11,12
or acting as an ATP-gated chloride channel (CFTR)
13
. Many human ABC
transporters are bio-medically and clinically relevant: For example, multidrug transporters such as
ABCB1, ABCG2, or ABCC1 protect various organs from toxic insult, but also contribute to drug
resistance of cancer cells
14-17
; In liver, a series of ABC transporters (ABCB11, ABCB4, ABCG5/G8)
catalyzes the generation of bile, with dysfunction of either transporter leading to liver disease
18-22
; The
transporter of antigenic peptides (TAP, a heterodimer of ABCB2 and ABCB3) has an important role in
the adaptive immune response
23,24
. Archaea, yeasts, plants, and human parasites also have ABC
transporters that catalyze vital functions (reviewed in
25-27
).
In addition to the fundamental question of how they translocate large substrates across lipid bilayers,
a detailed structural and mechanistic understanding of ABC transporters may contribute to drug
discovery, for example in the case of multidrug or liver ABC transporters
28-31
.
It has been established that the closed NBD dimer conformation is essential for ATPase activity of full
ABC transporters, because physical NBD separation results in the transporter inactivation
32-34
. Given
the tight interaction between the NBDs and the coupling helices, it is generally accepted that the
conserved power stroke of ABC transporters is the pushing together and pulling apart of the

"""""""""–"
2
cytoplasmic ends of the TMDs during the ATP hydrolysis cycle (reviewed in
35-38
). However, recent
structural and mechanistic studies challenged the notion that a single, conserved mechanism can
explain the reactions even within individual sub-families of ABC transporters. This review evaluates
transport mechanisms derived from structures of full transporters as well as from functional,
spectroscopic, and biophysical studies.
Alternating access in ABC importers
Type I ABC importers. Type I ABC importers facilitate medium-affinity uptake of diverse nutrients
including ions, sugars, amino acids, short peptides, and oligosaccharides, into bacteria. The E. coli
maltose transporter has been extensively studied (REFS) and can serve as a mechanistic model
of a
generic mechanism of type 1 ABC importers (Fig. 2a). The transport cycle can be thought to start in
an inward-facing conformation, with substrate-loaded binding protein approaching the external side of
the transporter. There is a possibility of futile ATPase activity at this state, resulting in a "basal
ATPase rate". However, this activity is slow in type 1 ABC importers and often additionally inhibited.
Docking of the substrate binding protein stimulates the basal ATPase rate of the maltose
39
and
alginate transporters
40
, but not of the molybdate/tungstate transporters
32,41
. The key conformational
transition during the transport cycle (state 1 to 2) is the closing of the NBDs, which pushes the
coupling helices towards each other and converts the TMDs to an outward-facing conformation. This
generates a tunnel from the attached binding protein to a low-affinity substrate-binding pocket in the
translocation pathway, halfway across the membrane. In the maltose transporter, two effects were
found to ensure the release of substrate from the binding protein: First, the binding pocket is slightly
distorted. Second, periplasmic TMD loops (in particular a "scoop loop") cause a partial steric clash
with bound substrate
42
. Although the affinity of maltose to the temporary binding site between the
TMDs is only moderate (Kd in the millimolar range), the substrate populates this pocket because the
local concentration in the tunnel is at least 2 orders of magnitude above the dissociation constant. In
the subsequent, irreversible step (state 2 to 3) ATP is hydrolyzed and inorganic phosphate released.
This causes the NBD dimer to open, pulling the coupling helices outwards and triggering the
conversion to an inward-facing conformation. The substrate can now dissociate into the cytoplasm.
Finally, the exchange of ADP for ATP resets the system (state 3 to 1).
There are mechanistic variations of the theme in certain type 1 ABC importers. Because of their role
in medium-affinity pathways, efficient coupling of ATP hydrolysis to transport is important, and the
observed basal ATPase activities are generally low. While some transporters such as OpuA have
strict coupling of 2 ATP per transport cycle
43
, additional regulatory strategies have been observed for
other transporters in the form of trans-inhibition
32,33,44
or the binding of regulatory proteins
45
.
Type 2 ABC importers. These are generally part of high-affinity uptake pathways for metal chelates
including heme and other iron-containing complexes, and cobalamin (reviewd in
46,47
). These
substrates are not only larger and more hydrophobic than those of type I ABC importers, but available
at low concentrations only. The mechanism proposed for the E. coli vitamin B12 transporter BtuCD-F
can serve as a model for other type 2 ABC importers. Unlike observed in the maltose transporter,
there is no measurable affinity (and thus no substrate binding pocket) for B12 in the TMDs of BtuCD-
F. The obtained crystal structures revealed three gate regions in the transmembrane BtuC subunits,
two on the cytoplasmic side (cyto-gates 1 and 2) and one on the periplasmic side (peri-gate)
48,49
. The
key difference to type 1 ABC importers is that in BtuCD-F, ATP binding and hydrolysis does not
generate an inward-facing conformation by itself. Rather, the two cytoplasmic gates separate the
cytoplasm from the central translocation cavity both in the ATP-bound and nucleotide-free states, as
long as transport substrate is absent. Figure 3b shows a schematic of the transport mechanism
deduced from structural, biochemical, and spectroscopic data
48-52
. The cycle can be thought to start in

"""""""""–"
3
an ATP-bound state (state 1) with a closed NBD dimer conformation, and coupling helices pushed
together, which causes cyto-gate 2 to close. Because the peri-gate is open, state 1 is an outward-
facing conformation. Futile ATPase activity (arrows to the left) has been observed in all type 2 ABC
importers studied to date, although the measured rates are very low and the loss of cytoplasmic ATP
apparently manageable for the cell. State 2 is reached upon docking of the substrate binding protein
and the release of substrate into a hydrophobic ("teflon") cavity with no measurable affinity. As in type
1 importers, the high-affinity pocket in the substrate binding protein is distorted upon docking, and
loops of the TMD cause a steric clash, "scooping" the substrate from its pocket and enclosing it in the
translocation pathway between the TMDs. In BtuCD, trapping of substrate in this cavity could only be
demonstrated in liposomes. In detergent, the substrate escapes from the cavity, probably due to
increased protein dynamics or competition for binding with detergent molecules. The following,
irreversible step (state 2 to 3) is the hydrolysis of ATP and the release of phosphate, which requires
(at least partial) opening of the NBD dimer, pulling the coupling helices apart and opening cyto-gate 2.
Due to the size of the trapped transport substrate, cyto-gate 1 is now unable to close, which probably
causes a strained conformation and pressure from the sides onto the substrate. This resembles a
peristaltic force and may contribute to substrate dissociation into the cytoplasm. Upon substrate
release, BtuCD relaxes into an asymmetric conformation (state 4) that does not feature a central
cavity anymore, because both cyto-gate 1 and the peri-gate are now closed
51
. This state is very stable
in vitro and probably probably prevents potential ion leakage during the subsequent re-setting of the
system. The asymmetric conformation of state 4 appears unique to type 2 importers because the
structural elements involved are not present in type 1 importers. Notably, the mechanistic proposal
does not assign any role to the BtuCD conformation first observed, an apo-state with outward-facing
TMDs
53
. Although such a conformation may transiently occur in the cell (before ADP is replaced by
ATP) and contribute to futile ATP hydrolysis, it is not essential to formulate a productive transport
cycle. Another important point is that nucleotide-bound BtuCD with a closed NBD dimer could only be
stably trapped and crystallized upon introduction of a disulfide cross-link that covalently linked the D-
loops of the two NBDs.
Distinct mechanistic proposals for B-family ABC exporters
Despite a number of high-resolution structures reported, no common mechanism has been
established for B-family ABC exporters. One problem is that most structures do not reveal bound
substrates, a consequence of their hydrophobicity and low binding affinity in detergent solution.
Another issue is that when removed from the membrane and in the absence of ATP (or ATP-ADP
mixtures reflecting average cytoplasmic concentrations), B-family ABC exporters are prone to
adopting inward-facing conformations with occasionally very large separations of the NBDs. When
bound to nucleotides, the conformations tend to show a smaller separation of the NBDs as well as
occluded or outward-facing conformations. Figure 3 shows a selection of structures of B-family ABC
exporters, demonstrating the wide conformational range.
Early mechanistic proposals stated that alternating access in B-family ABC exporters is reached by
converting the observed, inward-facing conformations to outward-facing states with bound ATP.
However, several crystal structures are inconsistent with this simplified view
54-58
. The relevance of the
observed inward-facing conformations has therefore been controversially discussed (see box 2), and
the discussion is not simply about semantics. The critical issue is whether an observed structure
reflects a functionally relevant state. If it is concluded that a conformation is not physiologically
relevant (as were the inward-facing conformation of the LLO flippase PglK), there would be no point in
attempting to dock a transport substrates into the observed cavities. The two extreme views outlined
in box 2 could be reconciled by viewing B-family ABC exporters as springs, whose architecture works
to push their NBDs apart. In cellular membranes, the combined effects of lipids, ATP, and substrate

"""""""""–"
4
would allow the transporters to reach outward-facing conformations. Inward-facing conformations
might reflect physiological states if they provide substrate binding sites or may occur transiently
because the NBD dimer needs to release the ATP hydrolysis products. Inward-facing conformations
without NBD contact or substrate binding pockets are likely unphysiological.
As was observed for ABC importers, substrates and inhibitors can modulate the ATPase activity of B-
family ABC exporters. The molecular basis of this phenomenon is unknown, but stimulation or
inhibition is often used to identify specific interactions. Two distinct mechanisms of B-family ABC
exporters are shown in Figure 4.
Alternating access. An essential component of this mechanistic proposal (Fig. 4a) is the binding of
transport substrate to an inward-facing pocket of the TMDs (state 1). The relevant binding site may
not only be accessible from the cytoplasm, but also from the inner leaflet of the lipid bilayer. Highly
hydrophobic substrates (drugs, phospholipids) could thus be directly recruited from within the
membrane. The inward-facing conformation is linked to an open NBD conformation. As for ABC
importers, futile ATP hydrolysis may occur in the absence of substrate, albeit at a slower rate than
when substrate is bound. For poly-specific transporters (including the multidrug transporter ABCB1), it
is unclear if there is only one binding pocket
59
, as some findings suggest multiple sites
60,61
. In the case
of TAP, it was concluded that the N- and C-termini of all transported peptides are firmly bound,
whereas the central aminoacids may only weakly interact with the transporter
62
. Once a substrate is
recruited, the transporter probably converts to an occluded conformation (state 1 to 2), with ATP
bound between closed NBDs, but without access to the central cavity from either side of the
membrane. Such a conformation was observed in the structures of AMPPNP-bound McjD
63
and
ATPγS-bound PCAT
57
, albeit without bound substrate. It is unclear if the occluded conformation is a
mandatory intermediate. The following step (state 2 to 3) reflects the conversion to the outward-facing
conformation, allowing substrate release into the external medium or into the outer leaflet of the lipid
bilayer. Given that there is a re-arrangement of the TM helices from states 1 to 3, with different sets of
helices surrounding the translocation pathway and forming the two external wings (Fig. 4), there is a
change in the cavity surface surrounding the substrate. In the AMPPNP-bound, outward-facing
Sav1866 structure, a rather hydrophilic cavity was observed, suggesting that a chemical mismatch to
the hydrophobic drugs could be driving substrate release
64
. This notion was in line with observations
that the apparent affinity of multidrug ABC transporters for substrates was lower from the external
than from the cytoplasmic side
65
. If a substrate is highly hydrophobic (such as in the case of certain
tumor drugs or phospholipids), it is likely to re-partition into the membrane and may only remain in the
external medium when bound to a carrier protein (e.g. BSA) and swept away in the blood stream, or if
stabilized in a mixed micelle such as in bile. In the final step (state 3 to 1), hydrolysis of ATP and the
release of inorganic phosphate drives the NBDs apart and, by transmitting this motion to the TMDs
via the coupling helices, converts the outward-facing into an inward-facing conformation. This step is
irreversible given the large amount of energy gained from the hydrolysis of ATP (approaching 50kJ
mol
-1
for one ATP). It is unclear if the hydrolysis of two ATP molecules is simultaneous or consecutive,
nor is it certain that both ATP molecules are hydrolyzed during every transport cycle (see below).
"
Outward-only mechanism. This mechanism was proposed for the LLO flippase PglK
66
(Fig. 4b). As
its name implies, the outward-only mechanism does not invoke an inward-facing cavity to interact with
the substrate. Although two distinct inward-facing conformations were determined for PglK, the
observed cavities did not appear to contribute to the interaction with the substrate. The cycle of the
outward-only mechanism can be thought to start in an ATP-bound state and an outward-facing TMD
conformation (state 1). Whereas the lipidic polyprenyl tail of the substrate probably remains attached
to the lipid-facing surface of the transporter, the pyrophosphate-glycan head-group is proposed to be
transferred directly into the outward-facing cavity, where strong, electrostatic interactions between

Citations
More filters
Journal ArticleDOI
TL;DR: Recent advances that have increased understanding of the structures and molecular mechanisms of multidrug efflux pumps in bacteria are described, suggesting opportunities for countering their activities.
Abstract: Infections arising from multidrug-resistant pathogenic bacteria are spreading rapidly throughout the world and threaten to become untreatable. The origins of resistance are numerous and complex, but one underlying factor is the capacity of bacteria to rapidly export drugs through the intrinsic activity of efflux pumps. In this Review, we describe recent advances that have increased our understanding of the structures and molecular mechanisms of multidrug efflux pumps in bacteria. Clinical and laboratory data indicate that efflux pumps function not only in the drug extrusion process but also in virulence and the adaptive responses that contribute to antimicrobial resistance during infection. The emerging picture of the structure, function and regulation of efflux pumps suggests opportunities for countering their activities.

463 citations

Journal ArticleDOI
29 May 2017-Nature
TL;DR: The results suggest a multidrug recognition and transport mechanism of ABCG2, rationalize disease-causing single nucleotide polymorphisms and the allosteric inhibition by the 5D3 antibody, and provide the structural basis of cholesterol recognition by other G-subfamily ABC transporters.
Abstract: ABCG2 is a constitutively expressed ATP-binding cassette (ABC) transporter that protects many tissues against xenobiotic molecules Its activity affects the pharmacokinetics of commonly used drugs and limits the delivery of therapeutics into tumour cells, thus contributing to multidrug resistance Here we present the structure of human ABCG2 determined by cryo-electron microscopy, providing the first high-resolution insight into a human multidrug transporter We visualize ABCG2 in complex with two antigen-binding fragments of the human-specific, inhibitory antibody 5D3 that recognizes extracellular loops of the transporter We observe two cholesterol molecules bound in the multidrug-binding pocket that is located in a central, hydrophobic, inward-facing translocation pathway between the transmembrane domains Combined with functional in vitro analyses, our results suggest a multidrug recognition and transport mechanism of ABCG2, rationalize disease-causing single nucleotide polymorphisms and the allosteric inhibition by the 5D3 antibody, and provide the structural basis of cholesterol recognition by other G-subfamily ABC transporters

302 citations

Journal ArticleDOI
01 Dec 2016-Cell
TL;DR: The structure of zebrafish CFTR in the absence of ATP is determined by electron cryo-microscopy to 3.7 Å resolution and reveals why many cystic-fibrosis-causing mutations would lead to defects either in folding, ion conduction, or gating and suggests new avenues for therapeutic intervention.

265 citations


Cites background from "Mechanistic diversity in ATP-bindin..."

  • ...Different structures of ABC exporters captured in the inward-facing conformation by X-ray crystallography and EM display various degrees of separation between the NBDs (Locher, 2016)....

    [...]

Journal ArticleDOI
TL;DR: Overall, a complex picture of lipid–protein interactions emerges, through a range of mechanisms including modulation of the physical properties of the lipid environment, detailed chemical interactions between lipids and proteins, and key functional roles of very specific lipids binding to well-defined binding sites on proteins.
Abstract: Membrane lipids interact with proteins in a variety of ways, ranging from providing a stable membrane environment for proteins to being embedded in to detailed roles in complicated and well-regulated protein functions. Experimental and computational advances are converging in a rapidly expanding research area of lipid–protein interactions. Experimentally, the database of high-resolution membrane protein structures is growing, as are capabilities to identify the complex lipid composition of different membranes, to probe the challenging time and length scales of lipid–protein interactions, and to link lipid–protein interactions to protein function in a variety of proteins. Computationally, more accurate membrane models and more powerful computers now enable a detailed look at lipid–protein interactions and increasing overlap with experimental observations for validation and joint interpretation of simulation and experiment. Here we review papers that use computational approaches to study detailed lipid–prot...

263 citations

Journal ArticleDOI
06 Sep 2017-Nature
TL;DR: This study uncovers the structural basis for LPS recognition, delineates the conformational transitions of MsbA to flip LPS, and paves the way for structural characterization of other lipid flippases.
Abstract: Lipopolysaccharide (LPS) in the outer membrane of Gram-negative bacteria is critical for the assembly of their cell envelopes. LPS synthesized in the cytoplasmic leaflet of the inner membrane is flipped to the periplasmic leaflet by MsbA, an ATP-binding cassette transporter. Despite substantial efforts, the structural mechanisms underlying MsbA-driven LPS flipping remain elusive. Here we use single-particle cryo-electron microscopy to elucidate the structures of lipid-nanodisc-embedded MsbA in three functional states. The 4.2 A-resolution structure of the transmembrane domains of nucleotide-free MsbA reveals that LPS binds deep inside MsbA at the height of the periplasmic leaflet, establishing extensive hydrophilic and hydrophobic interactions with MsbA. Two sub-nanometre-resolution structures of MsbA with ADP-vanadate and ADP reveal an unprecedented closed and an inward-facing conformation, respectively. Our study uncovers the structural basis for LPS recognition, delineates the conformational transitions of MsbA to flip LPS, and paves the way for structural characterization of other lipid flippases. Cryo-electron microscopy snapshots of the E. coli flippase MsbA at discrete functional states reveal a ‘trap and flip’ mechanism for lipopolysaccharide flipping and the conformational transitions of MsbA during its substrate transport cycle. The translocation and flipping of lipids across membrane bilayers is important for maintaining lipid asymmetry, but it also plays a part in membrane trafficking and signalling. This process is catalysed by flippases, bacterial ATP-binding cassette transporters that flip lipopolysaccharide (LPS). Because of the importance of LPS transport to bacterial survival, these transporters are also a target for the development of antibiotics. Here, Maofu Liao and colleagues present the structural basis of LPS recognition by the Escherichia coli flippase MsbA and reveal the trajectory of LPS. Using cryo-electron microscopy the authors provide snapshots of several of the states involved in lipid translocation across the bacterial membrane. In one of these states, they detected electron density within the transmembrane domain of MsbA corresponding to LPS. On the basis of these insights, the researchers propose a 'trap and flip' mechanism in which LPS translocates far into the transporter without flipping.

200 citations

References
More filters
Journal ArticleDOI
TL;DR: The ability to predict and circumvent drug resistance is likely to improve chemotherapy, and it has become apparent that resistance exists against every effective drug, even the authors' newest agents.
Abstract: Chemotherapeutics are the most effective treatment for metastatic tumours. However, the ability of cancer cells to become simultaneously resistant to different drugs--a trait known as multidrug resistance--remains a significant impediment to successful chemotherapy. Three decades of multidrug-resistance research have identified a myriad of ways in which cancer cells can elude chemotherapy, and it has become apparent that resistance exists against every effective drug, even our newest agents. Therefore, the ability to predict and circumvent drug resistance is likely to improve chemotherapy.

5,105 citations

Journal ArticleDOI
14 Sep 2006-Nature
TL;DR: The observed, outward-facing conformation reflects the ATP-bound state, with the two nucleotide-binding domains in close contact and the two transmembrane domains forming a central cavity—presumably the drug translocation pathway—that is shielded from the inner leaflet of the lipid bilayer and from the cytoplasm, but exposed to the outer leaflet and the extracellular space.
Abstract: Multidrug transporters of the ABC family facilitate the export of diverse cytotoxic drugs across cell membranes. This is clinically relevant, as tumour cells may become resistant to agents used in chemotherapy. To understand the molecular basis of this process, we have determined the 3.0 A crystal structure of a bacterial ABC transporter (Sav1866) from Staphylococcus aureus. The homodimeric protein consists of 12 transmembrane helices in an arrangement that is consistent with cross-linking studies and electron microscopic imaging of the human multidrug resistance protein MDR1, but critically different from that reported for the bacterial lipid flippase MsbA. The observed, outward-facing conformation reflects the ATP-bound state, with the two nucleotide-binding domains in close contact and the two transmembrane domains forming a central cavity—presumably the drug translocation pathway—that is shielded from the inner leaflet of the lipid bilayer and from the cytoplasm, but exposed to the outer leaflet and the extracellular space. Multidrug efflux transporters cause serious problems in cancer chemotherapy and in the treatment of bacterial infections. A puzzling aspect of their biology is how a single transporter can recognize and transport such a wide variety of structurally dissimilar compounds. The publication of the crystal structures of two quite different multidrug efflux transporters will help to solve the mystery. In the first study, the structure of AcrB — a multidrug efflux transporter from E. coli — was determined. Its three constituent subunits were captured at different steps in the transport cycle: prior to substrate binding, substrate-bound, and post-extrusion. The voluminous multidrug binding pocket handles multiple substrates via multi-site binding. The second study determined the structure of an ATP-driven multidrug transporter from S. aureus. The clinical relevance of this 'ABC' family of transporters derives from the fact that they catalyse the extrusion of various cytotoxic compounds used in cancer therapy. The structure, with the transporter in the outward-facing conformation, is a useful model of human homologues and may initiate the rational design of drugs aimed at interfering with the extrusion of agents used in chemotherapy.

1,244 citations

Journal ArticleDOI
TL;DR: The availability of an increasing number of high-resolution structures has provided a valuable framework for interpretation of recent studies, and realistic models have been proposed to explain how these fascinating molecular machines use complex dynamic processes to fulfill their numerous biological functions.
Abstract: Summary: ATP-binding cassette (ABC) systems are universally distributed among living organisms and function in many different aspects of bacterial physiology. ABC transporters are best known for their role in the import of essential nutrients and the export of toxic molecules, but they can also mediate the transport of many other physiological substrates. In a classical transport reaction, two highly conserved ATP-binding domains or subunits couple the binding/hydrolysis of ATP to the translocation of particular substrates across the membrane, through interactions with membrane-spanning domains of the transporter. Variations on this basic theme involve soluble ABC ATP-binding proteins that couple ATP hydrolysis to nontransport processes, such as DNA repair and gene expression regulation. Insights into the structure, function, and mechanism of action of bacterial ABC proteins are reported, based on phylogenetic comparisons as well as classic biochemical and genetic approaches. The availability of an increasing number of high-resolution structures has provided a valuable framework for interpretation of recent studies, and realistic models have been proposed to explain how these fascinating molecular machines use complex dynamic processes to fulfill their numerous biological functions. These advances are also important for elucidating the mechanism of action of eukaryotic ABC proteins, because functional defects in many of them are responsible for severe human inherited diseases.

1,194 citations

Journal ArticleDOI
TL;DR: Diverse covalent modifications of the lipid A moiety may occur during its transit from the outer surface of the inner membrane to the outer membrane of most gram-negative bacteria.
Abstract: The lipid A moiety of lipopolysaccharide forms the outer monolayer of the outer membrane of most gram-negative bacteria. Escherichia coli lipid A is synthesized on the cytoplasmic surface of the inner membrane by a conserved pathway of nine constitutive enzymes. Following attachment of the core oligosaccharide, nascent core-lipid A is flipped to the outer surface of the inner membrane by the ABC transporter MsbA, where the O-antigen polymer is attached. Diverse covalent modifications of the lipid A moiety may occur during its transit from the outer surface of the inner membrane to the outer membrane. Lipid A modification enzymes are reporters for lipopolysaccharide trafficking within the bacterial envelope. Modification systems are variable and often regulated by environmental conditions. Although not required for growth, the modification enzymes modulate virulence of some gram-negative pathogens. Heterologous expression of lipid A modification enzymes may enable the development of new vaccines.

1,148 citations

Journal ArticleDOI
TL;DR: An important goal is to develop quantitative models that detail the kinetic and molecular mechanisms by which ABC transporters couple the binding and hydrolysis of ATP to substrate translocation.
Abstract: ATP-binding cassette (ABC) transporters constitute a ubiquitous superfamily of integral membrane proteins that are responsible for the ATP-powered translocation of many substrates across membranes. The highly conserved ABC domains of ABC transporters provide the nucleotide-dependent engine that drives transport. By contrast, the transmembrane domains that create the translocation pathway are more variable. Recent structural advances with prokaryotic ABC transporters have provided a qualitative molecular framework for deciphering the transport cycle. An important goal is to develop quantitative models that detail the kinetic and molecular mechanisms by which ABC transporters couple the binding and hydrolysis of ATP to substrate translocation.

1,087 citations