ALL Metrics
-
Views
-
Downloads
Get PDF
Get XML
Cite
Export
Track
Research Article

In silico prediction of structure and function for a large family of transmembrane proteins that includes human Tmem41b

[version 1; peer review: 1 approved, 2 approved with reservations]
PUBLISHED 03 Dec 2020
Author details Author details
OPEN PEER REVIEW
REVIEWER STATUS

This article is included in the Bioinformatics gateway.

Abstract

Background: Recent strides in computational structural biology have opened up an opportunity to understand previously uncharacterised proteins.  The under-representation of transmembrane proteins in the Protein Data Bank highlights the need to apply new and advanced bioinformatics methods to shed light on their structure and function.  This study focuses on a family of transmembrane proteins containing the Pfam domain PF09335 ('SNARE_ASSOC'/ ‘VTT ‘/’Tvp38’). One prominent member, Tmem41b, has been shown to be involved in early stages of autophagosome formation and is vital in mouse embryonic development as well as being identified as a viral host factor of SARS-CoV-2.
Methods: We used evolutionary covariance-derived information to construct and validate ab initio models, make domain boundary predictions and infer local structural features. 
Results: The results from the structural bioinformatics analysis of Tmem41b and its homologues showed that they contain a tandem repeat that is clearly visible in evolutionary covariance data but much less so by sequence analysis.  Furthermore, cross-referencing of other prediction data with covariance analysis showed that the internal repeat features two-fold rotational symmetry.  Ab initio modelling of Tmem41b and homologues reinforces these structural predictions.  Local structural features predicted to be present in Tmem41b were also present in Cl-/H+ antiporters. 
Conclusions: The results of this study strongly point to Tmem41b and its homologues being transporters for an as-yet uncharacterised substrate and possibly using H+ antiporter activity as its mechanism for transport.

Keywords

ab initio modelling, bioinformatics, autophagy, contact predictions, evolutionary covariance, DedA, SARS-CoV-2, Tmem41b, VTT domain

Introduction

A protein’s structural information is crucial to understand it’s function and evolution. Currently, there is only experimental structural data for a tiny fraction of proteins (Khafizov et al., 2014). For instance, membrane proteins are encoded by 30% of the protein-coding genes of the human genome (Almén et al., 2009), but they only have a 2% representation in the Protein Data Bank (PDB) (Koehler Leman et al., 2015). Membrane protein families are particularly poorly understood due to experimental difficulties, such as over-expression, which can result in toxicity to host cells (Grisshammer & Tateu, 1995), as well as difficulty in finding a suitable membrane mimetic to reconstitute the protein. Additionally, membrane proteins are much less conserved across species compared to water-soluble proteins (Sojo et al., 2016), making sequence-based homologue identification a challenge, and in turn rendering homology modelling of these proteins more difficult. Membrane proteins can be grouped according to their interaction with various cell membranes: integral membrane proteins (IMPs) are permanently anchored whereas peripheral membrane proteins transiently adhere to cell membranes. IMPs that span the membrane are known as transmembrane proteins (TMEMs) as opposed to IMPs that adhere to one side of the membrane (Fowler & Coveney, 2006).

One IMP protein family is Tmem41, which has two human representatives, namely Tmem41a and Tmem41b; both share the PF09335 ('SNARE_ASSOC'/ ‘VTT ‘/’Tvp38’) Pfam (El-Gebali et al., 2019) domain. The profile of Tmem41b has recently risen due to experimental evidence pointing to its involvement in macroautophagy regulation (making it a possible Atg protein, i.e. an autophagy related protein) and lipid mobilisation (Moretti et al., 2018). Other studies identify Tmem41b to be involved in motor circuit function, with TMEM41B-knockout Drosophila showing neuromuscular junction defects and aberrant motor neuron development in knockout zebrafish (Lotti et al., 2012). Also, it has been reported that in TMEM41B-knockout HeLa cells there is an inhibition of Zika virus replication (Scaturro et al., 2018). Tmem41b has also been identified as a host cell factor for SARS-CoV-2 (Schneider et al., 2020). Tmem41b is the only common host cell factor identified for flaviviruses and coronaviruses and is the only autophagy-related protein identified as a viral host factor (Hoffmann et al., 2020).

Additionally, Tmem41b has been shown to be essential for mouse embryonic development: homozygous knockout mice embryos suffer early termination of their development after 7–8 weeks (Van Alstyne et al., 2018). Tmem41b is a structurally uncharacterised 291-residue protein found in the endoplasmic reticulum (ER) localising at the mitochondria-associated ER membranes (Moretti et al., 2018). Disruption of the PF09335 domain by various residue substitutions (Tábara et al., 2019) or its removal (Morita et al., 2018) results in inhibition of autophagosome formation and impaired lipid mobilisation in human embryonic kidney (HEK) cells.

Tmem41b homologues, hereafter referred to as VTT proteins (Morita et al., 2019), are present in all domains of life (Keller & Schneider, 2013). The Pfam PF09335 domain was first identified in the Saccharomyces cerevisiae protein Tvp38 (Inadome et al., 2007), and the authors concluded that Tvp38 associates with the tSNAREs in Tlg2-containing compartments, suggesting a role in membrane transport. Investigations into the bacterial and archaeal prevalence of these proteins showed that 90% of bacterial species and 70% of archaeal species encoded proteins with the PF09335 domain (Doerrler et al., 2013). Bacterial and archaeal PF09335-containing proteins are collectively known as the Death Effector Domain A (DedA) family (Doerrler et al., 2013). Detailed studies of the Escherichia coli DedA proteins have indicated that there are eight E. coli representatives of the DedA family (YqjA, YghB, YabI, Yoh, DedA, YdjX, YdjZ, and YqaA) with overlapping functions (Doerrler et al., 2013; Keller & Schneider, 2013), with Ydjx and Ydjz being the most closely related to human Tmem41b in terms of sequence similarity (Doerrler et al., 2013). Phenotypically, DedA knock-out E. coli cells display increased temperature sensitivity, cell division defects, activation envelope stress pathways, compromised proton motive force, sensitivity to alkaline pH and increased antibiotic susceptibility (Doerrler et al., 2013; Keller et al., 2014). As E. coli expresses multiple DedA homologues, the redundancy protects the cells from the phenotypical effects of single or multiple knock-outs as long as at least one DedA is expressed (Kumar & Doerrler, 2014). Borrelia burgdorferi contains only one DedA protein in its genome and knockout cells display the same phenotype as the E. coli knockout strains. Interestingly, E. coli knockout cells can be rescued with the B. burgdorferi homologue that shows only 19% sequence identity with YdjA. Attempts to rescue B. burgdorferi DedA knockout cells with E. coli homologues have resulted in more complex observations, with different homologues rescuing different phenotypes (Doerrler et al., 2013). The functions of DedA have also been studied in the opportunistic pathogen Pseudomonas aeruginosa where it was concluded that DedA proteins are required for its low antibiotic susceptibility. P. aeruginosa DedA is able to rescue E.coli DedA knockout cells (Justice et al., 2016).

Until the structure of poorly characterised protein families such as Pfam family PF09335 can be elucidated experimentally, ab initio protein modelling can be used to predict a fold allowing for structure-based function inferences (Rigden et al., 2017). Such methods have made significant strides recently due to the availability of contact predictions (Kinch et al., 2016). Prediction of residue-residue contacts relies on the fact that each pair of contacting residues covaries during evolution. The process of co-variation occurs as the properties of the two residues complement each other in order to maintain structural integrity of that local region and, consequently, its original functionality. Therefore, if one residue from the pair is replaced, the other must also change to compensate the physical chemical variation and hence preserve the original structure (Lapedes et al., 1999). The link between two residues can be then reliably detected in multiple sequence alignments by using direct coupling analysis (Morcos et al., 2011) as well as machine learning algorithms (Wu et al., 2020). The predicted contacts can be used for a range of analyses such as the identification of domain boundaries (Rigden, 2002; Simkovic et al., 2017a), but their main application is for contact-based modelling methods which can address larger targets than conventional fragment-assembly-based ab initio methods (Yang et al., 2020).

In the current study, we utilised state of the art methods to make structural predictions for two prominent members of the Pfam family PF09335 (Tmem41b and Yqja) by exploiting data derived from sequence, evolutionary covariance and ab initio modelling. We are able to predict that PF09335 homologues (VTT proteins) contain re-entrant loops (stretches of protein that enter the bilayer but exit on the same side of the membrane) as well as a pseudo-inverted repeat topology. The predicted presence of both of these structural features strongly suggests that VTT proteins are secondary active transporters for an uncharacterised substrate.

Methods

Pfam database screening

Searches using the sequences of VTT domain proteins Tmem41b, Yqja, Ydjx, Ydjz, Tvp38 and Mt2055 were made against the Pfam-A_v32.0 (RRID:SCR_004726) (El-Gebali et al., 2019) database using the HHPred (RRID:SCR_010276) v3.0 server (Zimmermann et al., 2018) with default parameters (-p 20 -Z 10000 -loc -z 1 -b 1 -B 10000 -ssm 2 -sc 1 -seq 1 -dbstrlen 10000 -norealign -maxres 32000 -contxt /cluster/toolkit/production/bioprogs/tools/hh-suite-build-new/data/context_data.crf) and eight iterations for MSA generation in the HHblits (Remmert et al., 2012) stage.

Contact map predictions

The DeepMetapsicov v1.0 server (Kandathil et al., 2019) was used to generate contact predictions with ConKit v0.12 (Simkovic et al., 2017b) utilised to visualise the contact maps. ConPlot (RRID:SCR_019216) was used to overlay additional prediction data (Sánchez Rodríguez et al., unpublished work).

Other prediction data

Transmembrane helical topology predictions were obtained from the Topcons server (Tsirigos et al., 2015). Secondary structure predictions were made employing a local installation of PSIPRED (RRID:SCR_010246) v4.0 (McGuffin et al., 2000). ConKit was also used to predict and visualise potential structural domain boundaries (Rigden, 2002; Simkovic et al., 2017a). Residue analysis of putative amphipathic regions were performed using HELIQUEST (Gautier et al., 2008) to determine the presence, direction and magnitude of any hydrophobic moment. Residue conservation was determined using the Consurf server (Ashkenazy et al., 2016).

Dataset for custom re-entrant database

A library of re-entrant loop sequences together with the putative re-entrant loop sequences from the query proteins were clustered to establish any visible relationships of the sequences. The library was built by obtaining a non-redundant set of 56 re-entrant helix sequences by first retrieving all 714 TM proteins that contain at least one re-entrant loop from the PDBTM (RRID:SCR_011962) (Kozma et al., 2013) and removing redundancy with a 40% identity threshold. The resulting 127 protein structures were split into their component chains, eliminating any chain lacking a re-entrant loop. The subsequent set of 188 unique re-entrant loop sequences were then filtered removing any sequences of less than 10 residues and more than 20, thereby ensuring the collection of sequences conformed to the length of typical (Yan & Luo, 2010) re-entrant loops. The remaining 56 sequences were clustered, supplemented by candidate re-entrant sequences from the proteins studied here. Clustering was performed using CLANS v1.0 (Frickey & Lupas, 2004) with the BLAST results (p-value cut-off threshold of 0.1) (Altschul et al., 1997) used to calculate strengths of similarity.

Model building

Ab initio models were built using the trRosetta (Yang et al., 2020) server with default settings. Conservation was mapped on to the models using the ConSurf server (Ashkenazy et al., 2016). Visualisation of models was achieved using PyMOL (RRID:SCR_000305) v2.3.0 (DeLano, 2002).

Structural alignments

Dali (RRID:SCR_013433) v4.0 (Holm & Laakso, 2016) was used to structurally align the output models and to query against the PDBTM (Kozma et al., 2013).

An earlier version of this article can be found on bioRxiv (doi: https://doi.org/10.1101/2020.06.27.174763)

Results and discussion

Sequence comparisons suggest Pfam families PF09335 and PF06695 are related

HHpred (Zimmermann et al., 2018) was used to screen a selection of VTT proteins against the Pfam database (El-Gebali et al., 2019). Hits were observed in the same region against both PF09335 and the Pfam domain PF06695 (‘Sm_multidrug_ex’) which is strongly indicative of homology: a probability of 99.4% with an E-value of 9E-17 for the PF09335 hit and 98.3% and 2E-10 respectively for PF06695. A HHpred search against the Pfam database using a member of PF06695 - the short archaeal sequence Mt2055 (UniProt code W9DY28) (Apweiler et al., 2004) - returned similar results (Table 1). The Mt2055 sequence originates from the unpublished draft genome of the archaebacterium Methanolobus tindarius DSM 2278. For many of the subsequent analyses, the shorter archaeal sequence was used initially but the clear homology among this set of proteins means that inferences can be drawn across the group.

Table 1. HHpred results for Tmem41b and homologues demonstrate homology between Pfam families PF09335 and PF06695.

PF09335
'SNARE_ASSOC'/ ‘VTT ‘/’Tvp38’
PF06695
‘Sm_multidrug_ex’
SpeciesUniProt
Code
LengthProbabilityE-ValueProbabilityE-Value
Tmem41b Homo sapiensQ5BJD529199.49E-1798.32E-10
YdjxEscherichia coliP7621923699.62.1E-1799.19.9E-13
YdjzEscherichia coliP7622123599.61.1E-1799.04.5E-16
YqjaEscherichia coliP0AA6322099.625.6E-1599.411.3E-12
Tvp38Saccharomyces cerevisiaeP3616433799.47.9E-1598.72.7E-10
Mt2055Methanolobus tindariusW9DY2816899.02.4E-1099.81.8E-20

There are no known experimental protein structures representing PF09335 or PF06695, but both Gremlin and DMPfold have constructed ab initio models for these Pfam domains (Greener et al., 2019; Ovchinnikov et al., 2017).

The predicted Pfam domains are inconsistent with a structural domain

Analysis of the HHpred results obtained for the archaeal protein Mt2055 revealed the presence of additional hits for both PF06695 and PF09335 Pfam domains, in which the C-terminal half of the domains aligned with the N-terminal half of the Archaea protein. For example, residues 1-69 of the archaeal protein aligned with residues 52-117 of the Pfam PF09335 profile with a probability of 74.15%. Interestingly, contact density analysis (Rigden, 2002; Sadowski, 2013) supported the existence of a domain boundary around residue 60, in broad agreement with the HHpred results (Figure 1). Both the HHpred and contact density results therefore pointed to a specific domain structure being present.

5a16f7bc-3e9d-4db6-9810-5d8181a1372a_figure1.gif

Figure 1. Mt2055 domain analysis.

(a) Contact density profile constructed by ConKit (Simkovic et al., 2017b) utilising DeepMetaPSICOV contact prediction. Solid black line represents contact density and dotted red lines mark density minima corresponding to possible domain boundaries. (b) HHalign alignments for the N-terminal and C-terminal Mt2055 halves, formatted using Jalview (Waterhouse et al., 2009) and coloured according to the ClustalX scheme. Red bars represent helical secondary structure. (c) Maps of predicted contacts generated by DeepMetaPSICOV and plotted using ConKit; left is N-terminal half (residues 1-84) and right is C-terminal half (residues 85-168). Black points represent predicted intramolecular contacts.

Sequence & contact prediction map analysis indicate that PF06695 is made up of a tandem repeat

When the Mt2055 sequence was split at residue 60-61, the resulting N-terminal region of 60 residues and the C-terminal section of 79 residues could be aligned using HHalign (Soding, 2005) with a 78% probability and an E-value of 1.9E-3. Examination of the map of predicted contacts for Mt2055 reveals features that are present in both the N- and C-terminal halves of the protein (Figure 1c). Taken together, these data strongly support the existence of a tandem repeat within the Mt2055 protein and hence across the PF06695 and PF09335 protein families.

Interestingly, however, an equivalent sequence analysis with HHpred of other PF09335 homologues including Tmem41b itself does not reveal a repeat. However, inspection of their corresponding predicted contact maps does reveal features repeated when N- and C-halves of the protein are compared (Figure 2). Apparently, evolutionary divergence has removed all trace of the repeat sequence signal in bacterial and eukaryotic proteins, although the feature remains visible by evolutionary covariance analysis.

5a16f7bc-3e9d-4db6-9810-5d8181a1372a_figure2.gif

Figure 2. Tmem41b Contact map constructed using DeepMetaPSICOV and plotted using Conkit.

The highlighted areas represent repeat units that have been revealed through evolutionary covariance analysis.

Ab initio modelling of Mt2055 reveals an unusual topology

Several authors have deposited structures of uncharacterised Pfam families in databases (El-Gebali et al., 2019); however, Pfam domain boundaries for PF09335/PF06695, which define the limits of these previous modelling exercises, do not reflect the conserved structural domain that we predict. Given the fact that the available ab initio models were inconsistent with the transmembrane helix, secondary structure and contact predictions (data not shown), we constructed our own models of Mt2055 as well as Tmem41b and Yqja with trRosetta.

The Mt2055, Tmem41b and Yqja models had estimated TM scores from the trRosetta server of 0.633, 0.624 and 0.635 respectively, suggesting that they were likely to have captured the native fold of the family. All-against-all pairwise structural superposition of the models with DALI gave a mean Z-score of 11.9 confirming their strong similarity. We also used satisfaction of predicted contacts to validate the models (Figure 3) (Simkovic et al., 2017a). This showed that 80% of the top L predicted contacts (where L is the length of the protein) are satisfied by the model contacts for both Mt2055 and Yqja and a value of 60% was achieved for Tmem41b suggestive of good quality models (de Oliveira et al., 2017).

5a16f7bc-3e9d-4db6-9810-5d8181a1372a_figure3.gif

Figure 3.

(a) trRosetta model of MT2055 - amphipathic helix (green) and a re-entrant loop (orange) packed with a TM helix (red) (b) Superposition of DMP predicted contact map for Mt2055 and contacts from the Mt2055 model. Black points are matching contacts, red are mismatches and grey are contacts predicted but not present in the model. Diagonal is a visual representation of transmembrane helix and secondary structure prediction – central diagonal is the visualisation of the TopCons transmembrane prediction (orange being a TM helix) and the outer diagonals are the visual representation of the PsiPred secondary structure prediction (pink – alpha helix and yellow – coil). Red boxes highlight the re-entrant loop and TM helix packing. c) trRosetta model of Tmem41b only showing the conserved structural domain (residues 39-217) d) trRosetta model of Yqja only showing the conserved structural domain (residues 14-176). e) Proposed topology for (extended) VTT domain.

The models (Figure 3) contained interesting features: two inversely symmetrical repeated units each possessing an amphipathic helix (green) and a re-entrant loop (orange) packed with a TM helix (red).

The presence of a re-entrant loop packed against each TM helix can also be seen on predicted contact maps for these proteins (Figure 3b). Interestingly, each of the re-entrant helices is predicted as a single transmembrane region in the TopCons predictions (see the diagonal of Figure 3b) with a two-residue region of coil in the centre. Such a prediction would more obviously be treated as indicative of some kind of kink in the helix (Law et al., 2016) but the explanation here is these regions form re-entrant helices. Similar contact map features, indicative of re-entrant loops packing against TM helices, can be seen clearly on the contact maps of other VTT proteins (data not shown).

The analysis was performed by HELIQUEST (Gautier et al., 2008) which constructed helical wheel diagrams and provided a quantitative measure of the hydrophobic moment for the region being analysed (Figure 4).

5a16f7bc-3e9d-4db6-9810-5d8181a1372a_figure4.gif

Figure 4. Helical wheel diagrams generated using the HELIQUEST server.

Hydrophobic residues are shown in yellow, serine and threonine in purple, basic residues in dark blue, acidic residues in red, asparagine and glutamine in pink, alanine and glycine in grey, histidine in light blue and proline in green circles. Arrows represent direction and magnitude of the hydrophobic moment and residue marked with ‘N’ is the N-terminal end of the putative amphipathic helix with the residue marked ‘C’ being the C-terminal end. (a) Mt2055 putative amphipathic helix 1 (hydrophobic moment of 0.298). (b) Mt2055 putative amphipathic helix 2 (hydrophobic moment of 0.546). (c) Tmem41b putative amphipathic helix 1 (hydrophobic moment of 0.471). (d) Tmem41b putative amphipathic helix 2 (hydrophobic moment of 0.420). (e). Yqja putative amphipathic helix 1 (hydrophobic moment of 0.295). (f) Yqja putative amphipathic helix 2 (hydrophobic moment of 0.396).

Mapping conservation onto the models, using the Consurf server, indicates that the re-entrant loops are highly conserved and therefore likely to be functionally and/or structurally important (Figure 5).

5a16f7bc-3e9d-4db6-9810-5d8181a1372a_figure5.gif

Figure 5.

trRosetta models with Consurf conservation mapping for (a) Mt2055 (b) Tmem41b (c) Yqja. Conservation is shown as a spectrum from blue (highly conserved) to red (not conserved).

Re-entrant loops are also present in Cl-/H+ Antiporters

The presence of re-entrant loops and the high density of conserved residues within them caused us to examine experimentally characterised re-entrant loops in the PDBTM database. A total of 56 non-redundant re-entrant helices were identified (see Methods). All 56 were clustered with the putative re-entrant loops from Mt2055 and four PF09335 homologues (Tmem41b, Tvp38, Ydjx and Ydjz) using relative E-values derived from an all-against-all BLAST run in CLANS (Frickey & Lupas, 2004) with a 0.1 p-value cut-off. The largest cluster contained 14 sequences, of which four were putative re-entrant sequences from the query proteins (Mt2055 C-terminal re-entrant, Ydjx C-terminal re-entrant, Ydjz N-terminal re-entrant & Ydjz C-terminal re-entrant), seven (3org, 5tqq, 3nd0, 3det and 6coy) were re-entrant loop sequences from Cl-/H+ antiporters, one was from a boron exchanger (5l25), one from an electron transporter (2n4x) [albeit classified as a member of the lysine exporter superfamily (Saier et al., 2016)] and one from a mechanogated channel (5z10).

Analysis of the Cl-/H+ antiporter structures show that they contain a similar inverted repeat as we infer for the VTT homologues, resulting in pseudo-2-fold axis of symmetry running along the membrane (Duran & Meiler, 2013). Again similarly, the Cl-/H+ antiporter 3orgA also contains the amphipathic helices on the N-terminal side of the re-entrant loops. The fact that the presence of the amphipathic helices is restricted only to 3orgA and not found in all homologues suggest that these features are not essential for function (Figure 5).

A possible antiporter role for VTT proteins

The presence of re-entrant loops in a transmembrane protein strongly indicates a transporter or pore functionality since this structural feature has, hitherto, only been found in proteins of this kind (Yan & Luo, 2010). The structural similarities between the VTT proteins and the Cl-/H+ antiporters raise the possibility that the families studied here are, in fact, unsuspected distant homologues having this putative pore feature in common. In that regard it is relevant to recall a hypothesis that DedA proteins are H+ antiporters resulting from SDM experiments (Justice et al., 2016).

A recent study has identified key residues (Figure 6) in the E. coli DedA protein Yqja that, when replaced in site directed mutagenesis experiments, resulted in decreased proton motive force across the E. coli inner membrane (Panta et al., 2019). Highlighting the essential residues (E39, D51, R130 and R136) on the Yqja model show that they come together in three-dimensional space with the N-terminal side of the first re-entrant possessing E39 and the C-terminal side possessing D51. R130 and R136 are similarly positioned on the second re-entrant loop (Figure 7). Re-entrant loops are known to form pores and here we have two proton-titratable residues (E39, D51) in close proximity to essential basic residues (R130 and R136) within a putative pore. This three-dimensional arrangement of key residues could serve a role in the coupling of the protonation status with the binding of a yet to be characterised substrate as is postulated for the multi-drug H+ antiporter MdfA (Heng et al., 2015) where these same residues are located inside a central cavity.

5a16f7bc-3e9d-4db6-9810-5d8181a1372a_figure6.gif

Figure 6. Essential residues determined by SDM experiments highlighted in pink on the YqjA model.

5a16f7bc-3e9d-4db6-9810-5d8181a1372a_figure7.gif

Figure 7.

(a) Left - Predicted Contact map with repeating units highlighted in yellow boxes, contact map signature of re-entrant loop packed with TM helix in red boxes.; Right - The Experimental Contact map obtained from the PDB structure with repeating units highlighted in yellow boxes, contact map signature of re-entrant loop packed with TM helix in red boxes. (b) Actual 3orgA topology; grey: TM Helices that are additional to the core; red: TM helices contributing to the formation of the core; orange; re-entrant loops contributing to the formation of the core; green: amphipathic helices contributing to the formation of the core. (c) The 2-fold pseudo symmetry of the amphipathic/re-entrant loop/TM helix core inverted repeat structure of 3orgA with membrane positions shown as grey planes obtained from PDBTM.

Conclusions

This study demonstrates how covariance prediction data have multiple roles in modern structural bioinformatics: not just by acting as restraints for model making and serving for validation of the final models but by predicting domain boundaries and revealing the presence of cryptic internal repeats not evidenced by sequence analysis. Furthermore, we characterised a contact map feature characteristic of a re-entrant helix which may in future allow detection of this feature in other protein families.

Sequence, co-variance and ab initio modelling analyses show that the Pfam PF09335 and PF06695 domains are distantly homologous. These domains contain a structural core composed of a pseudo-inverse repeat of an amphipathic helix, a re-entrant loop and a TM helix. All PF09335 homologues contain this central core with additional TM- helices flanking either side.

Querying the models against the PDB using Dali did not yield any significant hits. However, analysis of the prediction data revealed two features of VTT proteins that independently suggest that they are secondary transporters: both an inverted repeat architecture and the presence of a re-entrant loop, which are both independently and strongly associated with transporter function (Duran & Meiler, 2013; Yan & Luo, 2010). Additionally, the fact that VTT proteins show structural similarities with H+ antiporters indicate that these proteins may also couple substrate transport with an opposing H+ current. Indeed, the Yqja homologue also contains strategically placed residues known to be involved in H+ antiporter activity. The ab initio models show that the essential residues come together in the region that would be buried in the membrane potentially forming a substrate chamber consistent with the transport of a specific substrate. Further research needs to be carried out to determine what this substrate is and confirm the mechanism of transport.

Data availability

All data underlying the results are available as part of the article and no additional source data are required.

Comments on this article Comments (0)

Version 2
VERSION 2 PUBLISHED 03 Dec 2020
Comment
Author details Author details
Competing interests
Grant information
Copyright
Download
 
Export To
metrics
Views Downloads
F1000Research - -
PubMed Central
Data from PMC are received and updated monthly.
- -
Citations
CITE
how to cite this article
Mesdaghi S, Murphy DL, Sánchez Rodríguez F et al. In silico prediction of structure and function for a large family of transmembrane proteins that includes human Tmem41b [version 1; peer review: 1 approved, 2 approved with reservations] F1000Research 2020, 9:1395 (https://doi.org/10.12688/f1000research.27676.1)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.
track
receive updates on this article
Track an article to receive email alerts on any updates to this article.

Open Peer Review

Current Reviewer Status: ?
Key to Reviewer Statuses VIEW
ApprovedThe paper is scientifically sound in its current form and only minor, if any, improvements are suggested
Approved with reservations A number of small changes, sometimes more significant revisions are required to address specific details and improve the papers academic merit.
Not approvedFundamental flaws in the paper seriously undermine the findings and conclusions
Version 1
VERSION 1
PUBLISHED 03 Dec 2020
Views
24
Cite
Reviewer Report 20 Jan 2021
Claudio Bassot, Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Stockholm, Sweden 
Approved with Reservations
VIEWS 24
The authors model ab-initio Tmem41b and homologues, characterizing them as secondary transporters. The models are reliable and the study is scientifically robust and worthy of indexing. 
However, two points are unclear in the text and need to be clarified, ... Continue reading
CITE
CITE
HOW TO CITE THIS REPORT
Bassot C. Reviewer Report For: In silico prediction of structure and function for a large family of transmembrane proteins that includes human Tmem41b [version 1; peer review: 1 approved, 2 approved with reservations]. F1000Research 2020, 9:1395 (https://doi.org/10.5256/f1000research.30592.r75805)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.
  • Author Response 25 Mar 2021
    Daniel Rigden, University of Liverpool, UK
    25 Mar 2021
    Author Response
    The authors model ab-initio Tmem41b and homologues, characterizing them as secondary transporters. The models are reliable and the study is scientifically robust and worthy of indexing. 
    However, two points are ... Continue reading
COMMENTS ON THIS REPORT
  • Author Response 25 Mar 2021
    Daniel Rigden, University of Liverpool, UK
    25 Mar 2021
    Author Response
    The authors model ab-initio Tmem41b and homologues, characterizing them as secondary transporters. The models are reliable and the study is scientifically robust and worthy of indexing. 
    However, two points are ... Continue reading
Views
88
Cite
Reviewer Report 07 Jan 2021
László Dobson, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary 
Gábor Tusnády, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary 
Approved with Reservations
VIEWS 88
In this manuscript Mesdaghi et al. describe the in silico structure modeling of three homologous integral membrane proteins Mt2055, Yqja and human Tmem41b. Structure determination of transmembrane proteins lacks behind globular ones for several reasons, giving space for computational tools. ... Continue reading
CITE
CITE
HOW TO CITE THIS REPORT
Dobson L and Tusnády G. Reviewer Report For: In silico prediction of structure and function for a large family of transmembrane proteins that includes human Tmem41b [version 1; peer review: 1 approved, 2 approved with reservations]. F1000Research 2020, 9:1395 (https://doi.org/10.5256/f1000research.30592.r75806)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.
  • Author Response 25 Mar 2021
    Daniel Rigden, University of Liverpool, UK
    25 Mar 2021
    Author Response
    Major:
    - The authors should be more specific about the exact boundaries of Pfam domains in different proteins as well as the sequence relations of proteins presented in Table 1. ... Continue reading
COMMENTS ON THIS REPORT
  • Author Response 25 Mar 2021
    Daniel Rigden, University of Liverpool, UK
    25 Mar 2021
    Author Response
    Major:
    - The authors should be more specific about the exact boundaries of Pfam domains in different proteins as well as the sequence relations of proteins presented in Table 1. ... Continue reading
Views
49
Cite
Reviewer Report 17 Dec 2020
Pradip Panta, Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA 
William T. Doerrler, Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA 
Approved
VIEWS 49
This work describes the computational structural modeling of a conserved membrane protein family that includes human TMEM41B, a protein with a number of reported functions. Membrane proteins are poorly represented in the structural database and computational methods are increasingly valuable ... Continue reading
CITE
CITE
HOW TO CITE THIS REPORT
Panta P and Doerrler WT. Reviewer Report For: In silico prediction of structure and function for a large family of transmembrane proteins that includes human Tmem41b [version 1; peer review: 1 approved, 2 approved with reservations]. F1000Research 2020, 9:1395 (https://doi.org/10.5256/f1000research.30592.r75807)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.
  • Author Response 25 Mar 2021
    Daniel Rigden, University of Liverpool, UK
    25 Mar 2021
    Author Response
    I would like to begin with nomenclature. I received an email from Dr. Noburo Mizushima several months ago. He has published work on the TMEM41B protein. Also included on the ... Continue reading
COMMENTS ON THIS REPORT
  • Author Response 25 Mar 2021
    Daniel Rigden, University of Liverpool, UK
    25 Mar 2021
    Author Response
    I would like to begin with nomenclature. I received an email from Dr. Noburo Mizushima several months ago. He has published work on the TMEM41B protein. Also included on the ... Continue reading

Comments on this article Comments (0)

Version 2
VERSION 2 PUBLISHED 03 Dec 2020
Comment
Alongside their report, reviewers assign a status to the article:
Approved - the paper is scientifically sound in its current form and only minor, if any, improvements are suggested
Approved with reservations - A number of small changes, sometimes more significant revisions are required to address specific details and improve the papers academic merit.
Not approved - fundamental flaws in the paper seriously undermine the findings and conclusions
Sign In
If you've forgotten your password, please enter your email address below and we'll send you instructions on how to reset your password.

The email address should be the one you originally registered with F1000.

Email address not valid, please try again

You registered with F1000 via Google, so we cannot reset your password.

To sign in, please click here.

If you still need help with your Google account password, please click here.

You registered with F1000 via Facebook, so we cannot reset your password.

To sign in, please click here.

If you still need help with your Facebook account password, please click here.

Code not correct, please try again
Email us for further assistance.
Server error, please try again.