Integral membrane proteins are involved in a plethora of biological processes including cellular signalling, molecular transport, and catalysis. Many of these functions are mediated by non-covalent interactions with other proteins, substrates, metabolites, and surrounding lipids. Uncovering such interactions and deciphering their effect on protein activity is essential for understanding the regulatory mechanisms underlying integral membrane protein function. However, the detection of such dynamic complexes has proven to be challenging using traditional approaches in structural biology. Native mass spectrometry has emerged as a powerful technique for the structural characterisation of membrane proteins and their complexes, enabling the detection and identification of protein-binding partners. In this review, we discuss recent native mass spectrometry-based studies that have characterised non-covalent interactions of membrane proteins in the presence of detergents or membrane mimetics. We additionally highlight recent progress towards the study of membrane proteins within native membranes and provide our perspective on how these could be combined with recent developments in instrumentation to investigate increasingly complex biomolecular systems.

Integral membrane proteins comprise approximately one-third of the human proteome [1] and undertake key roles in signal and energy transduction, cell adhesion, molecular transport, and enzymatic catalysis [2]. Such functions are carefully regulated by post-translational modifications and biomolecular interactions that fine-tune protein activity in response to internal and external stimuli. When these mechanisms fail, the subsequent dysregulation of membrane protein function can induce a wide range of pathological conditions including cardiovascular, central nervous system, and immune disorders. Accordingly, certain classes of membrane proteins (e.g. G-protein coupled receptors [GPCRs] and ion channels) have been extensively employed as biological targets within drug discovery programs investigating such indications [3–5].

Understanding how endogenous binding partners and drugs impact the structures and function of membrane proteins is a key step towards elucidating protein structure–function relationships. Advances in experimental and computational techniques have enabled access to hitherto elusive knowledge of membrane proteins’ working mechanisms and their interactions with other biomolecules. These methodological advancements range from developments in membrane protein crystallisation [6] and solubilisation methods [7] to high-resolution cryo-electron microscopy [8] and tools for predicting protein structure [9,10]. In parallel, native mass spectrometry (native MS) has emerged as a powerful technique that can provide unique insights [11–16]. Native MS analysis of purified membrane proteins provides direct information regarding purity, folding, modifications, oligomeric state, and sub-unit interactions in homo- or heterooligomers. Owing to the possibility of monitoring protein-ligand interactions with molecular-level precision, native MS has developed into a valuable tool for high-throughput drug screening platforms [17,18].

By integrating native MS alongside other structural techniques, deep insights into the relationship between protein sequence, structure, dynamics, and function can be derived. The ease of coupling native MS with ion-mobility and hydrogen-deuterium exchange MS enables access to information regarding protein folding, conformation, and dynamics [19–23]. Similarly, combining native MS directly with cryo-EM via selective deposition using electrospray ionisation offers additional benefits of increasing the scope and throughput of structural determination [24–26]. Native MS can serve as a preliminary tool to screen for the best protein homologues and optimal purification conditions for subsequent interrogation by other structural techniques [27,28]. Additionally, native MS enables the detection of lipids bound to membrane proteins [29,30] – offering valuable insights into the critical role of lipids in membrane protein structures and functions.

There has been a multitude of excellent, in-depth reviews focused on the study of membrane protein structure by native MS [15,31–33]. Herein we focus on recent studies investigating interactions between membrane proteins and endogenous ligands captured throughout the purification process and the dynamics of membrane proteins in signalling pathways observed directly in the native membrane. Finally, we will draw attention to the native top-down MS approach that combines native MS with top-down proteomics in a single experiment and give our perspective on how native top-down can be used to study complex signalling pathways in native membranes.

A fundamental challenge associated with handling membrane proteins outside of their cellular context is to maintain their native fold in aqueous buffers. This is often achieved using detergents, which mimic the phospholipid bilayer [34,35]. Detergents can drastically impact the structure and function of solubilised membrane proteins [36], and an optimal detergent for a biophysical technique must be determined on a case-by-case basis [37]. Fortunately, there exists a wide range of detergents with varying properties to select from, with both commercial and specialist detergents offering assorted head groups and chain chemistries. Many detergents typically used to maintain protein stability and function are not always suitable for native MS measurements. For example, DDM (n-dodecyl β-D-maltoside) and LMNG (lauryl maltose neopentyl glycol) are often considered the preferred detergents for protein extraction and purification from cellular membranes as they create a stable environment for proteins [38]. However, these surfactants form relatively larger micelles which can only be removed from the protein in the gas phase using extensive collisional activation [39,40]. Therefore, when using DDM or LMNG in native MS measurements, it can be difficult to liberate the protein from the micelle without inducing ligand and/or subunit dissociation. By contrast, native MS-friendly detergents such as the charge-reducing C8E4 and LDAO, considered to provide a harsher environment for proteins, can be removed at lower collisional energies, assisting in the preservation of non-covalent interactions between membrane proteins and their binding partners. However, such detergents may be less effective at maintaining the protein’s fold in solution. There is a delicate balance between activation energies and preserving interactions of membrane proteins in native MS analyses, so care must be taken in selecting the detergents for MS analyses [41,42]. Table 1 exemplifies the variety of detergents used for protein solubilisation and subsequent native MS analyses over the past 3 years. We hope that a future database presenting ‘reference’ mass spectra of membrane proteins characterised to date, along with suitable experimental conditions and instrument settings can guide the community in selecting optimal detergents suitable for new protein targets.

Table 1
Selected list of membrane proteins purified in various detergents and analysed by native MS in recent years (2020–2022)
Membrane proteinDetergent used for purificationDetergent used for native MS analysisReferences
2022 
Kirbac3.1 DDM C8E4 [79
Aqp0  C8E4 [12
Aqp0 OG C8E4 [56
OmpF, FepA, BtuB OG OG [65
TqsA DDM LDAO, C8E4 [80
FhaC OG OG [81
GCGR DDM, CHS OGDs G1 [82
Glut5
PfHT1
GlpG
NTR1 
DDM
DDM
DDM
LMNG 
DDM
DDM
DDM, LDAO
DDM, NM, DDTM, LDAO, C8E4, C12E8, LMNG, OGNG, Fos-choline-12, -14, -16, HEGA-11, MEGA-10, sucrose monodecanoate 
[83
NHA2 DDM, CHS C12E9 [84
β1AR DDM DDM/Foscholine16/cholesteryl hemisuccinate [85
MraY
MurJ 
DDM DDM, OG, C8E4, LDAO, LDAO [48
NIP2;1 DDM, DM LDAO [86
PA2880 DDM DDM [87
PatAB DDM OGD G1 [88
PMP22 Empigen BB, DM C12E9 [59
Rho and opsin LMNG LMNG [54
SthK DDM OGNG [89
TRAAK
TREK2 
C10E5 C10E5 [90
2021 
A2aR
GCGR 
LMNG
G1, CHS 
LMNG
G1/CHS 
[72
AKCR1 LMNG, CHA DDM [91
BAM complex DDM C8E4 [68
bR, AR3 OG OG [30
Kir3.4
Kir3.2 
C10E5 C10E5 [92
LptDE DDM C8E4 [67
MFSD2A DDM, CHS DDM [93
RAGE Triton X-100 Triton X-100 [94
Rhodopsin CHAPS C8E4 [95
SusCD LDAO C8E4 [74
TmrAB DDM DDM [96
TolC OG OG [97
tOmpA, OmpAAG, OmpTrans2, OmpTrans3 DDM DDM [98
Wzc LMNG C8E4, LDAO [57
2020 
AceI DDM LDAO, C8E4, DDM, OGNG [66
AcrB DDM DDM, Triton X-100 [99
AmtB DDM OGD [100
AqpZ
MATE
AmtB
BAM
NTSR1
OmpT 
DDM and OGDs OGDs [43
CD9, CD81 DDM DDM [101
Cytochrome bc1 complex GDN GDN [102
DGAT1   [103
EmbB DDM C12E8, DDM [104
FapF, FapD C8E4 C8E4 [76
FptA OG OG [105
Get1/2 C12E9, Cymal-5/6, DM, UDM, DDM, LMNG, Cymal-5-NG UDM, LMNG, C12E9, Cymal-5-NG [106
HAS DM DM [107
K2p4.1 C10E5 C10E5 [108
Lci1 OG LDAO [109
NHE9 DDM DDM [110
OmpF OG OG [111
PSH
LeuT
MurJ 
NG, LDAO
DDM
DDM 
NG, LDAO
OG
LDAO 
[66
Semisweet
DHODH
CB1
Beta-1 AR
OmpF
AqpZ
AmtB
TSPO 
DDM
LDAO
LMNG
DDM
OG
DDM
DDM
DDM 
C8E4
LDAO
DDM, foscholine, CHS
DDM, foscholine, CHS
C8E4
C8E4
C8E4
C8E4 
[78
V-ATPase DDM LMNG [60
Membrane proteinDetergent used for purificationDetergent used for native MS analysisReferences
2022 
Kirbac3.1 DDM C8E4 [79
Aqp0  C8E4 [12
Aqp0 OG C8E4 [56
OmpF, FepA, BtuB OG OG [65
TqsA DDM LDAO, C8E4 [80
FhaC OG OG [81
GCGR DDM, CHS OGDs G1 [82
Glut5
PfHT1
GlpG
NTR1 
DDM
DDM
DDM
LMNG 
DDM
DDM
DDM, LDAO
DDM, NM, DDTM, LDAO, C8E4, C12E8, LMNG, OGNG, Fos-choline-12, -14, -16, HEGA-11, MEGA-10, sucrose monodecanoate 
[83
NHA2 DDM, CHS C12E9 [84
β1AR DDM DDM/Foscholine16/cholesteryl hemisuccinate [85
MraY
MurJ 
DDM DDM, OG, C8E4, LDAO, LDAO [48
NIP2;1 DDM, DM LDAO [86
PA2880 DDM DDM [87
PatAB DDM OGD G1 [88
PMP22 Empigen BB, DM C12E9 [59
Rho and opsin LMNG LMNG [54
SthK DDM OGNG [89
TRAAK
TREK2 
C10E5 C10E5 [90
2021 
A2aR
GCGR 
LMNG
G1, CHS 
LMNG
G1/CHS 
[72
AKCR1 LMNG, CHA DDM [91
BAM complex DDM C8E4 [68
bR, AR3 OG OG [30
Kir3.4
Kir3.2 
C10E5 C10E5 [92
LptDE DDM C8E4 [67
MFSD2A DDM, CHS DDM [93
RAGE Triton X-100 Triton X-100 [94
Rhodopsin CHAPS C8E4 [95
SusCD LDAO C8E4 [74
TmrAB DDM DDM [96
TolC OG OG [97
tOmpA, OmpAAG, OmpTrans2, OmpTrans3 DDM DDM [98
Wzc LMNG C8E4, LDAO [57
2020 
AceI DDM LDAO, C8E4, DDM, OGNG [66
AcrB DDM DDM, Triton X-100 [99
AmtB DDM OGD [100
AqpZ
MATE
AmtB
BAM
NTSR1
OmpT 
DDM and OGDs OGDs [43
CD9, CD81 DDM DDM [101
Cytochrome bc1 complex GDN GDN [102
DGAT1   [103
EmbB DDM C12E8, DDM [104
FapF, FapD C8E4 C8E4 [76
FptA OG OG [105
Get1/2 C12E9, Cymal-5/6, DM, UDM, DDM, LMNG, Cymal-5-NG UDM, LMNG, C12E9, Cymal-5-NG [106
HAS DM DM [107
K2p4.1 C10E5 C10E5 [108
Lci1 OG LDAO [109
NHE9 DDM DDM [110
OmpF OG OG [111
PSH
LeuT
MurJ 
NG, LDAO
DDM
DDM 
NG, LDAO
OG
LDAO 
[66
Semisweet
DHODH
CB1
Beta-1 AR
OmpF
AqpZ
AmtB
TSPO 
DDM
LDAO
LMNG
DDM
OG
DDM
DDM
DDM 
C8E4
LDAO
DDM, foscholine, CHS
DDM, foscholine, CHS
C8E4
C8E4
C8E4
C8E4 
[78
V-ATPase DDM LMNG [60

Excitingly, new detergents with modified headgroups, linkers, and chain configurations continue to emerge. A notable example in this regard is the development of a ‘tailored’ OGD-based detergent that combines the membrane solubilisation and protein extraction efficacy of DDM with the ease of protein release from C8E4 micelles [43]. Synthesis of such hybrid detergents containing two different headgroups [44] can foster the creation of detergents with predictable biophysical properties, paving the way for an ideal detergent tailored towards a given membrane protein target and downstream analysis technique. Nevertheless, an optimal detergent for solubilising and stabilising a given membrane protein target is currently difficult to predict.

Detergent-purified proteins can also be reconstituted into other membrane mimetics, such as bicelles, amphiphilic polymers, liposomes and nanodiscs, for native MS analyses [45,46]. However, the initial exposure to detergents may impact the observed oligomeric state of the protein, as well as strip away structurally or functionally important endogenous lipids and ligands. In those cases, adopting a completely ‘detergent-free’ strategy is desirable. To this end, membrane proteins can be directly solubilised by synthetic polymers, including SMA and DIBMA, forming the so-called native nanodiscs [47,48]. This detergent-free strategy is highly appealing since the protein is maintained in a lipid bilayer composed of its own native lipids. However, releasing intact protein complexes from SMALPs is nontrivial, potentially due to low charge acquisition by proteins embedded in the nanodiscs [49], the heterogeneity of the system, and strong interactions between the protein–lipid complex and the scaffold polymer. Over the past decade, significant efforts have been dedicated to optimising the protocols to eject samples from these membrane mimetics and in several instances, it was possible to capture protein-lipid interactions at even lower activation energies than those used to remove detergent micelles [33]. Additionally, selection of the appropriate membrane mimetic can have a significant impact on the observed results; for example, the proportion of PMP22 dimer was shown to be different in detergent micelles or when reconstituted into SCOR bicelles that better mimic the cholesterol and sphingolipid-rich native membrane environment (PMP22 is further discussed in a later section) [50]. Nanodiscs, that consist of a lipid bilayer surrounded by membrane scaffolding protein, have also been gaining popularity recently. Using nanodiscs with a 50:50 distribution of two different lipids allowed the investigation of lipid selectivity for two membrane proteins, AmtB and AqpZ [51]. AqpZ displayed the preference for PG over PE, and PC over either lipid; while AmtB was shown to be broadly enriched in bound PG, with a few tightly bound PC molecules. More complex mixed lipid nanodiscs that mimic various biological membranes were also recently investigated [52]. In combination with methodological improvements, such as controlling the stability of membrane protein nanodiscs by adding charge manipulation reagents [53], these developments could lead to even more widespread use of nanodiscs in native MS in the near future. Nevertheless, the possibility of scrutinising membrane proteins directly solubilised and purified in the complete absence of traditional detergents continues to gain interest. For example, a recent study demonstrates that SMA copolymer can selectively solubilise and maintain bacteriorhodopsin and archeerrhodopsin-3 in a correctly folded conformation, unlike detergents [30]. Further method developments, such as supercharging or new activation methods, can further improve the analysis of SMALP-solubilised membrane proteins. An exciting detergent-free method has recently emerged that involves the ejection of lipid vesicles derived directly from native membranes [54,55]; this is discussed in more detail in a later section.

Although the use of native MS in probing the interactions of membrane proteins has been well described in recent reviews [32,33], here we highlight the recent observations made in the past 3 years.

Covalent modifications

While native MS (not coupled with doing multiple rounds of MS/MS), does not provide localisation information on covalent modifications to proteins, it can still play a role in the characterisation of individual proteoforms. Such measurements can enable the effects of PTMs on protein interactions to be directly identified. For example, Harvey et al. [56] employed native MS to study post-translational modifications of AQP0, the most abundant membrane protein in the eye lens. By performing surface-induced dissociation on doubly phosphorylated tetramers, they discovered that these forms contain exclusively singly phosphorylated monomers, despite the presence of two phosphorylation sites on each monomer. In a different study, native MS was used to monitor the number of phosphorylations on Wzc, a protein involved in EPS regulation in Gram-negative bacteria, to complement high-resolution structural studies [57]. The non-phosphorylated form of the protein assembled into octamers while the multiply phosphorylated form was monomeric, indicating a potential regulatory mechanism for glycan polymerisation and translocation.

On the other hand, coupling native MS with tandem MS (MSn), known as native top-down MS, can enable the identification and characterisation of specific proteoforms and post-translational modifications. In an elegant study, Ro et al. [58] recently coupled nanodiscs with native top-down MS and gained valuable insights into the active form of particulate methane monooxygenase, pMMO by determining the stoichiometry of copper ions bound to each subunit and detecting post-translational modifications.

Non-covalent interactions

Protein–protein

A more common application of native MS is in the investigation of non-covalent biomolecular interactions, such as protein–protein interactions. Fantin et al. [59] compared the oligomerisation of PMP22 between the wild-type protein and disease-associated mutants and detected decreased proportions of dimer for mild and moderate disease phenotypes but enhanced dimerisation for the severe disease mutants. By employing ion-mobility MS, the authors observed greater gas-phase stability of dimers but lower stability of monomers of all mutants compared with the wild-type protein with respect to unfolding. This led to a proposed pathophysiological mechanism in which disease-associated mutants are more likely to aggregate, resulting in extensive mistrafficking. As another example, native MS has been extremely useful for confirming the subunit composition of the V1 region of the mammalian V-ATPase [60].

Protein–lipid

Lipids can influence the structure and function of membrane proteins. For example, while some oligomeric membrane proteins contain strongly bound inter-subunit interfaces, other weakly oligomerise and readily dissociate into monomers or lower oligomeric forms upon extraction from their native lipid environment. It has been suggested [61] that in these cases, specific lipid interactions may be required for oligomeric assembly in the phospholipid bilayer. One approach to elucidate the effect of lipids on membrane proteins is to titrate a detergent-purified, fully delipidated sample with the most abundant lipids in their native membrane and quantify the relative binding affinities [62]. The specificity of bound ligands can then be ascertained by systematic titration with a delipidating detergent such as NG as demonstrated for the lipid II flippase MurJ [41]. In another study, MraY dimer, the first integral membrane enzyme in the biosynthesis of bacterial cell wall peptidoglycan, was found to be stabilised by endogenous lipids [63]. Further investigation by MS/MS and functional assays revealed the identity of these ligands as substrate and product of MraY-catalysed reaction (Figure 1). Furthermore, Patil et al. [64] found densities at the cavity formed by the transmembrane helices of human GPR158. PE and PI lipids were identified by native MS and lipidomics and remodelled into the EM structure with high confidence and their roles were ascribed to stabilising the dimer interface. More recently, native MS combined with chemical crosslinking was used to probe the organisation of bacterial outer membrane proteins and lipids [65]. The pattern of lipid crosslinks captured by native MS amongst various OmpF mutants reflects that the outermost leaflet of the outer membrane is enriched with lipopolysaccharides while the inner leaflets are mainly composed of phospholipids.

Native mass spectrometry of purified MraY
Figure 1
Native mass spectrometry of purified MraY

(A) Mass spectrum of MraY released from OG (octylglucoside) detergent micelles. Peaks in the spectrum correspond to the MraY monomer, the ligand-free dimer, and the dimer bound to undecaprenyl phosphate (C55P), lipid I and lipid II. (B and C) Dimer-bound to C55P (B) and lipid I (C) were separately isolated and subjected to collision-induced dissociation (CID) to liberate MraY protomers. (D) Schematic depiction of a ligand stabilised MraY dimer in detergent micelles. Removal of bound ligand caused the dimer to dissociate. Data reproduced from [63].

Figure 1
Native mass spectrometry of purified MraY

(A) Mass spectrum of MraY released from OG (octylglucoside) detergent micelles. Peaks in the spectrum correspond to the MraY monomer, the ligand-free dimer, and the dimer bound to undecaprenyl phosphate (C55P), lipid I and lipid II. (B and C) Dimer-bound to C55P (B) and lipid I (C) were separately isolated and subjected to collision-induced dissociation (CID) to liberate MraY protomers. (D) Schematic depiction of a ligand stabilised MraY dimer in detergent micelles. Removal of bound ligand caused the dimer to dissociate. Data reproduced from [63].

Close modal

Protein–drug/antibiotics

Another interesting area of research is the interaction of membrane proteins with drugs or antibiotics. Using native MS, the antibiotic chlorhexidine was shown to induce a change in the oligomeric state of the chlorhexidine efflux pump AceI [66]. Chlorhexidine was also shown to cause an increase in the formation of the functional dimeric form of AceI. In 2021, Fiorentino et al. [67] investigated the mechanism by which an antibacterial peptide thanatin impairs LPS translocation through LptDE complex. Native MS confirmed the simultaneous binding of thanatin and LPS to LptDE, ruling out inhibition by competitive binding. This result, in combination with hydrogen-deuterium exchange MS and molecular dynamics simulations data, suggested a mechanism whereby thanatin obstructs LPS transport by stabilising the inactive form of LptDE. In another study, a combination of native MS and MD simulations enabled the elucidation of the lipid-binding preference of the Bam complex [68]. The study revealed that the Bam complex preferentially binds cardiolipin and that the latter enhances its interaction with darobactin, a peptidomimetic antibiotic.

Protein–metal ion

One type of interaction that is challenging to observe by native MS is the binding of proteins to non-volatile salt ions, such as Na+. Such ions have been previously shown to regulate the assembly of several membrane protein complexes [69]. In native MS, the presence of even relatively low concentrations of salts results in signal suppression, loss of resolution and spectra that are often too difficult to interpret. Consequently, native MS typically employs volatile salts, such as ammonium acetate, that are readily removed upon gentle activation [70]. Nevertheless, there are instances where metal ions are necessary cofactors for protein stability and enzymatic reactions. One way to study the effect of non-volatile salts on protein structure and stability is to use emitters with submicron tip diameters that form smaller electrospray droplets and thus greatly reduce the number of observed protein–salt adducts [71]. This approach was recently used by [72] to study the effects of sodium ions on two GPCRs: a Class A GPCR, A2aR, and a Class B GPCR, GCGR. For A2aR, native MS revealed enhanced Na+ binding in the presence of antagonists and suppressed Na+ binding in the presence of agonists. This result is consistent with previous structural studies that showed the presence of an allosteric Na+ binding pocket in inactive conformations of Class A GPCRs, which is absent in the active conformations. For GCGR, a more stable conformation was observed in NaCl/Tris buffer compared to ammonium acetate, as indicated by the lower charge state distribution and enhanced binding of the stabilising negative allosteric modulator, NNC0666, observed in the former buffer. These results highlight the applicability of native MS to study membrane proteins from physiologically relevant salt buffers. More recently, another native MS approach was developed to interrogate the propensity of β1-adrenergic receptor coupling to Gs and Gi. Upon capturing the G-protein-coupling complexes in presence of different ligands, it was observed that endogenous zinc ions specifically stabilise the complexes between G-proteins and receptors. Overall, this study uncovered a novel role for metal ions in modulating the selectivity of G-protein coupling [73].

Protein–ligand/Enzyme–substrate

To probe the interactions with ligands, in one study Gray et al. [74] combined native MS analysis with other structural tools to identify the finite size of the substrate molecules (approximately 2.5 kDa) that are imported by the SusCD complex, a major fructooligosaccharides uptake system in Bacteroidetes. Native MS was also used to study the interactions of membrane proteins, involved in peptidoglycan biosynthesis pathway, with lipid-like substrates. The key step in the synthesis of peptidoglycan is the translocation of lipid II from the inner to the outer leaflet of the plasma membrane. The flippase MurJ is thought to play a dominant role over FtsW in this process. A previous native MS study revealed a stronger lipid II binding preference for MurJ over FtsW, and that the lipid II binding to MurJ is reduced in the presence of cardiolipins, which is suggestive of lipid-regulated precursor translocation [75]. Similarly, an increase in the binding affinities of MraY towards C55P, MurG towards lipid I, and MurJ towards lipid II were observed using native MS recently, suggesting that the affinity of each membrane enzyme for its native substrate promotes their trajectory along the peptidoglycan pathway [63].

When using even the most ‘native-like’ membrane mimetic, there is always a risk of experimental artefacts caused by the extraction of the protein from its native membrane environment. Recently, membrane proteins and their complexes have been released into the mass spectrometer directly from their native membrane, marking a significant shift towards native MS of membrane proteins in the most native-like contexts. In 2018, Chorev et al. [55] used native MS to investigate protein complexes directly from native membrane vesicles. This technique named sonicated lipid vesicles MS (SoLVe MS) was successful in revealing a range of subunit interactions and lipid preferences for endogenous proteins that are abundant in the membrane. Several of the observed complexes were not previously reported, highlighting the potential of SoLVe MS. For example, the Bam complex with either a second BamE subunit or up to three cardiolipin molecules was observed, providing more structural insight about this complex. The interactions between SecYEG translocon and an F-type ATPase were also observed directly.

In a very recent study, Chen et al. [54] ejected retinal rod segments (ROSs) directly from the native bovine disc membrane into the mass spectrometer under controlled light conditions to uncover signalling events of a GPCR (rhodopsin) in the context of its own native membrane (Figure 2). Crucially, all components of the signalling pathway were observed in real-time and without any extensive sample pre-treatments. The study found that one of the key steps in the pathway, the hydrolysis of retinal, was significantly slower in native membranes compared to LMNG detergent micelle. In addition, the presence of PE lipids in ROS membranes was shown to be important for the regeneration of rho species following the original activation by light. Unsaturated PC lipids were observed to be enriched around rho shortly after light activation, shedding light on the role of lipid microenvironments for signalling facilitation. The ability to observe all the steps of the signalling pathway directly from the native membranes enabled kinetic studies in the presence of exogenously added downstream effector molecules, highlighting the potential application of this methodology in drug discovery workflows.

Rhodopsin signalling cascade in the native membrane captured by mass spectrometry

Figure 2
Rhodopsin signalling cascade in the native membrane captured by mass spectrometry

(A–C) Disc membranes of rod cells (A) are homogenised to form a heterogeneous distribution of vesicles (B) that are introduced directly into the mass spectrometer (C), a UHMR equipped with an LED light source. (D,E) Changes in the population of rho and opsin in LMNG (blue and green, respectively) and in the native membrane vesicles (red and yellow, respectively; Individual spectra are shown as zero-charge plots with illumination times stated. (F) Relative abundance of rho as a function of illumination time in LMNG micelles (blue) or native membranes (red). Figures reproduced from [54].

Figure 2
Rhodopsin signalling cascade in the native membrane captured by mass spectrometry

(A–C) Disc membranes of rod cells (A) are homogenised to form a heterogeneous distribution of vesicles (B) that are introduced directly into the mass spectrometer (C), a UHMR equipped with an LED light source. (D,E) Changes in the population of rho and opsin in LMNG (blue and green, respectively) and in the native membrane vesicles (red and yellow, respectively; Individual spectra are shown as zero-charge plots with illumination times stated. (F) Relative abundance of rho as a function of illumination time in LMNG micelles (blue) or native membranes (red). Figures reproduced from [54].

Close modal

Despite the numerous advantages and a recently published protocol [76], the practical application of SoLVe MS remains challenging. The main obstacle is that the identities of various complexes observed in the spectrum are difficult to achieve through mass alone. Various other techniques exist that can be combined with native MS to further characterise species observed in SoLVe MS spectra. Native top-down mass spectrometry employing high energy collision- (CID HCD), electron- (ECD, ETD and EID) and photon- (IRMPD and UVPD) based activation techniques can be used to break covalent bonds (while preserving non-covalent interactions in some cases) to obtain information about protein sequence and identify proteoforms. Recent advances in this field have been outlined in a recent review [31]. In addition, co-purified lipids and other small molecules often need to be identified. A traditional approach to this problem is to perform lipidomics on the co-purified lipids in a separate experiment to native MS [77]; however, while this method can be very informative, the direct protein–lipid binding information is lost during the extraction step. The recent advances in MS instrumentation enabled the development of a hybrid method termed ‘nativeomics’ (also known as native top-down), this allows observation and identification of protein-bound lipids and ligands in a single experiment [78]. For instance, the application of this workflow to an outer membrane protein TSPO allowed previously unidentified electron density in the crystal structure of this protein to be assigned as PE lipid species [78].

Native MS is a versatile and rapid technique for obtaining non-covalent interactions of membrane proteins that are often difficult or outright intractable to other techniques. It can be used to study membrane proteins in a varied range of membrane mimetic systems, notably in their own native lipid bilayer. Advances in the synthesis and commercialisation of mild and native MS-compatible detergents will continue to be relevant for investigating protein–drug/lipid binding where highly purified samples are required. Increasing the repertoire of activation regimes that enables release of intact oligomers from lipid bilayer nanodiscs will allow selective disruptions of the nanodiscs with minimal dissociation of the encapsulated protein complexes. Such an advance will pave the way for releasing intact proteins from more complex biological membranes and tissues for which strong lipid–protein interactions must be overcome.

Recent advances in native MS have enabled protein assemblies to be released from intact lens tissue [12]. For such a method to be widely applicable, it will be critical to identify novel sample preparation strategies and develop gas phase activation techniques that ensure efficient but selective dissociation. Tagging of endogenous proteins for enrichment with affinity binders and the development of new affinity probes to enrich the target proteins over other noninteracting members of the proteome will enhance the signal-to-noise ratio in applying this method to investigations of specific pathways. It will be interesting to be able to analyse various proteoforms in a healthy versus diseased state in a single experiment to accelerate diagnosis, monitor disease progression, and perform drug screening in the native context of their protein targets – towards this goal, native top-down holds great promise.

  • In the modern era of structural biology, native MS has emerged as a powerful tool for defining the molecular sociology of a variety of membrane protein targets including transporters, channels, and receptors. Starting off with detergents and membrane mimetics, this area of research is now going through a transformational period towards more native environments.

  • Although still in its infancy, analysis of complexes directly from native membranes by native MS has already managed to capture several key signalling events. However, the full potential and widespread use of this approach are yet to happen.

  • In addition to optimising sample preparation strategies, there is a clear need for new instrumentation, which combines sensitivity, dynamic mass range, resolution, and access to diverse ion activation techniques including HCD, ECD, ETD, IRMPD and UVPD.

The authors declare that there are no competing interests associated with the manuscript.

J.R.B. acknowledges the Royal Society for funding and is supported by a University Research Fellowship (URF\R1\211567).

Open access for this article was enabled by the participation of University of Oxford in an all-inclusive Read & Publish agreement with Portland Press and the Biochemical Society under a transformative agreement with JISC.

The authors would like to thank Prof Carol Robinson for kindly providing figure 2 and for helpful discussions during the preparation of this manuscript. We apologise to colleagues whose work was not included due to space considerations. AOO is a junior research fellow at St. Anne's College, University of Oxford.

A2aR

adenosine 2A receptor

C8E4

tetraethylene Glycol Monooctyl Ether

CID

collision induced dissociation

Cryo-EM

cryogenic electron microscopy

DDM

n-dodecyl β-D-maltoside

DIBMA

diisobutylene-maleic acid

ECD

electron-capture dissociation

EID

electron-induced dissociation

ETD

electron-transfer dissociation

GCGR

glucagon receptor

GPCR

G-protein-coupled receptor

HCD

higher-energy collisional dissociation

HDX

hydrogen-deuterium exchange

IRMPD

infrared multiple photon dissociation

LDAO

n-Dodecyl-N,N-Dimethylamine-N-Oxide

LMNG

lauryl maltose neopentyl glycol

MS

mass spectrometry

NG

nonylglucoside

OG

octylglucoside

OGD

oligoglycerol detergents

PC

phosphatidylcholine

PE

phosphatidylethanolamine

SMA

styrene maleic anhydride

SMALPS

styrene-maleic acid lipid particles

SoLVe-MS

sonicated lipid vesicle mass spectrometry

UVPD

ultraviolet photodissociation

XL-MS

cross-linking mass spectrometry

1.
Dobson
L.
,
Reményi
I.
and
Tusnády
G.E.
(
2015
)
The human transmembrane proteome
.
Biol. Direct
10
,
31
[PubMed]
2.
Marinko
J.T.
,
Huang
H.
,
Penn
W.D.
,
Capra
J.A.
,
Schlebach
J.P.
and
Sanders
C.R.
(
2019
)
Folding and misfolding of human membrane proteins in health and disease: from single molecules to cellular proteostasis
.
Chem. Rev.
119
,
5537
5606
[PubMed]
3.
Santos
R.
,
Ursu
O.
,
Gaulton
A.
,
Bento
A.P.
,
Donadi
R.S.
,
Bologa
C.G.
et al.
(
2017
)
A comprehensive map of molecular drug targets
.
Nat. Rev. Drug Discovery
16
,
19
34
4.
Sriram
K.
and
Insel
P.A.
(
2018
)
G protein-coupled receptors as targets for approved drugs: how many targets and how many drugs?
Mol. Pharmacol.
93
,
251
258
[PubMed]
5.
Hutchings
C.J.
,
Colussi
P.
and
Clark
T.G.
(
2019
)
Ion channels as therapeutic antibody targets
.
MAbs
11
,
265
296
[PubMed]
6.
Nageswar
K.V.
,
Sharma
M.
,
Patil
D.N.
,
Nayak
S.
,
Roy
A.
and
Singh
A.K.
(
2022
)
Current trends in membrane protein crystallography
.
Advances in Protein Molecular and Structural Biology Methods
, pp.
277
290
,
Elsevier
7.
Brown
C.J.
,
Trieber
C.
and
Overduin
M.
(
2021
)
Structural biology of endogenous membrane protein assemblies in native nanodiscs
.
Curr. Opin. Struct. Biol.
69
,
70
77
[PubMed]
8.
Kühlbrandt
W.
(
2022
)
Forty years in cryoEM of membrane proteins
.
Microscopy
71
,
i30
i50
[PubMed]
9.
Jumper
J.
,
Evans
R.
,
Pritzel
A.
,
Green
T.
,
Figurnov
M.
,
Ronneberger
O.
et al.
(
2021
)
Highly accurate protein structure prediction with AlphaFold
.
Nature
596
,
583
589
[PubMed]
10.
Baek
M.
,
DiMaio
F.
,
Anishchenko
I.
,
Dauparas
J.
,
Ovchinnikov
S.
,
Lee
G.R.
et al.
(
2021
)
Accurate prediction of protein structures and interactions using a three-track neural network
.
Science
373
,
871
876
[PubMed]
11.
Barth
M.
and
Schmidt
C.
(
2020
)
Native mass spectrometry-a valuable tool in structural biology
.
J. Mass Spectrom.
55
,
e4578
[PubMed]
12.
Hale
O.J.
and
Cooper
H.J.
(
2022
)
Native ambient mass spectrometry of an intact membrane protein assembly and soluble protein assemblies directly from lens tissue
.
Angew. Chem. Int. Ed.
61
,
e202201458
13.
Hammerschmid
D.
,
van Dyck
J.F.
,
Sobott
F.
and
Calabrese
A.N.
(
2020
)
Interrogating membrane protein structure and lipid interactions by native mass spectrometry
.
Methods Mol. Biol.
2168
,
233
261
[PubMed]
14.
Jooß
K.
,
McGee
J.P.
and
Kelleher
N.L.
(
2022
)
Native mass spectrometry at the convergence of structural biology and compositional proteomics
.
Acc. Chem. Res.
55
,
1928
1937
[PubMed]
15.
Karch
K.R.
,
Snyder
D.T.
,
Harvey
S.R.
and
Wysocki
V.H.
(
2022
)
Native mass spectrometry: recent progress and remaining challenges
.
Annu. Rev. Biophys.
51
,
157
179
[PubMed]
16.
Bennett
J.L.
,
Nguyen
G.T.
and
Donald
W.A.
(
2021
)
Protein–small molecule interactions in native mass spectrometry
.
Chem. Rev.
122
,
7327
7385
[PubMed]
17.
Gavriilidou
A.F.
,
Sokratous
K.
,
Yen
H.-Y.
and
De Colibus
L.
(
2022
)
High-throughput native mass spectrometry screening in drug discovery
.
Front. Mol. Biosci.
9
,
837901
[PubMed]
18.
Reher
R.
,
Aron
A.T.
,
Fajtová
P.
,
Stincone
P.
,
Wagner
B.
,
Pérez-Lorente
A.I.
et al.
(
2022
)
Native metabolomics identifies the rivulariapeptolide family of protease inhibitors
.
Nat. Commun.
13
,
4619
[PubMed]
19.
Dafun
A.S.
and
Marcoux
J.
(
2022
)
Structural mass spectrometry of membrane proteins
.
Biochim. Biophys. Acta
1870
,
140813
20.
Giladi
M.
and
Khananshvili
D.
(
2020
)
Hydrogen-deuterium exchange mass-spectrometry of secondary active transporters: from structural dynamics to molecular mechanisms
.
Front. Pharmacol.
11
,
70
[PubMed]
21.
Martens
C.
and
Politis
A.
(
2020
)
A glimpse into the molecular mechanism of integral membrane proteins through hydrogen–deuterium exchange mass spectrometry
.
Protein Sci.
29
,
1285
1301
[PubMed]
22.
Shukla
A.K.
,
Westfield
G.H.
,
Xiao
K.
,
Reis
R.I.
,
Huang
L.-Y.
,
Tripathi-Shukla
P.
et al.
(
2014
)
Visualization of arrestin recruitment by a G-protein-coupled receptor
.
Nature
512
,
218
222
[PubMed]
23.
Jia
R.
,
Martens
C.
,
Shekhar
M.
,
Pant
S.
,
Pellowe
G.A.
,
Lau
A.M.
et al.
(
2020
)
Hydrogen-deuterium exchange mass spectrometry captures distinct dynamics upon substrate and inhibitor binding to a transporter
.
Nat. Commun.
11
,
6162
[PubMed]
24.
Mikhailov
V.A.
,
Mize
T.H.
,
Benesch
J.L.
and
Robinson
C.V.
(
2014
)
Mass-selective soft-landing of protein assemblies with controlled landing energies
.
Anal. Chem.
86
,
8321
8328
[PubMed]
25.
Esser
T.K.
,
Böhning
J.
,
Fremdling
P.
,
Agasid
M.T.
,
Costin
A.
,
Fort
K.
et al.
(
2022
)
Mass-selective and ice-free electron cryomicroscopy protein sample preparation via native electrospray ion-beam deposition
.
PNAS Nexus
1
,
pgac153
26.
Westphall
M.S.
,
Lee
K.W.
,
Salome
A.Z.
,
Lodge
J.M.
,
Grant
T.
and
Coon
J.J.
(
2022
)
Three-dimensional structure determination of protein complexes using matrix-landing mass spectrometry
.
Nat. Commun.
13
,
2276
[PubMed]
27.
Niebling
S.
,
Veith
K.
,
Vollmer
B.
,
Lizarrondo
J.
,
Burastero
O.
,
Schiller
J.
et al.
(
2022
)
Biophysical screening pipeline for Cryo-EM grid preparation of membrane proteins
.
Front. Mol. Biosci.
9
,
882288
[PubMed]
28.
Olinares
P.D.B.
,
Kang
J.Y.
,
Llewellyn
E.
,
Chiu
C.
,
Chen
J.
,
Malone
B.
et al.
(
2021
)
Native mass spectrometry-based screening for optimal sample preparation in single-particle Cryo-EM
.
Structure
29
,
186.e186
195.e186
[PubMed]
29.
Pyle
E.
,
Kalli
A.C.
,
Amillis
S.
,
Hall
Z.
,
Lau
A.M.
,
Hanyaloglu
A.C.
et al.
(
2018
)
Structural lipids enable the formation of functional oligomers of the eukaryotic purine symporter UapA
.
Cell Chem. Biol.
25
,
840.e844
848.e844
[PubMed]
30.
Hoi
K.K.
,
Bada Juarez
J.F.
,
Judge
P.J.
,
Yen
H.Y.
,
Wu
D.
,
Vinals
J.
et al.
(
2021
)
Detergent-free lipodisq nanoparticles facilitate high-resolution mass spectrometry of folded integral membrane proteins
.
Nano Lett.
21
,
2824
2831
[PubMed]
31.
Liu
R.
,
Xia
S.
and
Li
H.
(
2022
)
Native top-down mass spectrometry for higher-order structural characterization of proteins and complexes
.
Mass Spectrom. Rev.
e21793
[PubMed]
32.
Cong
X.
,
Patrick
J.W.
,
Liu
Y.
,
Liang
X.
,
Liu
W.
and
Laganowsky
A.
(
2022
)
Investigation of protein-lipid interactions using native mass spectrometry
.
Methods Mol. Biol.
2349
,
41
64
[PubMed]
33.
Keener
J.E.
,
Zhang
G.
and
Marty
M.T.
(
2021
)
Native mass spectrometry of membrane proteins
.
Anal. Chem.
93
,
583
597
[PubMed]
34.
Anandan
A.
and
Vrielink
A.
(
2016
)
Detergents in membrane protein purification and crystallisation
.
Adv. Exp. Med. Biol.
922
,
13
28
[PubMed]
35.
Seddon
A.M.
,
Curnow
P.
and
Booth
P.J.
(
2004
)
Membrane proteins, lipids and detergents: not just a soap opera
.
Biochim. Biophys. Acta
1666
,
105
117
[PubMed]
36.
Kurauskas
V.
,
Hessel
A.
,
Ma
P.
,
Lunetti
P.
,
Weinhäupl
K.
,
Imbert
L.
et al.
(
2018
)
How detergent impacts membrane proteins: atomic-level views of mitochondrial carriers in dodecylphosphocholine
.
J. Phys. Chem. Lett.
9
,
933
938
[PubMed]
37.
Bordag
N.
and
Keller
S.
(
2010
)
α-Helical transmembrane peptides: A “Divide and Conquer” approach to membrane proteins
.
Chem. Phys. Lipids
163
,
1
26
[PubMed]
38.
Kotov
V.
,
Bartels
K.
,
Veith
K.
,
Josts
I.
,
Subhramanyam
U.K.T.
,
Günther
C.
et al.
(
2019
)
High-throughput stability screening for detergent-solubilized membrane proteins
.
Sci. Rep.
9
,
10379
[PubMed]
39.
Landreh
M.
,
Costeira-Paulo
J.
,
Gault
J.
,
Marklund
E.G.
and
Robinson
C.V.
(
2017
)
Effects of detergent micelles on lipid binding to proteins in electrospray ionization mass spectrometry
.
Anal. Chem.
89
,
7425
7430
[PubMed]
40.
Reading
E.
,
Walton
T.A.
,
Liko
I.
,
Marty
M.T.
,
Laganowsky
A.
,
Rees
D.C.
et al.
(
2015
)
The effect of detergent, temperature, and lipid on the oligomeric state of MscL constructs: insights from mass spectrometry
.
Chem. Biol.
22
,
593
603
[PubMed]
41.
Bolla
J.R.
,
Corey
R.A.
,
Sahin
C.
,
Gault
J.
,
Hummer
A.
,
Hopper
J.T.S.
et al.
(
2020
)
A mass-spectrometry-based approach to distinguish annular and specific lipid binding to membrane proteins
.
Angew. Chem. Int. Ed. Engl.
59
,
3523
3528
[PubMed]
42.
Reading
E.
,
Liko
I.
,
Allison
T.M.
,
Benesch
J.L.
,
Laganowsky
A.
and
Robinson
C.V.
(
2015
)
The role of the detergent micelle in preserving the structure of membrane proteins in the gas phase
.
Angew. Chem. Int. Ed.
54
,
4577
4581
43.
Urner
L.H.
,
Liko
I.
,
Yen
H.Y.
,
Hoi
K.K.
,
Bolla
J.R.
,
Gault
J.
et al.
(
2020
)
Modular detergents tailor the purification and structural analysis of membrane proteins including G-protein coupled receptors
.
Nat. Commun.
11
,
564
[PubMed]
44.
Urner
L.H.
,
Schulze
M.
,
Maier
Y.B.
,
Hoffmann
W.
,
Warnke
S.
,
Liko
I.
et al.
(
2020
)
A new azobenzene-based design strategy for detergents in membrane protein research
.
Chem. Sci.
11
,
3538
3546
[PubMed]
45.
Marty
M.T.
,
Hoi
K.K.
and
Robinson
C.V.
(
2016
)
Interfacing membrane mimetics with mass spectrometry
.
Acc. Chem. Res.
49
,
2459
2467
[PubMed]
46.
Hopper
J.T.S.
,
Yu
Y.T.-C.
,
Li
D.
,
Raymond
A.
,
Bostock
M.
,
Liko
I.
et al.
(
2013
)
Detergent-free mass spectrometry of membrane protein complexes
.
Nat. Methods
10
,
1206
1208
[PubMed]
47.
Knowles
T.J.
,
Finka
R.
,
Smith
C.
,
Lin
Y.-P.
,
Dafforn
T.
and
Overduin
M.
(
2009
)
Membrane proteins solubilized intact in lipid containing nanoparticles bounded by styrene maleic acid copolymer
.
J. Am. Chem. Soc.
131
,
7484
7485
[PubMed]
48.
Oluwole
A.O.
,
Danielczak
B.
,
Meister
A.
,
Babalola
J.O.
,
Vargas
C.
and
Keller
S.
(
2017
)
Solubilization of membrane proteins into functional lipid-bilayer nanodiscs using a diisobutylene/maleic acid copolymer
.
Angew. Chem. Int. Ed. Engl.
56
,
1919
1924
[PubMed]
49.
Hellwig
N.
,
Peetz
O.
,
Ahdash
Z.
,
Tascón
I.
,
Booth
P.J.
,
Mikusevic
V.
et al.
(
2018
)
Native mass spectrometry goes more native: investigation of membrane protein complexes directly from SMALPs
.
Chem. Commun.
54
,
13702
13705
50.
Fantin
S.M.
,
Parson
K.F.
,
Yadav
P.
,
Juliano
B.
,
Li
G.C.
,
Sanders
C.R.
et al.
(
2021
)
Ion mobility–mass spectrometry reveals the role of peripheral myelin protein dimers in peripheral neuropathy
.
Proc. Natl. Acad. Sci.
118
,
e2015331118
51.
Keener
J.E.
,
Jayasekera
H.S.
and
Marty
M.T.
(
2022
)
Investigating the lipid selectivity of membrane proteins in heterogeneous nanodiscs
.
Anal. Chem.
94
,
8497
8505
[PubMed]
52.
Kostelic
M.M.
,
Zak
C.K.
,
Jayasekera
H.S.
and
Marty
M.T.
(
2021
)
Assembly of model membrane nanodiscs for native mass spectrometry
.
Anal. Chem.
93
,
5972
5979
[PubMed]
53.
Keener
J.E.
,
Zambrano
D.E.
,
Zhang
G.
,
Zak
C.K.
,
Reid
D.J.
,
Deodhar
B.S.
et al.
(
2019
)
Chemical additives enable native mass spectrometry measurement of membrane protein oligomeric state within intact nanodiscs
.
J. Am. Chem. Soc.
141
,
1054
1061
[PubMed]
54.
Chen
S.
,
Getter
T.
,
Salom
D.
,
Wu
D.
,
Quetschlich
D.
,
Chorev
D.S.
et al.
(
2022
)
Capturing a rhodopsin receptor signalling cascade across a native membrane
.
Nature
604
,
384
390
[PubMed]
55.
Chorev
D.S.
,
Baker
L.A.
,
Wu
D.
,
Beilsten-Edmands
V.
,
Rouse
S.L.
,
Zeev-Ben-Mordehai
T.
et al.
(
2018
)
Protein assemblies ejected directly from native membranes yield complexes for mass spectrometry
.
Science
362
,
829
834
[PubMed]
56.
Harvey
S.R.
,
O'Neale
C.
,
Schey
K.L.
and
Wysocki
V.H.
(
2022
)
Native mass spectrometry and surface induced dissociation provide insight into the post-translational modifications of tetrameric AQP0 isolated from bovine eye lens
.
Anal. Chem.
94
,
1515
1519
[PubMed]
57.
Yang
Y.
,
Liu
J.
,
Clarke
B.R.
,
Seidel
L.
,
Bolla
J.R.
,
Ward
P.N.
et al.
(
2021
)
The molecular basis of regulation of bacterial capsule assembly by Wzc
.
Nat. Commun.
12
,
4349
[PubMed]
58.
Ro
S.Y.
,
Schachner
L.F.
,
Koo
C.W.
,
Purohit
R.
,
Remis
J.P.
,
Kenney
G.E.
et al.
(
2019
)
Native top-down mass spectrometry provides insights into the copper centers of membrane-bound methane monooxygenase
.
Nat. Commun.
10
,
2675
[PubMed]
59.
Fantin
S.M.
,
Parson
K.F.
,
Yadav
P.
,
Juliano
B.
,
Li
G.C.
,
Sanders
C.R.
et al.
(
2021
)
Ion mobility-mass spectrometry reveals the role of peripheral myelin protein dimers in peripheral neuropathy
.
Proc. Natl. Acad. Sci. U.S.A.
118
,
e2015331118
[PubMed]
60.
Abbas
Y.M.
,
Wu
D.
,
Bueler
S.A.
,
Robinson
C.V.
and
Rubinstein
J.L.
(
2020
)
Structure of V-ATPase from the mammalian brain
.
Science
367
,
1240
1246
[PubMed]
61.
Landreh
M.
,
Marty
M.T.
,
Gault
J.
and
Robinson
C.V.
(
2016
)
A sliding selectivity scale for lipid binding to membrane proteins
.
Curr. Opin. Struct. Biol.
39
,
54
60
[PubMed]
62.
Laganowsky
A.
,
Reading
E.
,
Allison
T.M.
,
Ulmschneider
M.B.
,
Degiacomi
M.T.
,
Baldwin
A.J.
et al.
(
2014
)
Membrane proteins bind lipids selectively to modulate their structure and function
.
Nature
510
,
172
175
[PubMed]
63.
Oluwole
A.O.
,
Corey
R.A.
,
Brown
C.M.
,
Hernández-Rocamora
V.M.
,
Stansfeld
P.J.
,
Vollmer
W.
et al.
(
2022
)
Peptidoglycan biosynthesis is driven by lipid transfer along enzyme-substrate affinity gradients
.
Nat. Commun.
13
,
1
12
[PubMed]
64.
Patil
D.N.
,
Singh
S.
,
Laboute
T.
,
Strutzenberg
T.S.
,
Qiu
X.
,
Wu
D.
et al.
(
2022
)
Cryo-EM structure of human GPR158 receptor coupled to the RGS7-Gβ5 signaling complex
.
Science
375
,
86
91
[PubMed]
65.
Webby
M.N.
,
Oluwole
A.O.
,
Pedebos
C.
,
Inns
P.G.
,
Olerinyova
A.
,
Prakaash
D.
et al.
(
2022
)
Lipids mediate supramolecular outer membrane protein assembly in bacteria
.
Sci. Adv.
8
,
eadc9566
[PubMed]
66.
Bolla
J.R.
,
Howes
A.C.
,
Fiorentino
F.
and
Robinson
C.V.
(
2020
)
Assembly and regulation of the chlorhexidine-specific efflux pump AceI
.
Proc. Natl. Acad. Sci.
117
,
17011
17018
67.
Fiorentino
F.
,
Sauer
J.B.
,
Qiu
X.
,
Corey
R.A.
,
Cassidy
C.K.
,
Mynors-Wallis
B.
et al.
(
2021
)
Dynamics of an LPS translocon induced by substrate and an antimicrobial peptide
.
Nat. Chem. Biol.
17
,
187
195
[PubMed]
68.
Kaur
H.
,
Jakob
R.P.
,
Marzinek
J.K.
,
Green
R.
,
Imai
Y.
,
Bolla
J.R.
et al.
(
2021
)
The antibiotic darobactin mimics a β-strand to inhibit outer membrane insertase
.
Nature
593
,
125
129
[PubMed]
69.
Petroff
J.T.
,
Tong
I.I.
,
Chen
A.
,
Dekoster
L.J.
,
Khan
G.T.
,
Abramson
F.
et al.
(
2020
)
Charge reduction of membrane proteins in native mass spectrometry using alkali metal acetate salts
.
Anal. Chem.
92
,
6622
6630
[PubMed]
70.
Hernández
H.
and
Robinson
C.V.
(
2007
)
Determining the stoichiometry and interactions of macromolecular assemblies from mass spectrometry
.
Nat. Protoc.
2
,
715
726
[PubMed]
71.
Xia
Z.
and
Williams
E.R.
(
2019
)
Effect of droplet lifetime on where ions are formed in electrospray ionization
.
Analyst
144
,
237
248
72.
Agasid
M.T.
,
Sørensen
L.
,
Urner
L.H.
,
Yan
J.
and
Robinson
C.V.
(
2021
)
The Effects of Sodium Ions on Ligand Binding and Conformational States of G Protein-Coupled Receptors—Insights from Mass Spectrometry
.
J. Am. Chem. Soc.
143
,
4085
4089
[PubMed]
73.
Yen
H.-Y.
,
Liko
I.
,
Song
W.
,
Kapoor
P.
,
Almeida
F.
,
Toporowska
J.
et al.
(
2022
)
Mass spectrometry captures biased signalling and allosteric modulation of a G-protein-coupled receptor
.
Nat. Chem.
14
,
1375
1382
[PubMed]
74.
Gray
D.A.
,
White
J.B.R.
,
Oluwole
A.O.
,
Rath
P.
,
Glenwright
A.J.
,
Mazur
A.
et al.
(
2021
)
Insights into SusCD-mediated glycan import by a prominent gut symbiont
.
Nat. Commun.
12
,
44
[PubMed]
75.
Bolla
J.R.
,
Sauer
J.B.
,
Wu
D.
,
Mehmood
S.
,
Allison
T.M.
and
Robinson
C.V.
(
2018
)
Direct observation of the influence of cardiolipin and antibiotics on lipid II binding to MurJ
.
Nat. Chem.
10
,
363
371
[PubMed]
76.
Chorev
D.S.
,
Tang
H.
,
Rouse
S.L.
,
Bolla
J.R.
,
von Kügelgen
A.
,
Baker
L.A.
et al.
(
2020
)
The use of sonicated lipid vesicles for mass spectrometry of membrane protein complexes
.
Nat. Protoc.
15
,
1690
1706
[PubMed]
77.
Gupta
K.
,
Li
J.
,
Liko
I.
,
Gault
J.
,
Bechara
C.
,
Wu
D.
et al.
(
2018
)
Identifying key membrane protein lipid interactions using mass spectrometry
.
Nat. Protoc.
13
,
1106
1120
[PubMed]
78.
Gault
J.
,
Liko
I.
,
Landreh
M.
,
Shutin
D.
,
Bolla
J.R.
,
Jefferies
D.
et al.
(
2020
)
Combining native and ‘omics’ mass spectrometry to identify endogenous ligands bound to membrane proteins
.
Nat. Methods
17
,
505
508
[PubMed]
79.
Jin
R.
,
He
S.
,
Black
K.A.
,
Clarke
O.B.
,
Wu
D.
,
Bolla
J.R.
et al.
(
2022
)
Ion currents through Kir potassium channels are gated by anionic lipids
.
Nat. Commun.
13
,
490
[PubMed]
80.
Khera
R.
,
Mehdipour
A.R.
,
Bolla
J.R.
,
Kahnt
J.
,
Welsch
S.
,
Ermler
U.
et al.
(
2022
)
Cryo-EM structures of pentameric autoinducer-2 exporter from Escherichia coli reveal its transport mechanism
.
EMBO J.
41
,
e109990
[PubMed]
81.
Sicoli
G.
,
Konijnenberg
A.
,
Guérin
J.
,
Hessmann
S.
,
Del Nero
E.
,
Hernandez-Alba
O.
et al.
(
2022
)
Large-scale conformational changes of FhaC provide insights into the two-partner secretion mechanism
.
Front. Mol. Biosci.
9
,
950871
[PubMed]
82.
Kjølbye
L.R.
,
Sørensen
L.
,
Yan
J.
,
Berglund
N.A.
,
Ferkinghoff-Borg
J.
,
Robinson
C.V.
et al.
(
2022
)
Lipid modulation of a class B GPCR: elucidating the modulatory role of PI(4,5)P2 Lipids
.
J. Chem. Inf. Model.
62
,
6788
6802
[PubMed]
83.
Yen
H.Y.
,
Abramsson
M.L.
,
Agasid
M.T.
,
Lama
D.
,
Gault
J.
,
Liko
I.
et al.
(
2022
)
Electrospray ionization of native membrane proteins proceeds via a charge equilibration step
.
RSC Adv.
12
,
9671
9680
[PubMed]
84.
Matsuoka
R.
,
Fudim
R.
,
Jung
S.
,
Zhang
C.
,
Bazzone
A.
,
Chatzikyriakidou
Y.
et al.
(
2022
)
Structure, mechanism and lipid-mediated remodeling of the mammalian Na(+)/H(+) exchanger NHA2
.
Nat. Struct. Mol. Biol.
29
,
108
120
[PubMed]
85.
Yen
H.-Y.
,
Liko
I.
,
Song
W.
,
Kapoor
P.
,
Almeida
F.
,
Toporowska
J.
et al.
(
2022
)
Mass spectrometry captures biased signalling and allosteric modulation of a G-protein-coupled receptor
.
Nat. Chem.
14
,
1375
1382
[PubMed]
86.
van den Berg
B.
,
Pedebos
C.
,
Bolla
J.R.
,
Robinson
C.V.
,
Baslé
A.
and
Khalid
S.
(
2021
)
Structural basis for silicic acid uptake by higher plants
.
J. Mol. Biol.
433
,
167226
[PubMed]
87.
Zhao
J.
,
Hellwig
N.
,
Djahanschiri
B.
,
Khera
R.
,
Morgner
N.
,
Ebersberger
I.
et al.
(
2022
)
Assembly and functional role of PACE transporter PA2880 from Pseudomonas aeruginosa
.
Microbiol. Spectrum
10
,
e01453
e01421
88.
Guffick
C.
,
Hsieh
P.-Y.
,
Ali
A.
,
Shi
W.
,
Howard
J.
,
Chinthapalli
D.K.
et al.
(
2022
)
Drug-dependent inhibition of nucleotide hydrolysis in the heterodimeric ABC multidrug transporter PatAB from Streptococcus pneumoniae
.
FEBS J.
289
,
3770
3788
[PubMed]
89.
Schmidpeter
P.A.M.
,
Wu
D.
,
Rheinberger
J.
,
Riegelhaupt
P.M.
,
Tang
H.
,
Robinson
C.V.
et al.
(
2022
)
Anionic lipids unlock the gates of select ion channels in the pacemaker family
.
Nat. Struct. Mol. Biol.
29
,
1092
1100
[PubMed]
90.
Zhu
Y.
,
Schrecke
S.
,
Tang
S.
,
Odenkirk
M.T.
,
Walker
T.
,
Stover
L.
et al.
(
2022
)
Cupric Ions Selectively Modulate TRAAK–Phosphatidylserine Interactions
.
J. Am. Chem. Soc.
144
,
7048
7053
[PubMed]
91.
Grison
C.M.
,
Lambey
P.
,
Jeannot
S.
,
Del Nero
E.
,
Fontanel
S.
,
Peysson
F.
et al.
(
2021
)
Molecular insights into mechanisms of GPCR hijacking by Staphylococcus aureus
.
Proc. Natl. Acad. Sci.
118
,
e2108856118
92.
Qiao
P.
,
Schrecke
S.
,
Lyu
J.
,
Zhu
Y.
,
Zhang
T.
,
Benavides
A.
et al.
(
2021
)
Insight into the Phospholipid-binding preferences of Kir3.4
.
Biochemistry
60
,
3813
3821
[PubMed]
93.
Cater
R.J.
,
Chua
G.L.
,
Erramilli
S.K.
,
Keener
J.E.
,
Choy
B.C.
,
Tokarz
P.
et al.
(
2021
)
Structural basis of omega-3 fatty acid transport across the blood–brain barrier
.
Nature
595
,
315
319
[PubMed]
94.
Moysa
A.
,
Steczkiewicz
K.
,
Niedzialek
D.
,
Hammerschmid
D.
,
Zhukova
L.
,
Sobott
F.
et al.
(
2021
)
A model of full-length RAGE in complex with S100B
.
Structure
29
,
989.e1006
1002.e1006
[PubMed]
95.
Norris
C.E.
,
Keener
J.E.
,
Perera
S.M.D.C.
,
Weerasinghe
N.
,
Fried
S.D.E.
,
Resager
W.C.
et al.
(
2021
)
Native mass spectrometry reveals the simultaneous binding of lipids and zinc to rhodopsin
.
Int. J. Mass Spectrom.
460
,
116477
[PubMed]
96.
Stefan
E.
,
Obexer
R.
,
Hofmann
S.
,
Vu Huu
K.
,
Huang
Y.
,
Morgner
N.
et al.
(
2021
)
De novo macrocyclic peptides dissect energy coupling of a heterodimeric ABC transporter by multimode allosteric inhibition
.
eLife
10
,
e67732
[PubMed]
97.
Housden
N.G.
,
Webby
M.N.
,
Lowe
E.D.
,
El-Baba
T.J.
,
Kaminska
R.
,
Redfield
C.
et al.
(
2021
)
Toxin import through the antibiotic efflux channel TolC
.
Nat. Commun.
12
,
4625
[PubMed]
98.
Vorobieva
A.A.
,
White
P.
,
Liang
B.
,
Horne
J.E.
,
Bera
A.K.
,
Chow
C.M.
et al.
(
2021
)
De novo design of transmembrane β-barrels
.
Science
371
,
eabc8182
[PubMed]
99.
Reading
E.
,
Ahdash
Z.
,
Fais
C.
,
Ricci
V.
,
Wang-Kan
X.
,
Grimsey
E.
et al.
(
2020
)
Perturbed structural dynamics underlie inhibition and altered efflux of the multidrug resistance pump AcrB
.
Nat. Commun.
11
,
5565
[PubMed]
100.
Urner
L.H.
,
Schulze
M.
,
Maier
Y.B.
,
Hoffmann
W.
,
Warnke
S.
,
Liko
I.
et al.
(
2020
)
A new azobenzene-based design strategy for detergents in membrane protein research
.
Chem. Sci.
11
,
3538
3546
[PubMed]
101.
Neviani
V.
,
van Deventer
S.
,
Wörner
T.P.
,
Xenaki
K.T.
,
van de Waterbeemd
M.
,
Rodenburg
R.N.P.
et al.
(
2020
)
Site-specific functionality and tryptophan mimicry of lipidation in tetraspanin CD9
.
FEBS J.
287
,
5323
5344
[PubMed]
102.
Zhu
G.
,
Zeng
H.
,
Zhang
S.
,
Juli
J.
,
Pang
X.
,
Hoffmann
J.
et al.
(
2020
)
A 3.3 Å-resolution structure of hyperthermophilic respiratory complex III reveals the mechanism of its thermal stability
.
Angew. Chem. Int. Ed. Engl.
59
,
343
351
[PubMed]
103.
Wang
L.
,
Qian
H.
,
Nian
Y.
,
Han
Y.
,
Ren
Z.
,
Zhang
H.
et al.
(
2020
)
Structure and mechanism of human diacylglycerol O-acyltransferase 1
.
Nature
581
,
329
332
[PubMed]
104.
Tan
Y.Z.
,
Rodrigues
J.
,
Keener
J.E.
,
Zheng
R.B.
,
Brunton
R.
,
Kloss
B.
et al.
(
2020
)
Cryo-EM structure of arabinosyltransferase EmbB from Mycobacterium smegmatis
.
Nat. Commun.
11
,
3396
[PubMed]
105.
Behrens
H.M.
,
Lowe
E.D.
,
Gault
J.
,
Housden
N.G.
,
Kaminska
R.
,
Weber
T.M.
et al.
(
2020
)
Pyocin S5 Import into Pseudomonas aeruginosa Reveals a Generic Mode of Bacteriocin Transport
.
mBio
11
,
e03230
19
[PubMed]
106.
McDowell
M.A.
,
Heimes
M.
,
Fiorentino
F.
,
Mehmood
S.
,
Farkas
Á.
,
Coy-Vergara
J.
et al.
(
2020
)
Structural basis of tail-anchored membrane protein biogenesis by the GET Insertase complex
.
Mol. Cell
80
,
72.e77
86.e77
[PubMed]
107.
Zeng
H.
,
Zhu
G.
,
Zhang
S.
,
Li
X.
,
Martin
J.
,
Morgner
N.
et al.
(
2020
)
Isolated Heme A Synthase from Aquifex aeolicus Is a Trimer
.
mBio
11
,
e02615
e02619
[PubMed]
108.
Schrecke
S.
,
Zhu
Y.
,
McCabe
J.W.
,
Bartz
M.
,
Packianathan
C.
,
Zhao
M.
et al.
(
2021
)
Selective regulation of human TRAAK channels by biologically active phospholipids
.
Nat. Chem. Biol.
17
,
89
95
[PubMed]
109.
Kono
A.
and
Spalding
M.H.
(
2020
)
LCI1, a Chlamydomonas reinhardtii plasma membrane protein, functions in active CO2 uptake under low CO2
.
Plant J.
102
,
1127
1141
[PubMed]
110.
Winkelmann
I.
,
Matsuoka
R.
,
Meier
P.F.
,
Shutin
D.
,
Zhang
C.
,
Orellana
L.
et al.
(
2020
)
Structure and elevator mechanism of the mammalian sodium/proton exchanger NHE9
.
EMBO J.
39
,
e105908
[PubMed]
111.
Jansen
K.B.
,
Inns
P.G.
,
Housden
N.G.
,
Hopper
J.T.S.
,
Kaminska
R.
,
Lee
S.
et al.
(
2020
)
Bifurcated binding of the OmpF receptor underpins import of the bacteriocin colicin N into Escherichia coli
.
J. Biol. Chem.
295
,
9147
9156
[PubMed]
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY). Open access for this article was enabled by the participation of University of Oxford in an all-inclusive Read & Publish agreement with Portland Press and the Biochemical Society under a transformative agreement with JISC.