WO2018150260A1 - Method for the identification of mutated proteins having modified thermal stability - Google Patents

Method for the identification of mutated proteins having modified thermal stability Download PDF

Info

Publication number
WO2018150260A1
WO2018150260A1 PCT/IB2018/000194 IB2018000194W WO2018150260A1 WO 2018150260 A1 WO2018150260 A1 WO 2018150260A1 IB 2018000194 W IB2018000194 W IB 2018000194W WO 2018150260 A1 WO2018150260 A1 WO 2018150260A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino acid
protein
acid sequence
target protein
sequence
Prior art date
Application number
PCT/IB2018/000194
Other languages
French (fr)
Inventor
Nicolas REYES
Original Assignee
Institut Pasteur
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Institut Pasteur filed Critical Institut Pasteur
Publication of WO2018150260A1 publication Critical patent/WO2018150260A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1058Directional evolution of libraries, e.g. evolution of libraries is achieved by mutagenesis and screening or selection of mixed population of organisms
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1089Design, preparation, screening or analysis of libraries using computer algorithms

Definitions

  • the invention relates to a method for the identification of mutated proteins having modified thermal stability and in particular having improved thermal stability.
  • the amino acid sequence of the mutated protein results from the substitution of amino acid residues by consensus amino acid residues determined following MSA and specific selection of each consensus residue based on sequence comparison of homologues.
  • mutant amino acid sequences for a target protein using the process of the invention enables the production of mutant proteins having modified thermal stability, in particular improved thermal stability.
  • AS is currently the most established method, and relies on screening the thermal stability of single alanine-mutants at each position in the target protein. This feature makes AS a labour-intensive and costly method, and difficult to apply to large IMP (>500 amino acids residues) or multi-protein complexes. Moreover, AS requires the use of radioactive compounds with high affinity (nano molar) for the target protein, which are not available for most membrane proteins.
  • the DE method relies on a key selection assay for protein expression and stability that requires a small fluorescent compound with high affinity for the protein target, which are not available for most IMP. Moreover, DE in its current stage can only be done using bacterial expression systems, which have been found problematic for most human membrane proteins. [7] The RD approach relies on previous knowledge of atomic-resolution structures of the protein target, which are unknown for most IMP, and requires a great deal of expertise to design the mutagenesis.
  • the process of the invention is based on the stabilizing effect of consensus mutagenesis, and provides major improvements on protein stability with minimal changes of the amino acid sequence compared to the target protein, and without affecting critical residues for function as illustrated for Integral Membrane Protein function.
  • Membrane proteins account for ⁇ 30% of all open reading frames in the human genome 18 They play essential roles in human physiology and pathology that range from synaptic transmission to viral infection 19 20 .
  • their molecular mechanisms or function and pharmacology remain largely unknown. This is mainly due to the tremendous experimental challenges associated to their production, purification and analysis by high-resolution structural and functional approaches 21 -24 Clearly, new approaches and efforts are needed to understand the molecular mechanisms of this important class of proteins.
  • the invention defines a process which is a solution to at least part of the drawbacks of the method disclosed in the art to improve thermal stability of proteins. Accordingly, mutant proteins were designed and obtained that showed improved thermal stability and that could be purified, and proved to be amenable for in vitro structural and functional studies, both of which have been performed successfully.
  • the invention accordingly provides a method to design mutated proteins that harbor improved thermal stability in detergents, wherein said proteins are membrane proteins, especially integral membrane proteins (IMP).
  • IMP integral membrane proteins
  • the invention also relates to mutant proteins, in particular membrane proteins, especially IMP, designed and recovered accordingly.
  • mutant protein in particular mutant IMP, for example mutant transporter
  • mutant transporter retained the functional and pharmacological properties of the Wild Type human protein, in particular IMP, for example transporter, and shares at least 85% amino acid sequence identity with it, more preferably at least 90% amino acid sequence identity with it, and most preferably at least 95% amino acid sequence identity with it. Therefore, it constitutes an excellent structural and pharmacological model of protein, as illustrated for human protein.
  • the method that was developed according to the invention should contribute to better understanding of the molecular basis or function and pharmacology of proteins such as IMP, including those of human origin, and to develop efficient drugs that target them.
  • the method can also be applied to water-soluble proteins, in particular to those used in pharmaceutical and non-pharmaceutical industries.
  • Proteins thermostable in detergent solutions may be useful in various applications, in particular structural analysis and biological analysis.
  • Structural and biological analysis of proteins that requires proteins to be stable in a detergent solution encompass methods such as crystallography (i.e. X-ray crystallography), electron microscopy (i.e. cryo-electron microscopy), protein NMR, X-ray scattering (Small-angle X-ray scattering), circular dichroism, and the like.
  • Methods for the biological analysis or pharmacological analysis encompass ligand-binding screening, affinity/kinetic screening, functional analysis of the proteins, drug targets screening, antagonist/agonist screening, and the like.
  • production of proteins in detergents for crystallization requires a well- ordered crystal. Disposing stable proteins in detergents remains both challenging and necessary in order to have proteins suitable for study of their properties, in particular of their structural or pharmacological properties.
  • the invention accordingly relates to a method of designing a protein with modified thermal stability in a detergent solution with respect to the thermal stability of a target protein comprising: a. providing a multiple sequence alignment (MSA) wherein at least a portion comprising at least 50 % of the full-length amino acid sequence of a target protein is aligned, in particular the full-length amino acid sequence of the target protein is aligned, with the amino acid sequence of homologous sequences of said protein present (i) in other species and/or (ii) in protein variants of the same species, using a determined alignment tool; wherein the homologous sequences are each different from the amino acid sequence of the target protein by at least one amino acid residue in their amino acid sequence, in particular wherein each homologous sequence shares at least 50% identity, more preferably at least 70% identity, with the amino acid sequence of the target protein or with the aligned portion thereof; b. defining for at least part of the positions of the amino acid residues in the target protein, in particular for each amino acid position of the aligne
  • iii a frequency of occurrence which is higher than 10% with respect to the frequency of occurrence of the residue at the same position in the target amino acid sequence when said frequency is evaluated among the aligned amino acid sequences;
  • step b providing a mutated amino acid sequence defining a consensus mutant protein of the target sequence by substituting one or more amino acid residues in the amino acid sequence of the target protein by its(their) respective consensus amino acid residue(s) defined for said each amino acid residue according to step b.
  • the target protein is a membrane protein, in particular an integral membrane protein.
  • the invention also relates to a process to produce a protein with modified thermal stability in a detergent solution with respect to the thermal stability of a target protein comprising: a. providing a multiple sequence alignment (MSA) wherein at least a portion comprising at least 50 % of the full-length amino acid sequence of a target protein is aligned, in particular the full-length amino acid sequence of the target protein is aligned, with the amino acid sequence of homologous sequences of said protein present (i) in other species and/or (ii) in protein variants of the same species, using a determined alignment tool; wherein the homologous sequences are each different from the amino acid sequence of the target protein by at least one amino acid residue in their amino acid sequence, in particular wherein each homologous sequence shares at least 50% identity, more preferably at least 70% identity, with the amino acid sequence of the target protein or with the aligned portion thereof;
  • MSA multiple sequence alignment
  • step b. providing a mutated amino acid sequence defining a consensus mutant protein of the target sequence by substituting one or more amino acid residues in the amino acid sequence of the target protein by its(their) respective consensus amino acid residue(s) defined for said each amino acid residue according to step b.;
  • step c. obtaining a polynucleotide encoding the mutant protein of step c. for expression in a cellular expression system
  • step d. producing the mutant protein from the polynucleotide encoding the mutated amino acid sequence provided in step d. in a cellular expression system;
  • step e evaluating thermal stability in a detergent solution of the mutant protein recovered from the production cells in step e.
  • the target protein is a membrane protein, in particular an integral membrane protein.
  • a portion of an amino acid sequence corresponds to at least 50% of the full-length amino acid sequence of the target protein, more preferably at least 75% of the full length amino acid sequence, more preferably at least 90% of the full-length amino acid sequence.
  • a portion of amino acid sequence may also correspond to a plurality of non-contiguous portions of the full-length amino acid sequence, said plurality representing at least 50% of the full-length amino acid sequence.
  • thermostable mutant proteins by oriented mutations
  • Thermal stability is accordingly the property of the target protein that is primarily altered when performing the process of the invention, in particular that is increased when providing or selecting a mutated sequence of the target protein. Thermal stability may be measured as disclosed hereafter in a denaturation assay assessing resistance to denaturation of the produced mutant protein.
  • the invention thus provides a process for the identification and for the preparation of thermostable mutant proteins and in particular provides a thermostable variant of a target protein.
  • properties may additionally be modified in relation or independently of thermal stability of the target protein in accordance with the process of the invention.
  • properties include the capability of the protein to efficiently keep their three-dimensional structure in detergent solutions and therefore keep their function.
  • production crystal that are well- ordered may involve determination of the capability of said crystal to diffract X- rays to high resolution and comparison to a control. It has been shown indeed that there is a measurable correlation between the stability of a protein, in particular a membrane protein in a detergent and the capability of proper crystallization.
  • the target protein for use in the invention is any protein present in any species.
  • the target protein is membrane protein of such organism, especially an IMP and in particular is a viral envelope protein, or a virus membrane receptor on a cell, especially a human cell.
  • the target protein is a wild-type protein.
  • the target protein is a mutant, in particular a naturally occurring mutant of a wild-type protein, in particular a mutant selected or designed to alter properties with respect to the wild-type protein, possibly in respect of thermal stability or crystallization capability.
  • the target protein may have any length determined in number of amino acids residues.
  • the protein has at least 300 amino acid residues, in particular at least 400 or 500 amino acid residues, but the method can also be performed with a target protein having less than 300 amino acid residues.
  • the protein has at least 1300 amino acid residues.
  • the protein comprises between 300 amino acid residues and 1500 amino acid residues, and more advantageously, the protein comprises between 350 and 1400 amino acid residues. It should be noted that the methods of the prior art are increasingly cumbersome with the length of the amino acid sequence of the target protein. The method disclosed therein allows the efficient production of thermal stable proteins, irrespective of their number of amino acid residues.
  • the target protein is one the amino acid sequence of which is known and especially disclosed in databases such as GenBank.
  • the amino acid sequences of the homologous sequences of the target protein are known and in particular disclosed in databases such as GenBank.
  • the amino acid sequences used for the performance of the alignment do not include a defined consensus sequence i.e., an artificial archetypal sequence defined using sequence data obtained from various known natural products but are rather representatives of isolated proteins.
  • the target protein and the homologous proteins are wild-type sequences.
  • the multiple sequence alignment follows a first round or mutations in the amino acid sequence of the target protein, such as function-improving mutation(s) and/or structure-improving mutation(s).
  • the amino acid sequence of the target protein may be an artificial sequence, i.e., a sequence that has been mutated with respect to a naturally occurring protein in order to modify properties of the target protein.
  • step c. of providing a mutated amino acid sequence defining a mutant protein of the target is followed by a further step of mutation in the sequence in order to alter other properties of the target protein.
  • the alignment performed according to the process of the invention is a (MSA) which as such is well known to the person skilled in the art. It may be performed through available tools including a suitable algorithm (such as Clustal W) and software, especially using an alignment software. Many software suitable to perform alignments are available such as JALVIEW software or others listed on https://en.wikipedia.org/wiki/List_of_sequence_alignment_software. In a particular embodiment of the invention, the determined alignment tool used for the MSA is JALVIEW software. https://en.wikipedia.org/wiki/List_of_sequence_alignment_software
  • homologous sequences of the target protein may be found in species different from the species providing the target sequence. They may additionally encompass or alternatively consist in variants of the target sequence available in the same species. In particular, when the target protein is a human protein, the homologous sequences may originate from other vertebrate species, in particular from other mammal species. They may also encompass variants of the target sequences disclosed in human or in other vertebrate species. A homologous sequence may also be defined as a variant of the target protein having at least 50% identity with the target protein, in particular at least 70% identity, whether wild-type or mutated as disclosed above.
  • each selected homologous sequence for the MSA has a difference with the target sequence such that both sequences share less than 95% sequence identity when aligned with the MSA tool.
  • step b. of the process the consensus amino acid residue may be defined for each amino acid position in the amino acid sequence of the target protein (also designated target sequence).
  • consensus amino acid residue may be defined for at least part of the amino acid residues in the target sequence and in particular for at least 25% or at least 50% or at least 75% or at least 90% of the amino acid positions in the target sequence.
  • the amino acid residues targeted for substitution by the consensus amino acid residues are contained in secondary structural elements of the target protein, such as in predicted or known (characterized) a-helices or in ⁇ -turns.
  • the amino acid residues targeted for substitution by the consensus amino acid residues are contained in loops, in particular in unstructured loops connecting the other structured regions.
  • such secondary structural elements and/or loops are involved in the alignment step performed on portion(s) of the amino acid sequence of the target protein.
  • a mutated amino acid sequence may be defined that results from the substitution of all amino acid residue(s) of the target sequence for which a consensus residue has been identified.
  • the thus obtained amino acid sequence defines a mutant protein of the target protein.
  • the mutated protein has a sequence identity with the target protein which is at least 80%, more preferably at least 85%, more preferably at least 90%, most preferably at least 95%, in order to minimize the effects of the mutations on the properties of the target protein.
  • Identity means to the extent to which two amino acid sequences have the same residue at the same position in an alignment.
  • Identity could be defined as the number of exact matches in the alignment of two sequences (the target protein and a mutated protein), divided by the length of the target protein or the length of the aligned portion of the target protein. An exact match occurs when the target protein and the mutated protein have identical amino acid residues in the same position.
  • a polynucleotide can be designed, especially by mutation of the polynucleotide encoding the target protein, for the expression of the mutant protein in a cellular expression system.
  • the polynucleotide encoding the mutant protein having the mutated amino acid sequence may further be mutated for codon optimization in accordance with methods well known from the person skilled in the art for its expression in the selected cellular expression system.
  • This polynucleotide may further be modified to add a sequence that encodes a purification handle at the terminus of the amino acid sequence of the mutant protein.
  • the invention thus relates to the mutated protein which is produced and whose amino acid sequence consists in the consensus mutated sequence of the target protein.
  • the invention also relates to the polynucleotide encoding the consensus mutated protein and to cells transformed with such polynucleotides in conditions enabling the expression of the polynucleotide as a mutated protein with respect to the target protein.
  • the process may additionally comprise, after step c. a further step (c(i)) of determination according to an additional parameter of the amino acid residues in the target sequence that may be mutated.
  • step c(i) is a step of calculating pairs of residues that co-evolve in the target protein and substituting these pairs of residues in the target sequence.
  • Step c(i) may be carried out using co-evolution software suitable for the determination of amino acid pairs that co-evolve such as
  • the additional but optional c(i) step has particular interest in situations where at the end of step c the mutant protein obtained has 80% or less sequence identity with the target sequence.
  • the co-evolution determination brings the advantage of pointing to amino acid residues that co-evolved in the 3-dimensional structure of the protein rather than in its amino acid sequence.
  • this step c(i) may be performed when the number of homologous sequences available for the MSA is at least 5 times the number of amino acid residues of the target sequence.
  • the homologous sequences may have to be found in species which are remote from the species of the target sequence, such as in prokaryotic homologues when the target protein is found in a eukaryotic species such as in human, and/or in divergent branches of the family protein of the target protein.
  • step c(i) the identity of the mutations to be introduced in the target sequence is still based on step c. but the additional step ci enables to lessen the number of residues to be mutated.
  • the obtained mutant sequence may thus be in the range of sequence identity of at least 90%, preferably at least 95% with the target sequence.
  • a variant (i.e. consensus mutant) protein obtained after step c and optionally c(i) may be prepared or produced in a cellular expression system and recovered for testing, in particular for thermal stability testing in detergent solution.
  • the target protein is a membrane protein, in particular a eukaryotic, such as a mammalian, especially a human, membrane protein.
  • the membrane protein is an integral membrane protein.
  • the membrane protein is selected among channels, enzymes, and primary or secondary active transporters.
  • Transporters may in particular be of the SLC1 transporters family which are ion- coupled amino acid transporters, and may comprise divergent branche(s) of this family, like sodium-dependent neutral amino acid transporters (ASCT1 -2).
  • the target protein is a soluble protein, especially a globular soluble protein.
  • the study of thermal stability of the mutant protein aims at determining whether mutations have improved, i.e., increased thermal stability in detergent solution.
  • a produced, especially crystallized mutant protein having improved thermal stability shows resistance to denaturation when the melting temperature is raised for at least 2°C, advantageously for at least 3 to 10°C and in a particular embodiment up to 20°C with respect to the melting temperature of the target protein.
  • the detergent solution is one known from the person skilled in the art in biochemistry, in particular one suitable for use in production, purification or analysis, especially by high-resolution structural and functional approaches.
  • Detergent solutions may in particular be those used for preparation of protein for structure determination or biological function analysis; for example detergent used for X-ray structure or crystallization.
  • detergent solutions encompass small micelle detergents, dodecyl-p-D-maltopyranoside (12M), n- dodecyl-N,N-dimethylamine-N-oxide (LDAO), n-nonyl-p-D-maltopyranoside (9M), nonyl-p-D-glucopyranoside (NG), n-decyl-p-D-maltopyranoside.
  • Detergent solutions may in particular be those used for preparation of protein for biophysical analysis and functional analysis using fluorescence spectroscopy (dodecyl-p-D- maltopyranoside, dodecyl-p-D-glucopyranoside, 7-Cyclohexyl-1 -Heptyl-p-D-
  • Maltoside as well methods for determination of high-resolution three- dimensional structures in particular X-ray crystallography and single-particle electron microscopy (dodecyl- decyl-, nonyl-, or octyl-p-D-maltopyranoside; dodecyl-, decyl-, nonyl-, or octyl-p-D-glucopyranoside; Neopentyl glycol based detergents like 2,2-dioctylpropane-1 ,3-bis-p-D-glucopyranoside).
  • Thermal stability of proteins may in general be characterized by the difference between the folded and unfolded states of the protein measured in kcalmol "1 . Measurement of stability may encompass thermal denaturation of the protein in a detergent solution and measurement by the increase of the apparent melting temperature of the mutant protein which is then compared to the melting temperature of the target protein by chromatographic observation, in particular by size-exclusion chromatography (SEC-TS).
  • SEC-TS size-exclusion chromatography
  • protein stability may be determined with or without ligand.
  • a sample of the purified protein is heated and then cooled and centrifuged to remove aggregated proteins and run on analytical HPLC. When the melting temperature is reached and proteins precipitates or aggregates, pic height decreases and void pic height increases.
  • the mutant protein when expressed in a cellular system, is fused to a detectable marker, such as a fluorescent marker, for the evaluation of the melting temperature of the mutant protein.
  • a detectable marker such as a fluorescent marker
  • the expression of the mutant protein may be performed in any available cellular system.
  • the protein is a eukaryotic protein, it is advantageously produced in a eukaryotic cell.
  • the process of the invention enables selecting and producing proteins having improved thermal stability with respect to the target protein.
  • the invention also relates to a process for modifying, in particular for improving thermal stability of a protein comprising carrying out consensus mutagenesis in the amino acid sequence of a target protein using the process defined herein in accordance to its embodiments and recovering the mutant protein having a modified, in particular improved thermal stability.
  • the invention relates to a process to identify an amino acid sequence which is mutated with respect to the amino acid sequence of a target protein said process comprising the steps of: a. providing a multiple sequence alignment (MSA) wherein at least a portion comprising at least 50% of the full-length amino acid sequence of a target protein is aligned, in particular wherein the full- length amino acid sequence of a target protein is aligned, with the amino acid sequence of homologous sequences of said protein present (i) in other species and/or (ii) in protein variants of the same species, using a determined alignment tool; wherein the homologous sequences are each different from the amino acid sequence of the target protein by at least one amino acid residue in their amino acid sequence, in particular wherein each homologous sequence shares at least 50% identity, more preferably at least 70% identity, with the amino acid sequence of the target protein or with the aligned portion thereof,
  • MSA multiple sequence alignment
  • step b providing a mutated amino acid sequence defining a mutant protein of the target sequence by substituting one or more amino acid residues in the amino acid sequence of the target protein by its(their) respective consensus amino acid residue(s) defined for said each amino acid residue according to step b.
  • the invention also relates to a process for the preparation of a protein with modified crystallization capability comprising preparing a protein having modified thermal stability in a detergent solution according to any of the embodiments of the process of the invention.
  • An optional step of further mutation may be performed after step c. and in particular before step d. of the process to prepare the mutant protein to further improve crystallization capability, such optional step of further mutation being in particular a step of mutation in the extracellular region of the target protein.
  • the process is performed, wherein the target protein is the Excitatory Amino Acid Transporter 1 (EAAT1 ) and the obtained mutant protein is EAAT1 of sequence SEQ ID No.1 SEQ ID No.2, SEQ ID No.3 or SEQ ID No.4.
  • EAAT1 Excitatory Amino Acid Transporter 1
  • the homologous sequences aligned within the MSA may comprise at least one sequence selected from the group comprising SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10 and SEQ ID No: 1 1 .
  • the homologous sequences alignment within the MSA comprise at least the following sequences: SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10 and SEQ ID No: 1 1 .
  • the invention thus also concerns a mutant protein of Excitatory Amino Acid Transporter 1 (EAAT1 ) the amino acid sequence of which is SEQ ID No.1 SEQ ID No.2, SEQ ID No.3 or SEQ ID No.4.
  • EAAT1 Excitatory Amino Acid Transporter 1
  • the process is performed, wherein the target protein is NPC1 L1 (Niemann-Pick C1 -Like 1 ), in particular human NPC1 L1 , in particular NPC1 L1 of Accession number Q9UHC9.2 (SEQ ID No: 12).
  • NPC1 L1 Naemann-Pick C1 -Like 1
  • human NPC1 L1 in particular NPC1 L1 of Accession number Q9UHC9.2 (SEQ ID No: 12).
  • Various isoforms of human NPC1 L1 , or ortholog proteins may be used in step a.
  • Niemann-Pick C1 - like protein 1 from Canis lupus familiaris (Accession number NP_001091019.1 - SEQ ID No: 14), from Bos Taurus (Accession number XP_588051 .4 - SEQ ID No: 15), from Loxodonta Africana (Accession number XP_003418622.1 - SEQ ID No: 16), from Mus musculus (Accession number Q6T3U4.1 - SEQ ID No: 17), from Monodelphis domestica (Accession number XP_001379744.2 - SEQ ID No: 18), from Dasypus novemcinctus (Accession number XP_004467651 .1 - SEQ ID No: 19), from Xenopus tropicalis (Accession number XP_002932620.2 - SEQ ID No: 20), from Oryzias latipes (Accession number XP_00293262
  • At least one of the above cited sequence may be aligned within the MSA when the target protein is human NPC1 L1 .
  • SEQ ID No: 12 to SEQ ID No: 25 may be aligned within the MSA when the target protein is human NPC1 L1 .
  • the process is performed, wherein the target protein is the Niemann-Pick C1 -like1 (NPC1 L1 ) and the obtained mutant protein is NPCL1 of sequence SEQ ID No.13.
  • the target protein is the Niemann-Pick C1 -like1 (NPC1 L1 ) and the obtained mutant protein is NPCL1 of sequence SEQ ID No.13.
  • the invention thus also concerns a mutant protein of Niemann-Pick C1 -like1
  • NPC1 L1 amino acid sequence of which is SEQ ID No.13.
  • the invention relates to a liposome or a collection of liposomes containing a mutant protein obtained from carrying out the process according to the invention.
  • the invention also relates to a cell expressing a mutant protein obtained from the process of the invention.
  • the invention relates to a cell or a liposome comprising a mutant protein of Excitatory Amino Acid Transporter 1 (EAAT1 ) the amino acid sequence of which is SEQ ID No.1 SEQ ID No.2, SEQ ID No.3 or
  • the invention relates to a cell or a liposome comprising a mutant protein of Niemann-Pick C1 -Iike1 (NPC1 L1 ) the amino acid sequence of which is SEQ ID No.13.
  • the process of the invention may be followed by additional steps of mutation in the mutant protein such as mutations that enable the improvement of purification or crystallization of the protein.
  • FIG. 1 Size exclusion chromatograms of detergent-solubilized GFP-fusions of EAATI WT and its thermostable variants at different temperatures from clear lysates of HEK293 cells (upper plots).
  • the apparent melting temperature (T m ) was estimated from the GFP-fluorescence value of the peak corresponding to the trimeric protein (lower plots).
  • FIG. 3 Uptake of radioactive L-glutamate by purified ⁇ 1 ⁇ (left), and ⁇ 1 ⁇ 2 (right) reconstituted in liposomes. Purified EAATI WT did not show significant levels of uptake above the control liposomes without reconstituted transporters (see Fig. 3a).
  • UCPH101 inhibits glutamate transport EAATl cryst (lower plot), but not in EAAT1 C ryst-ii (upper plot) in a concentration dependent manner (b).
  • Plots depict an average of three independent experiments performed with duplicate measurements, and error bars represent s.e.m. c-d, Structure of EAATI cryst trimer viewed from the extracellular solution (c) and from the membrane (d), with the ScaD and TranD as surfaces, and U PCH101 bound between them (spheres), e, EAATI cryst monomer viewed parallel to the membrane.
  • the ScaD domain is represented as surface, and several helices and loops in the TranD have been removed for clarity of display, f, Domain organization diagram of EAATI cryst monomer.
  • FIG. 4 Alignment of human SLC1 transporters. Amino acid sequences of EAAT1 -5, ASCT1 -2 and EAATI cryst are compared. The boundaries of the a-helices (cylinders) in the TranD and the ScaD seen in the EAATI cryst structure are shown. In order to confer crystallizability, the region between TM3 and TM4c (arrows) from ASCT2 was transferred to a thermally stabilized EAAT1 . To further improve crystal formation in the absence of U PCH101 , mutations M231 1 and F235I (circles) were introduced to generate EAAT1 cryst-n. These substitutions are found in EAAT2. Other residues involved in U PCH101 coordination are more conserved (triangles). Sequences were aligned with Jalview.
  • FIG. 1 UCPH101 binding site, a, Lateral view of EAATI cryst monomer from the membrane showing UCPH101 bound between the TranD and ScaD.
  • b-c UCP H101 coordination and Fo-Fc densities contoured at 2.0o in EAATI cryst (b) and EAAT1 C ryst-ii (c), respectively.
  • Side chains of residues in TM3, TM4c, and TM7 involved in coordination are shown.
  • F369 side chain moves outward in the EAAT1 C ryst-ii unbound state (c).
  • UCPHioi-substrate bound structures moving the carbonyl oxygen of A420 away from Na2 (black sphere), c, Omit map Fo-Fc (68) density for the TBOATFB molecule is contoured at 2.3o (black mesh), and some of the residues at Van deer Waals or H- bond distance from the compound are represented as sticks.
  • EAAT1 consensus mutants, a-b, Residues exchanged for consensus amino acids in ⁇ 1 ⁇ (a) and ⁇ 1 ⁇ 2 (b) are mapped into the structure of the EAATI CRYST (PDB 5LLM) trimer viewed from the extracellular medium (left panel), as well as the scaffold (cyan) and the transport (orange) domains viewed from the membrane. These domains are depicted separately for clarity of display, including two views of the transport domain separated ⁇ 180° from each other, showing its interface with the scaffold domain (left) and the membrane (right), respectively.
  • PDB 5LLM EAATI CRYST
  • Spheres correspond to the alpha carbon atoms of residues that were exchanged by conservative (grey) and non-conservative (black) consensus mutations, c-d, Radioactive L-glutamate uptake in cells expressing transporters (c), including control cells transfected with a vector lacking EAAT1 genes, and in liposomes with purified reconstituted transporters (d).
  • Yellow circles depict the liposomal bilayer separating sodium- (Na + ), potassium- (K + ), and choline-based (Ch + ) solutions, e, Rate of L-glutamate uptake by purified ⁇ 1 ⁇ (blue) and ⁇ 1 ⁇ 2 (red) reconstituted in liposomes, as a function of L-glutamate concentration.
  • Figure 8 Deuterium exchange at 20 °C. a-c, Deuterium uptake kinetics at 20 °C of examples peptides covering both helical and non-helical regions of EAATI WT (grey), ⁇ 1 - ⁇ (blue), and ⁇ 1 ⁇ 2 (red), respectively.
  • Solid lines represent double- exponential fits to the data, and dotted lines the expected deuterium kinetics of unfolded and solvent exposed peptides, d-f Deuterium incorporation after 1 h at 20 °C in EAAT1 TI (d), EAAT1 ⁇ 2 (e), and EAATI WT (f) mapped into the structure of the EAAT1 cRYTs (PDB 5LLM) trimer viewed from the extracellular medium (upper panel), as well as the scaffold (ScaD) and the transport (TranD) domains viewed from the membrane (lower panel), respectively. These domains are depicted separately for clarity of display. In the trimeric depiction, arrows point to the interface between protomers. Deuterium incorporation was calculated as an average of three experiments, and normalized to the maximal theoretical incorporation based on the number of backbone amide available for exchange in each peptide. A scale bar representing deuterium incorporation is depicted (d).
  • FIG. 9 Melting of trimeric transporters, a-c, Size-exclusion chromatograms of purified ⁇ 1 ⁇ (a), ⁇ 2 (b), and the chimeric transporter EAAT1 -ScaD-n-TranDT2 (c), respectively, pre-heated at different temperatures. Chromatograms show how the trimeric form of the transporters that elutes at ⁇ 3.0 ml melts into lower oligomeric state(s), most likely monomers, that elute at ⁇ 3.5 ml.
  • d-f Melting curves depicting the change in fractional area of the chromatographic peak corresponding to the trimeric transporters (black symbols), as a function of the temperature pre-pulse in ⁇ 1 ⁇ (d), ⁇ 2 (e), and EAAT1 -ScaD-n-TranDT2 (f), respectively.
  • Solid lines indicate fits of a Hill-like equation (see methods) to the data with TSO-FSEC values 49.3, 38.6 and 45.7 °C, and n values -21.5, -9.3, and -18.3 for EAAT1 T 1 , EAAT1 T2 , and EAAT1 - ScaD-n-TranDT2, respectively.
  • the total area under the chromatogram at each temperature, normalized to that at 4 °C, is also shown (empty symbols), and remains relatively constant at all temperatures.
  • FIG. 10 Bimodal m/z envelopes, a-b, m/z envelopes of an example peptide covering residues 174-184 of ⁇ 1 ⁇ (a), ⁇ 2 (b), at different pre-pulse temperatures.
  • Solid symbols represent the average of three experiments and error bars represent s.e.m, and are superimposed on the m/z spectrum of a representative experiment.
  • Solid lines represent fits of a double Gaussian equation to the data, as well as their low- and high-m/z components, c-d, melting plots of the low-m/z component of peptides covering residues 174-184 (c) and 187-194 (d) of ⁇ 1 ⁇ (solid circles), ⁇ 2 (solid triangles).
  • Solid lines indicate fits of a Hill-like equation to the data with T50-HDX-GAUSS values °C, and n values for peptide 174-184 in ⁇ 1 ⁇ and ⁇ 1 ⁇ 2, respectively, and TSO-HDX-GAUSS values °C, and n values for peptide 187-194 in ⁇ 1 ⁇ and EAAT1 T2, respectively.
  • Figure 1 1 Local thermal unfolding, a-b, Deuterium uptake kinetics at different pre- pulse temperatures of an example peptides containing residues 174-184 in ⁇ 1 ⁇ (a), and ⁇ 1 ⁇ 2 (b), respectively. Solid lines represent double-exponential fits to the data, and dotted lines the expected deuterium kinetics of the unfolded and solvent exposed peptide. Plots in a-b depict an average of three experiments, and error bars represent s.e.m.
  • the inset represents the temperature protocol used to prepare the protein samples, c-d Apparent melting temperatures of the peptides folded state in EAAT1 TI (C), and EAAT1 T 2 (d) are mapped into the structure of the EAATI CRYST (PDB 5LLM) trimer viewed from the extracellular medium (upper panel), as well as the scaffold (ScaD) and the transport (TranD) domains viewed from the membrane (lower panel), respectively. These domains are depicted separately for clarity of display.
  • the color code representing the apparent melting temperature values is depicted in a scale bar.
  • FIG. 12 Size exclusion chromatograms of detergent-solubilized GFP-fusions of NPC1 L1WT (front, row 1 ) and NPC1 L1 T 1 (back, row 2) from lysates of HEK293 cells.
  • Figure 13. Alignment of ⁇ 1 ⁇ , ⁇ 1 ⁇ 2 and EAATI WT. Amino acid residues of EAAT1 TI , EAAT1 T2 and EAATI WT are compared. The boundaries of the a-helices (cylinders) in the TranD and the ScaD issued from the structural analysis of the EAAT1 CRYST are shown. ⁇ 1 ⁇ and EAATI WT share 465 amino acid residues (85 % identity), while ⁇ 1 ⁇ 2 and EAATI WT share 513 amino acid residues (95% identity).
  • Example 1 THERMAL STABILIZATION OF HUMAN EXCITATORY AMINO ACID TRANSPORTER 1 FOR IN VITRO STRUCTURAL AND FUNCTIONAL STUDIES
  • This Example describes a process to improve the human Excitatory Amino Acid Transporter 1 (EAAT1 ) thermal stability in detergent solutions, as well as the determination of the X-ray structures of EAAT1 thermostable variants.
  • EAAT1 Human Excitatory Amino Acid Transporter 1
  • amino acid mutations that increase thermal stability have been introduced in EAAT1 that make the transporter amenable for purification in detergent solutions, while preserving the neurotransmitter transport function of the wild type protein.
  • This feature allowed to solve the first three-dimensional structures of thermostable EAAT1 variants and unravel a novel allosteric mechanism of inhibition.
  • Human members of the solute carrier 1 (SLC1 ) family of transporters take up excitatory neurotransmitters in the brain and amino acids in peripheral organs. Dysregulation of their functions is associated to neurodegenerative disorders and cancer.
  • the first crystal structures are presented for a human SLC1 transporter thermostabilized according to the method of the invention, the excitatory amino acid transporter 1 (EAAT1 ), with and without allosteric and competitive inhibitors bound.
  • the structures show novel architectural features of the human transporters, including intra- and extracellular domains with potential roles in transport function, as well as regulation by lipids and post-translational modifications.
  • SLC1 transporters constitute a large family of ion-coupled amino acid transporters present in all kingdoms of life 1 .
  • SLC1 transporters that share 40-70% amino acid identity (Fig. 4) and have evolved to serve two specialized functions 2 : in the central nervous system, SLC1 excitatory amino acid transporters (EAAT1 -5) take up the neurotransmitter glutamate into the cell.
  • EAATs take up glutamate and aspartate, while neutral amino acid transporters (ASCT1 -2) exchange small amino acids between the extra- and intracellular compartments, contributing to the cellular solute homeostasis.
  • Glutamate is the most important excitatory transmitter in the mammalian brain and is involved in most aspects of brain physiology, from development to cognition 3 .
  • most of the glutamate in the brain is intracellular, and it has to be continuously pumped into the cytoplasm to allow for rounds of transmission and prevent cytotoxicity.
  • This essential neurological function is done by the EAAT1 -5 isoforms expressed at the plasma membrane of astrocytes and neurons.
  • astroglial EAAT1 and EAAT2 orthologs are highly expressed in the hind- and forebrain, respectively, and are responsible for most of the glutamate uptake in the rodent brain 4 .
  • EAATs are powerful molecular pumps capable of maintaining up to 10 4 -fold glutamate gradients by using energy stored in sodium, proton and potassium gradients 5 .
  • their dysregulation has been associated with several neurological diseases, including amyotrophic lateral sclerosis 6 , ataxia 7 8 , stroke 9 , depression 10 and glioma 11 , making them important drug targets.
  • they are also expressed in intestine and kidney, where mutations in EAAT3 have been associated to dicarboxylic aminoaciduria 12 .
  • ASCTs are structurally similar to EAATs, and function as sodium-dependent neutral amino acid exchangers at the plasma membrane 13 . They are highly expressed in intestine, kidney and testis, where they play a key role in maintaining the amino acid cellular homeostasis. Importantly, ASCT2 is up-regulated in several forms of cancer, including melanoma 14 , lung 15 , prostate 16 and breast cancer 17 , and it is a key drug target for the treatment and diagnosis of these diseases.
  • the process used and disclosed herein to prepare stable and functional EAAT1 is general, fast, and cost-effective, and can be applied to other human membrane protein families. Moreover, with small variations the method can be applied to membrane proteins from other species, in particular pathogenic bacteria and viruses, or even to globular soluble proteins of different origin.
  • a consensus residue was defined when it met all the below criteria: i) the residue had the highest frequency of occurrence among the aligned sequences, and this frequency is >20%; ii) the residue occupied a position in the alignment in which gaps constitute ⁇ 30% of the entries; iii) the residue's frequency of occurrence differed by more >10% from the corresponding frequency of the residue in the target/parent sequence (human sequence: EAATI WT);
  • EAAT1 homologous transporter EAAT1 thermostable 1 (EAAT1 TI) has been generated using this approach and has the following sequence: Sequence ID No.1
  • EAAT1 TI is 85% identical and 92% similar to EAAT
  • an additional step consisted in calculating pairs of residues that strongly co-evolved, using EVcouplings software 67 (http://evfold.org/evfold-web/evfold.do) (or others having similar functionality), and substituting these pairs of residues in the target sequence for consensus residues.
  • this additional step is performed so that the substitutions performed in step c) are limited to co-varying residues.
  • the EAAT1 homologous transporter EAAT1 thermostable 2 ( ⁇ 1 ⁇ 2) has been generated using this approach and has the following sequence:
  • the homologous transporters ⁇ 1 ⁇ and ⁇ 1 ⁇ 2 have higher thermostability in detergent solutions and showed an increase in the apparent melting temperature (T m ) of ⁇ 13, and ⁇ 5 °C, respectively (Fig. 1 ), compared to EAATIWT.
  • the homologous transporters ⁇ 1 ⁇ and ⁇ 1 ⁇ 2 have conserved transport mechanism (i.e. functional) are when expressed in cells (Fig. 7c). In other words, these mutants are functional.
  • the homologous transporters ⁇ 1 ⁇ and ⁇ 1 ⁇ 2 also have a conserved transport mechanism (i.e. are functional) upon reconstitution of purified transporters in liposomes (Fig. 7d and 7e).
  • ⁇ 1 ⁇ and ⁇ 1 ⁇ 2 also have a good solubility in detergent solutions.
  • ⁇ 1 ⁇ shows similar level of neurotransmitter uptake in cells compared to EAATIWT, substantially higher solubilization yields in all detergents tested, and robust glutamate uptake upon purification and reconstitution in synthetic liposomes (Fig. 7d).
  • glutamate transport in liposomes was strictly dependent on opposite gradients of sodium and potassium across the bilayer, and the rate of transport was dependent on the concentration of L-glutamate, with a K m ( ⁇ 30 ⁇ ) similar to the one reported for EAATIWT (Fig. 7e). The transport mechanism is therefore conserved for ⁇ 1 ⁇ and EAAT1 T2.
  • the neurotransmitter experiment shows that the detergent solutions used for solubilization and purification induce unfolding events that render the EAAT1 WT inactive, while ⁇ 1 ⁇ and ⁇ 1 ⁇ 2 remain functional.
  • the homologous transporters ⁇ 1 ⁇ and ⁇ 1 ⁇ 2 but not EAATI WT, could be purified as properly folded transporters and reconstituted in liposomes as functional proteins (Fig. 2).
  • the stable and functional purified transporters could be used in high-resolution structural studies and would be suitable candidates for high- throughput screening of small compounds using in vitro techniques for drug discovery.
  • purified ⁇ 1 ⁇ and ⁇ 1 ⁇ 2 can be used to develop monoclonal molecules, like antibodies and nanobodies that recognize the transporters in the cell surface using three-dimensional epitopes, as opposed to the linear and intracellular epitopes currently used. Mutations based on consensus amino acids of animal SLC1 transporters kinetically trapped EAAT1 in folded and functional states, and allowed the analysis of intermediate unfolded states along their denaturation pathways.
  • thermostable EAAT1 variants in complex with a substrate (L-aspartate), and the allosteric inhibitor UCPH101 are presented.
  • the structures, and supporting functional data show new architectural features of the EAATs and ASCTs, and unravel the allosteric mechanism of UCPH101- like inhibitors in atomic detail. Taken together, these structural data can prove useful for the design of novel allosteric compounds with improved selectivity for both EAATs and ASCTs.
  • Purified wild-type EAAT1 shows a poly-disperse size-exclusion chromatogram in detergent solutions, and lacks transport activity upon reconstitution in synthetic liposomes (Fig. 3a).
  • consensus mutations 26 were introduced in the predicted transmembrane helices, and obtained a biochemically stable and functional transporter (Fig. 4).
  • TM3 predicted transmembrane helix 3
  • TM4c cytoplasmic half of TM4
  • EAATI cryst shares an overall ⁇ 75% sequence identity with wild type EAAT1 , and up to ⁇ 90% identity at the C-terminal core of the protein, where the transported substrate and coupled ions are expected to bind 25 27"31 .
  • purified EAATI cryst reconstituted in liposomes showed robust glutamate uptake that depended on opposite gradients of sodium and potassium ions across the bilayer (Fig.
  • EAATI cryst formed crystals in the presence of UCPH101 that diffract X- rays anisotropically and up to 3.25 A resolution, and its inhibitor-bound structure was solved by molecular replacement (see Methods and Table 1 ).
  • EAATI cryst was refractory to crystallization in the absence of UCPH101 , it was reasoned that mutations in the inhibitor-binding pocket could aid with the crystallization of the transporter.
  • a construct carrying M231 I and F235I mutations (EAAT1 C ryst-ii; Fig. 4), crystallized both in the presence and absence of the inhibitor, and diffracted X-rays up to 3.1 and 3.32 A resolution, respectively (Extended Data Table 1 ).
  • EAAT1 cr yst shows a symmetric homotrimer in a substrate- and UCPHi 01 -bound outward-facing conformation (Fig. 3c-e).
  • EAATI cryst adopts an overall GltPh-like fold 22 23 , in which each monomer is composed of two domains: a trimerization or scaffold domain (ScaD), including TM1 -2 and TM4-5; and a transport domain (TranD), including TM3, TM6-8 and re-entrant helical loops 1 -2 (HP1 -2; Fig. 3f).
  • the ScaD forms all inter-subunit contacts through residues in TM2, TM4 and TM5, that include six salt bridges and bury ⁇ 3,000 A 2 from each subunit.
  • the three ScaDs form a compact central structure with a propeller-like shape that ensures the trimeric form of the transporter and anchors it to the membrane.
  • the three TranDs are more peripheral and localize between the blades of the propeller, making protein contacts exclusively with the ScaDs of their own monomer.
  • the TranD-ScaD interface buries ⁇ 3,500 A 2 , including a conserved salt bridge between E256 and K364. This interface is mainly formed by cytoplasmic residues in HP1 , TM7, and TM3 (TranD), and TM2, TM4c, and TM5 (ScaD). However, on the extracellular side additional contacts occurred between HP2 and TM4, through residues that are well conserved among human transporters.
  • TM8 can be divided into extracellular (TM8a), transmembrane (TM8b), and cytoplasmic (TM8c) helices.
  • TM8a extracellular
  • TM8b transmembrane
  • TM8c cytoplasmic
  • HP2 is a key component of the gating machinery that controls the access of substrate and ions to their binding sites in the TranD 25 33-35 , and its interactions with TM8b likely play an important role in determining HP2 movements.
  • TM8a in EAAT1 36
  • EAAT2 ortholog 37 impaired glutamate transport, highlighting the significance of this extracellular region for function.
  • TM8b high electron density for the substrate (L-aspartate) was found and one of the sodium-binding sites previously observed in the archaeal homologs of SLC1 family (Na2), which was modeled with similar coordination than in their structures 25 31 .
  • the carboxylate group of D456 (TM8b) which coordinates the alpha-amino group of the substrate, is also at hydrogen bond distance with the hydroxyl group of S343 (HP1 ).
  • the guanidinium group of R457 (TM8b) engages in hydrogen bonding with HP1 residue G341 , and possibly L340 and T342 that point their backbone carbonyl oxygen atoms towards TM8b.
  • Residues S343 and R457 are well conserved in human SLC1 transporters, and substitutions at equivalent positions in EAAT1 (S363 and R477) 38 , and EAAT3 (R445) 39 inactivate transport.
  • the loss-of-function mutation R445W in EAAT3, equivalent to R457 in EAATI cryst causes human dicarboxylic aminoaciduria due to the lack of aspartate/glutamate reabsorption function in the kidney 12 .
  • the functional studies and our structural data converge to suggest that interactions between conserved human residues at HP1 and TM8b are important to the correct folding and function of the transporters.
  • hydrophilic TM8c extends beyond the membrane plane through a hydrophilic helix (TM8c), and makes contact with residues in TM3 and TM7a.
  • EAAT2 deletion mutants in this region have a deleterious effect on transport function and membrane trafficking 40 .
  • a deletion of TM8c beyond E501 in EAAT1 (equivalent to E500 in EAAT2 and E481 in EAATI cryst) decreased glutamate uptake rate by ⁇ 2-fold.
  • the functional data, and the amino acid conservation in TM8c among EAATs, underscore the pivotal role of this structural motif in protein folding and transport kinetics.
  • the ScaD is less conserved than the TranD in the SLC1 family.
  • the ScaD is less conserved than the TranD in the SLC1 family.
  • TM4 is highly divergent (Fig. 4), and shows several unique architectural features in EAATI cryst.
  • TM4a forms inter- and intra-monomeric contacts with TM2 and HP2, respectively.
  • TM4b-c loop amino acid insertion between TM4b and TM4c that appeared during the evolution of eukaryotic transporters protrudes into the central vestibule of the EAAT1 cr yst trimer.
  • the TM4b-c loop forms the center of the propeller, and makes extensive contacts within and between protomers.
  • TM4b-c loop Due to the lack of electron density it was not possible to model the outermost residues of the TM4b-c loop (Y200-V210), but they are expected to reach out to the bulk solvent, and expose one of the predicted N-glycosylation sites of the transporter (N204). Notably, despite the lack of sequence identity among human SLC1 transporters, all of them contain predicted N-glycosylation sites in the TM4b-c loop, suggesting a common role of this loop in the posttranslational processing of these proteins.
  • TM1 a amphipathic helix
  • TM1 a forms the tips of the blades in the propeller (Fig. 3c).
  • Fig. 3c amphipathic helix
  • TM1 a somehow interacts with the inner leaflet of the membrane.
  • a second hydrophobic crevice is observed between the extracellular part of TM4 and HP2, where there is also strong non-protein electron density.
  • a similar crevice was also noted in a substrate-bound structure of GltPh 22 .
  • TM1 a-HP1 a and TM4-HP2 hydrophobic crevices are at the interface between the TranD and ScaD, where large conformational changes are expected to occur during substrate translocation, they might constitute sites for lipid regulation of transport function.
  • EAAT1 cryst showed strong electron density for UCPH101 in a hydrophobic pocket facing the inner leaflet of the membrane on the interface between the TranD and ScaD (Fig. 3d,e and Fig. 5a, b). This pocket is formed by residues in TM3, TM7 and TM4c, and extends the TranD-ScaD interface by ⁇ 500 A 2 .
  • the chromene skeleton of UCPH101 is buried deeply in the domain interface, and coordinated by a direct ring- stacking interaction with F369 (TM7a), as well as hydrophobic interactions with G120 (TM3), V373 (TM7a) and M231 (TM4c) (Fig. 5b).
  • TM7a direct ring- stacking interaction with F369
  • TM3 hydrophobic interactions with G120
  • TM7a V373
  • TM4c M231
  • EAAT1 C ryst-n in the UCPHioi-bound state is nearly identical to that of the EAATI cryst with the exception of the mutated 1231 and I235 side chains, and a ⁇ 2 A movement of UCPH101 methoxy-phenyl and naphthalene groups away from them (Fig. 5c).
  • the EAAT1 C ryst-ii UCPHioi-unbound structure has an overall similar conformation to the UCPHioi-bound state, but shows notable differences.
  • the UCPH101 binding pocket contains no excess electron density, and the side chain of F369 moves outward by as much as 1.9 A, partly occupying the volume for UCPH101 chromene group (Fig.
  • HDX-MS hydrogen-deuterium exchange mass spectrometry
  • the HDX behavior of the EAAT1 cr yst was compared in the presence and absence of UCPH101.
  • the deuterium uptake pattern of EAAT1 cr yst shows dynamic structural elements in both the TranD and ScaD, and reveals the unstructured and solvent-exposed nature of several regions that were not resolved in the crystal structures, including the TM3-TM4a (peptide 153-173) and TM4b-c (peptide 200-208) loops, as well as the N- (peptide 1 -28) and C- termini (peptides 490-522).
  • UCPH101 significantly decreased deuterium uptake in several areas of the TranD including its binding pocket (residues 1 12-123 and 370-374), and the surrounding area (residues 354-369), while it left the uptake in the ScaD unchanged. It also decreased deuterium uptake in distant residues (336-349 and 420-430) at the tips of HP1 and HP2 involved in substrate coordination and occlusion, suggesting that UCPH101 induces conformational changes in the transporter upon binding.
  • the comparison between the structure and the model shows that the UCPHioi-modified areas detected by HDX-MS transit as rigid bodies from being solvent-exposed, in the inward-facing state, to buried at the TranD-ScaD interface, in the outward-facing state.
  • Such conformational change is expected to decrease the dynamics of alpha helices and/or the solvent accessibility of the loops in the UCPHioi-modified areas and thus, is consistent with the observed decrease in deuterium uptake.
  • the HDX-MS and structural analysis support the stabilization of the outward-facing state, at the expense of the inward-facing state(s), induced by UCPH101.
  • the UCPHI OI -TBOATFB bound structure is similar to that of the UCPHioi-bound state, with the exception of HP2 that adopts an "open" conformation and packs against the TM4b-c loop, disrupting the coordination of the Na2 (Fig. 6a, b).
  • These conformational changes resemble those previously observed in the structure of the GltPh-TBOA complex, and are in excellent agreement with the proposed competitive inhibitory mechanism of TBOA-like compounds 25 .
  • TBOATFB (Fig. 6c), but due to lack of resolution, it was not possible to unambiguously orient the compound.
  • the TBOA moiety of TBOATFB was initially positioned using the TBOA-bound GltPh structure as a guide. In this position, it remained stable during several successive cycles of refinement that yielded a reasonable fit into the electron density.
  • the additional benzoylamino and trifluoromethyl groups of TBOATFB localized in a hydrophobic cavity mainly formed by residues in HP1 b and TM7a, and possibly by residues in TM2 and TM4c.
  • the interactions of these groups with the transporter could explain the ⁇ 1500-fold increase in inhibitory potency of TBOATFB, compared to TBOA, that has been observed in EAAT1 transport assays 48 .
  • UCPH101 bound in this structure with an identical coordination than in the substrate-bound state. Therefore, despite the lower resolution of the TBOATFB bound structure, it shows that UCPH101 binding at its allosteric site does not preclude the movements of HP2 involved in substrate and sodium binding from the extracellular solution.
  • EAAT1 cryst reveal new architectural features of human SLC1 transporters, and the first non-competitive inhibitory mechanism of this family of proteins in molecular detail.
  • UCPH101 is an EAAT1 -selective inhibitor with a bipartite coordination by residues in both the TranD and ScaD, and the downward rigid-body movements of the TranD during transport disrupt such coordination. This implies that upon binding, UCPH101 "glues" the TranD to the ScaD in the outward-facing states, and precludes the translocation reaction of the transport cycle, but not the substrate binding/unbinding reactions from the extracellular solution.
  • the inhibitory mechanism of UCPH 101 contrasts with that of substrate-analog competitive inhibitors like TBOA.
  • the binding pocket of TBOA-like compounds overlap to some extent with that of the substrate 25 , and some of these molecules can bind the transporter from both the extra- and intracellular aqueous solutions 49 50 . Therefore, they inhibit transport by precluding substrate binding on either side of the membrane.
  • TBOA-like compounds are not selective among glutamate transporters due to the high amino acid conservation in the substrate-binding sites.
  • UCPH101 The mechanistic differences observed in UCPH101 over other known inhibitors make it an extremely valuable pharmacological tool, to isolate and study the conformational changes that EAAT1 undergoes upon substrate and ion binding.
  • the UCPH101 allosteric binding site observed in EAAT1 cr yst highlights a cavity that can facilitate the design of selective compounds for other human SLC1 transporters, and possibly the long-sought positive modulators of glutamate uptake.
  • the structure shows that the transporter is a homo-trimer, in which each protomer contains two structural and functional domains, the scaffold (ScaD) and transport (TranD) domains, previously observed in the structures of a glutamate transporter prokaryotic homologue.
  • the ScaD forms the inter-subunit interface through extensive contacts between transmembrane helices TM2 and TM4 on the extracellular half of the membrane, as well as between TM4 and TM5 on the cytoplasmic side.
  • alpha helical regions of the protein including TM3 and TM6-8, as well as two re-entrant helical loops (HP1 -2) fold into the TranD that encages the substrate and the coupled ions and translocates them across the membrane through rigid-body movements in an "elevator-like" fashion.
  • EAATI WT is inactive in transport assays, while ⁇ 1 ⁇ and ⁇ 1 ⁇ 2 are active.
  • the detergent solutions used for solubilization and purification may induce unfolding events that render the wild type transporter inactive.
  • the structural differences between the two mutants and the wild type transporter in detergent solutions have therefore been compared.
  • Hydrogen-deuterium exchange behavior of the three proteins using a protease-induced fragmentation approach linked to mass spectrometry (HDX-MS) was used.
  • HDX-MS measures the rate of deuterium exchange of the hydrogen amide in the protein backbone, which depends strongly on the presence of secondary structure due to the engagement of the amide group in hydrogen bonding as well as access to the aqueous solvent. Therefore, HDX-MS provides valuable information on protein folding, stability and dynamics.
  • EAAT1 TI and ⁇ 1 ⁇ 2 showed similar overall HDX patterns when assayed at 20 °C in detergent solutions, with sequence coverage of ⁇ 70 % and unimodal isotopic envelopes across all peptides measured.
  • Analysis of the HDX kinetics revealed that most peptides covering the alpha-helical regions of the TranD and ScaD take up deuterium at rates between 10 ⁇ 2 to 10 ⁇ 5 s ⁇ 1 , which are several orders of magnitude slower than predicted for unstructured peptides with the same sequence (10 2 s "1 ).
  • peptides partly covering at least TM4-5, as well as TM8 and HP2 were completely protected and do not show any deuterium uptake.
  • ⁇ 1 - ⁇ and ⁇ 1 ⁇ 2 showed an overall similar sequence of events during thermal inactivation, despite the difference in amino acid sequence, in which the loss of quaternary structure is an essential early event in the denaturation pathway.
  • the overall kinetic stability of the ScaD is lower than that of the TranD, and mutations in the ScaD determined the stability of trimeric form of the transporters, as shown by the chimera EAAT1 -ScaD-n-TranDT2;
  • the lowest local apparent melting (T50-HDX) observed in both ⁇ 1 ⁇ and ⁇ 1 ⁇ 2 map to an extensive area of the trimeric interface formed by TM4a-c and TM2 from neighboring subunits, respectively.
  • HP2 were constrained by lipid or detergent molecules in the monomeric form of the transporters, but not in the trimeric one. In fact, decreased in deuterium uptake at the level of HP2 in both ⁇ 1 ⁇ and EAAT1 T2 at high temperatures, is consistent with this line of thought.
  • Thermal denaturation is in general an irreversible process in detergent-solubilized membrane proteins. Applying temperature protocols that generate partial denaturation of the proteins offers the possibility to capture kinetic intermediates of the unfolding pathway. Size Exclusion Chromatography has first been used to analyze the effect of twenty-minute temperature pre-pulses on purified ⁇ 1 ⁇ and ⁇ 1 ⁇ 2 in detergent solutions, at a constant protein concentration, in order to quantify the irreversible effect of temperature on protein solubility, aggregation, and oligomeric state compared to reference transporters that were not pre-heated.
  • ⁇ 1 - ⁇ and ⁇ 1 ⁇ 2 reference samples maintained at 4 °C eluted as symmetric and monodisperse peaks that correspond to the trimeric form of the transporters ( ⁇ 3 ml elution volume), and remained stable up to 35 °C and 25 °C respectively (Fig. 9a, 9b and Fig. 9c,9d respectively).
  • the trimers melt into a lower oligomeric state that elutes at ⁇ 3.5 ml, without any signs of aggregation or insolubility, judging by the lack of high molecular-weight peaks and the constant area under the chromatograms at all temperatures respectively.
  • EAAT1 T2 samples pre-heated at 55 °C in liposomes were reconstituted and compared to the reference samples maintained at 4 °C.
  • the large decrease in neurotransmitter uptake observed in the pre-heated samples shows that the transport function of the "low molecular-weight" species has been largely impaired, and suggests that the monomeric form of the transporter is not functional.
  • the overall HDX pattern of ⁇ 1 ⁇ and ⁇ 1 ⁇ 2 was similar when the transporters were pre-heated at nearly their TSO-SEC, 40 °C and 50 °C respectively. Both transporters showed a significant increase in deuterium uptake in peptides covering TM4a-b (e.g. peptide 174-184) and the extracellular part of TM2 (e.g. peptide 81 -87) indicating partial loss of secondary structure in these helices. Additional increase in deuterium uptake was observed in the cytoplasmic parts of TM2 and TM3 (e.g.
  • EAAT1 TI and ⁇ 1 ⁇ 2 were also probed at pre-pulse temperatures above the
  • the peptides covering the extracellular helices TM4a-b showed clear bimodal isotopic envelops in the m/z spectra, while all other peptides were unimodal.
  • the bimodal envelopes were most apparent at pre-pulse temperatures close to the TSO-SEC (Fig. 10a and 10b), in which the amplitudes of the high- and low-mass components were similar. In contrast, at temperatures above and below the Tso-sEc the envelopes were nearly unimodal and dominated by the high- and low-mass components respectively.
  • TSO-HDX-GAUSS Using Gaussian fitting to calculate fraction of the low-mass components as a function of temperature, and estimated the apparent melting temperature of the peptides at the inter-subunit interface (TSO-HDX-GAUSS), peptides 174-184 and 187-194 in ⁇ 1 ⁇ showed TSO-HDX-GAUSS values of 49,5 °C and 48,8 °C respectively, while those of EAAT1 T 2 were 36,8 °C and 42,4 °C respectively (Fig. 10c).
  • the deuterium uptake kinetics of all peptides is well described by three components: an initial burst, which corresponds to the uptake measured at 10 s; a slow component with uptake rates ⁇ 10 ⁇ 3 s ⁇ 1 , and an intermediate component with rates ⁇ 10 "1 -10 "2 s "1 (Fig. 1 1 a and 1 1 b).
  • the deuterium uptake of the peptides covering the helical regions of the transporter is dominated by the slow component under reference conditions (e.g.
  • Mutagenesis based on consensus amino acids may allow the engineering of membrane protein with enhanced stability.
  • the two approaches disclosed therein substituteting one or more amino acid residues in the amino acid sequence of the target protein by its(their) respective consensus amino acid residue(s) or substituting one or more amino acid residues in the amino acid sequence of the target protein by substitution, wherein said amino acid residues are selected among residues which are present in pairs of residues that show co-evolution in the target protein
  • the consensus approach is especially dedicated for protein families that lack high selective pressure on their stability determinants.
  • the loss of stability observed in a full-consensus sequence of highly thermostable soluble proteins supports the above.
  • non-specialized ancestral proteins are expected to be more stable than the extant specialized structural homologs; the gain of stability has been observed in several laboratories during resurrection of ancestral soluble proteins.
  • the consensus approach may be adapted to include the condition of co- variance.
  • conserved amino acid interactions in the SLC1 family are important to the stability of the fold and therefore, restricting the consensus mutagenesis to conserved interacting positions may improve the protein stability.
  • the determination of pairs of position that show high co-variance in a curated PFAM alignment of the SLC1 family, as a proxy for conserved amino acid interactions, and application of the same consensus amino acid exchanges than in the consensus mutagenesis to those positions only permit to reduce the overall number of mutations. Indeed, the second method decreases the number of consensus mutations from 77 to 29, and yield a transporter that shares ⁇ 95% sequence identity with wild type EAAT1 .
  • Example 5 THERMAL STABILIZATION OF HUMAN Niemann-Pick C1 -like1 PROTEIN
  • NPCL1 Niemann-Pick C1 -Like1
  • NPCL1 is a cholesterol binding protein and is responsible for cholesterol re-adsorption in the apical plasma membrane of intestinal and hepatic epithelia.
  • Wild type human NPC1 L1 (NPC1 L1WT) contains 13 transmembrane helices and three large cytoplasmic domains, and although it expresses well in HEK293 cells, it is barely soluble in detergent solutions.
  • N PC1 L1WT The amino acid sequence of N PC1 L1WT includes 1 ,359 residues (SEQ ID No: 12), and it makes impractical and costly the implementation of methods like amino acid scanning mutagenesis to improve its stability. To overcome this problem, the method of the invention has been applied and a mutant construct called NPC1 L1 TI has been designed.
  • a consensus residue was defined when it met all the below criteria: i) the residue had the highest frequency of occurrence among the aligned sequences, and this frequency is >20%; ii) the residue occupied a position in the alignment in which gaps constitute ⁇ 30% of the entries; iii) the residue's frequency of occurrence differed by more >10% from the corresponding frequency of the residue in the target/parent sequence (human sequence: NPC1 L1WT);
  • NPC1 L1 homologous transporter NPC1 L1 thermostable T1 NPC1 L1 TI
  • NPC1 L1 TI shares ⁇ 91 % amino acid sequence identity with NPC1 L1WT (Sequence 12).
  • NPC1 L1 TI expresses at similar levels than NPC1 L1WT in HEK293 cells, but shows a strong improvement in detergent solubility giving rise to a major dominant peak in the size exclusion chromatogram of clear lysates (Fig. 12).
  • the consensus-based approaches illustrated by ⁇ 1 ⁇ , ⁇ 1 ⁇ 2 and NPC1 L1 TI can be generalized to other membrane proteins to obtain stable and functional proteins for structural, as well as biophysical analysis.
  • the methods of the invention allow reduction of the number of constructs to be screened or tested by nearly two orders of magnitude compared to current method like alanine screening and direct evolution, since only a few synthetic genes containing the consensus mutations have to be tested.
  • the methods of the invention allow an easy and eventually automated construct design; based on simple phylogenic analysis of protein sequences and algorithms available online through web-based servers, the calculation and of amino acid consensus sequences is simple.
  • Fluorescence-detection size-exclusion chromatography (FSEC) 51 was used to screen solubilization conditions and EAAT1 variants fused to enhanced green fluorescent protein (eGFP).
  • EAAT1 N-terminal fusions solubilized in dodecanoyl sucrose (DDS, Anatrace) were found to have good solubility and mono-dispersity by FSEC in clear lysates.
  • DDS dodecanoyl sucrose
  • EAAT1 loses its transport activity and chromatographic monodispersity upon purification.
  • consensus mutagenesis 26 was used according to the method of the invention as described in Example 1 , and EAAT1 variants were screened with different consensus mutations in the predicted transmembrane helices by FSEC.
  • the apparent melting temperature (Tm) of the most stable EAAT1 construct was >20 °C over that of the wild-type EAAT1 , but the mutated transporter was still refractory to crystallization. It The extracellular region between TM3-4c was changed for the shorter TM3-4c sequence from ASCT2 (Fig.4). In addition, the two predicted N-glycosylation sites of the transporter (N155T and N204T mutations) were mutated. The obtained mutant showed improved crystallizability.
  • EAAT1 cr yst The transporter obtained this way, called EAAT1 cr yst, retained the functional and pharmacological properties of the EAATI WT upon purification and has the following sequence:
  • Cells were transiently transfected in FreestyleTM293 medium (Invitrogen) using poly-ethylenimine (PEI) (Tebu-bio) at a density of 2.5 x 10 6 cells ml "1 , diluted with an equivalent volume of Ex- Cell®293 6 hours post-transfection, and treated with 2.2 mM valproic acid (Sigma) 12 hours after dilution of the cultures. Cells were collected at ⁇ 48 h post-transfection.
  • PEI poly-ethylenimine
  • Membranes were washed once with the above-mentioned buffer and finally homogenized with a douncer in a buffer containing 50 mM HEPES/Tris-base, pH 7.4, 200 mM NaCI, 1 mM L-asp, 1 mM EDTA, 1 mM TCEP, and 10% Glycerol, snap-frozen in liquid N2 and stored at -80°C at 0.5 g of membranes ml "1 .
  • Membrane solubilization was done by thawing out and supplementing the membrane homogenate with 2% DDS, 0.4% CHS, and 25 ⁇ UCPHioi (Abeam).
  • the eluted eGFP-transporter fusion was concentrated to 1 -2 mg ml "1 using 100- kDa cutoff membranes (Millipore), and digested with His-tagged PreScission protease overnight at 4 °C.
  • the protease was removed by reverse Ni-NTA (Qiagen) affinity chromatography, and the flow through containing the transporter was concentrated to 500 ⁇ , ultra-centrifuged (86,900 g, 20 min), and applied to a Superose 6 10/300 gel filtration column (GE Healthcare) equilibrated with 50 mM HEPES/Tris-base, pH 7.4, 200 mM NaCI, 1 mM L-asp, 1 mM TCEP, 5% Glycerol, 0.25% decanoyl sucrose (DS, Sigma), 0.05% CHS and 100 ⁇ UCPHioi .
  • the protocol was identical, but the allosteric inhibitor was omitted from all buffers.
  • the protein sample was supplemented with 3 mM TBOATFB (Tocris) before the injection in the gel filtration column equilibrated with 50 mM HEPES/Tris-base, pH 7.4, 200 mM NaCI, 1 mM TCEP, 5% Glycerol, 0.25% decanoyl sucrose (DS, Sigma), 0.05% CHS, 300 ⁇ TBOATFB and 100 ⁇ UCPHioi . Protein samples after the solubilization step were kept on ice or at 4 °C at all times.
  • Purified protein was concentrated to 3.5-4.0 mg ml "1 and 1 mM UCP H 101 was added in experiments with the inhibitor-bound transporters.
  • Initial vapor diffusion crystallization screens were done by mixing 300 nl of protein and reservoir solution in sitting drops, dispensed by a Mosquito® robot (TTP labtech) in 96-well Greiner plates.
  • the purified transporters form three-dimensional crystals in several conditions containing low molecular weight polyethylene glycols.
  • the best-diffracting crystals were obtained after manual optimization using 1.6 ⁇ hanging drops at 4 °C, obtained by mixing equal volumes of protein supplemented with 0.2% n-Octyl-b-D- glucopyranoside (BOG, Anatrace) and 0.04% CHS, and reservoir solutions containing 100 mM Tris, pH 8.2, 50 mM CaCI 2 , 50 mM BaCI 2 , and 28-30% PEG 400. Crystals appeared after 24-48 h and reached their maximum size after a week. Crystals were flash-frozen in liquid nitrogen before X-ray diffraction data collection without any further cryo protection.
  • BOG n-Octyl-b-D- glucopyranoside
  • X-ray diffraction data were collected at beamlines PROXIMA-1 at the SOLEIL synchrotron (St Aubin, France) and at beamlines at the European Synchrotron Radiation Facility (Grenoble, France). In general, 2-3 data sets from single crystals were collected, and indexed, integrated, scaled and merged using XDS package 52 . Due to the anisotropic nature of the diffraction data, the DEBYE and STARANISO programs were applied to scale it using the STARANISO server (http://staraniso.globalphasing.org/). The software performs an anisotropic cut-off of merged intensity data with a Bayesian estimation of the structure amplitudes, and applies an anisotropic correction to the data.
  • Table 1 shows the refinement statistics for the full sets of reflections truncated at the best high-resolution along h, k or I axis, values given by AIMLESS 53 , before the anisotropic corrections computed by the STARANISO software.
  • the corrected anisotropic amplitudes were then used for molecular replacement in PHASER 54 , using the scaffold and transport domains of GltPh (PDB code 2NWL) as independent search models.
  • the initial electron density maps were clearly interpretable, and the final model was obtained through rounds of manual building in COOT 55 and refinement in Buster 56 , until reaching good crystallographic statistics and stereochemistry.
  • the model contains one EAAT1 cr yst monomer per asymmetric unit and most of the EAAT1 cr yst polypeptide (residues 37- 487), with the exception of some residues in the extracellular loops between TM3-4a, TM4b-4c, TM5-6 and TM7b-HP2a. Sequence assignment was aided by anomalous difference Fourier maps from diffraction data collected with low energy X-rays (1.77 A) to highlight the sulfur atoms of methionine and cysteine residues.
  • EAAT1 C ryst-n as well as the EAATI cryst UCPHIOI -TBOATFB bound structures were solved by the same approach above mentioned, but using the EAAT1 cr yst TranD and ScaD as independent search models for molecular replacement.
  • Unilamellar liposomes were made at 9: 1 molar ratio of 1 -palmitoyl-2-oleoyl-sn- glycero-3-phosphocholine (Avanti Polar Lipids) and CHS, in a buffer containing 50 mM HEPES/Tris-base, pH 7.4, 200 mM NaCI and 1 mM L-asp.
  • the transporters were purified as described above, but excluding the reverse chromatography step after protease cleavage, and using a Superose 6 10/300 column equilibrated with 50 mM HEPES/Tris-base, pH 7.4, 200 mM NaCI, 1 mM L-asp, 0.5 mM TCEP, 0.0632% DDS, 0.01264% CHS, and 5% glycerol.
  • liposomes were first mixed with DDS at a 1 :2 (w/w) lipid-to-detergent ratio for 1 h, and then the purified transporters were added at a 1 :40 (w/w) protein-to-lipid ratio. Detergent removal was done at 4° C using SM-2 biobeads (BioRad) at 100 mg ml "1 . The internal solution of the liposomes was exchanged using 10 freeze-thaw cycles in the appropriate buffer.
  • proteoliposomes were concentrated by ultracentrifugation (150,000 g for 30 min at 4°C) and resuspended at 20 mg of lipids ml "1 , for immediate use.
  • Substrate transport was assayed at 37°C.
  • the uptake reaction was initiated by diluting the proteo-liposomes 10-fold into a buffer containing 50 mM HEPES/Tris-base, pH 7.4, 200 mM NaCI, 50 ⁇ L-glutamate, and 5 ⁇ [ 14 C]-L-glutamate (PerkinElmer), and 2.5% glycerol. After 30 min, 200- ⁇ aliquots were diluted 5-fold into ice-cold quench buffer (50 mM HEPES/Tris-base, pH 7.4, 200 mM ChCI, and 2.5% glycerol), followed by immediate filtration and wash on nitrocellulose 0.22- ⁇ filters (Millipore).
  • Radioactivity was quantified by liquid scintillation using a Tri-Carb 31 10TR counter (PerkinElmer).
  • Tri-Carb 31 10TR counter PerkinElmer
  • proteo-liposomes were both pre-incubated for 20 min at room temperature, and assayed in the presence of UCPH101. Background radioactivity was estimated from protein-free liposomes, and subtracted from the uptake data. Data was fitted to a Hill equation of the form:
  • proteo-liposomes were assayed in the presence of 0, 5, 50 or 200 ⁇ L-glutamate supplemented with 1 , 5, 5, or 5 ⁇ [ 14 C]-L-glutamate, respectively.
  • the initial rate of transport was calculated by a linear fit to 120 s and 180 s uptake measurements with origin fixed at zero. Background radioactivity was estimated from protein-free liposomes, and subtracted from the uptake data.
  • cells were collected 36 h post-transfection, and washed three times and resuspended at a density of 50 x 10 6 cells ml "1 in 1 1 mM HEPES/Tris-base, pH 7.4, 140 mM ChCI, 4.7 mM KCI, 2.5 mM CaCI 2 , 1.2 mM MgCI 2 , and 10 mM D-glucose, for immediate use.
  • the uptake assay was performed similarly to the one described for the proteo-liposomes, but using a reaction buffer containing 1 1 mM HEPES/Tris-base, pH 7.4, 140 mM NaCI, 4.7 mM KCI, 2.5 mM CaCI 2 , 1 .2 mM MgC , 10 mM D-glucose, 50 ⁇ L-glutamate, and 5 ⁇ [ 14 C]-L-glutamate, and 0.8- m nitrocellulose filters. Background radioactivity was estimated from cells transfected with empty vector, and subtracted from the uptake data.
  • HDX-MS experiments were performed with transporters purified as described in the proteo-liposome section, and using a Superose 6 5/150 gel filtration column equilibrated with 50 mM HEPES/Tris-base, 200 mM NaCI, pH7.4, 1 mM L-asp, 0.5 mM TCEP, 0.0632% DDS, 0.01264% CHS, and 5% glycerol.
  • the purified EAAT1 cr yst was incubated in ice for 30 min with 2.2% DMSO at a monomer concentration of 5.2 ⁇ , in the presence and absence of 102 ⁇ UCPHioi , respectively. Prior to labeling, 10 ⁇ _ of the unbound and UCPHioi-bound EAAT1 cr yst solution was equilibrated for 10 min at room temperature.
  • Deuterium exchange was initiated by adding 40 ⁇ _ of D 2 0 buffer (50 mM HEPES, pH 7.3, 200 mM NaCI, 1 mM L-asp, 5% glycerol, 0.0632% DDS, 0.01264% CHS, 0.5 mM TCEP) supplemented or not with 101 .2 ⁇ UCPHioi . Assuming that the K D and IC50 values of UCPH101 are similar, ⁇ 96% of the transporters would remain bound during deuterium labeling.
  • the resulting peptides were trapped and desalted onto a C18 Trap column (VanGuard BEH 1 .7 ⁇ , 2.1 x 5 mm, Waters corp.) at a flow rate of 100 ⁇ _ ⁇ of 0.15% formic acid, and then separated in 10 min by a linear gradient of acetonitrile from 5 to 40% at 40 ML/min using an ACQUITY UPLC ® BEH C18 analytical column (1 .7 Mm, 1 x 100 mm, Waters corp.). After each run, the pepsin cartridge was manually cleaned with two consecutive washes of 1 % formic acid, 5% acetonitrile, 1.5 M guanidinium chloride, pH 2.5. Blank injections were performed between each run to confirm the absence of carry-over.
  • Mass spectra were acquired in resolution and positive mode on a Synapt G2-Si HDMS mass spectrometer (Waters corp.) equipped with a standard electrospray ionization source, as described previously 57 .
  • Peptides were identified from undeuterated protein samples acquired in MS E mode by database searching in ProteinLynX Global Server 3.0 (Waters corp.). Each fragmentation spectrum was manually inspected for assignment validation. Deuterium uptake values were calculated for each peptide using DynamX 3.0 (Waters corp.). Only one unique charge state was considered per peptide and no adjustment was made for back-exchange.
  • HDX-MS results are reported as relative deuterium uptake values expressed in mass unit or fractional exchange 57 . A statistical analysis was performed with MEMHDX 58 using a False Discovery Rate of 1 %. References
  • ASC amino-acid transporter 2 (ASCT2) as a novel prognostic marker in non-small cell lung cancer.
  • Raunser S. et al. Heterologously expressed GLT-1 associates in approximately 200-nm protein-lipid islands. Biophys J 91 , 3718-3726, doi: 10.1529/biophysj.106.086900 (2006).

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Analytical Chemistry (AREA)
  • Ecology (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The invention relates to a method for the identification of mutated proteins having modified thermal stability and in particular having improved thermal stability. The amino acid sequence of the mutated protein results from the substitution of amino acid residues by consensus amino acid residues determined following MSA and selection of the consensus residue based on sequence comparison of homologues.

Description

METHOD FOR THE IDENTIFICATION OF MUTATED PROTEINS HAVING
MODIFIED THERMAL STABILITY
[1] The invention relates to a method for the identification of mutated proteins having modified thermal stability and in particular having improved thermal stability. The amino acid sequence of the mutated protein results from the substitution of amino acid residues by consensus amino acid residues determined following MSA and specific selection of each consensus residue based on sequence comparison of homologues.
[2] The identification of mutant amino acid sequences for a target protein using the process of the invention enables the production of mutant proteins having modified thermal stability, in particular improved thermal stability.
[3] Thermal stability of proteins is a key issue for their production and purification in cellular systems when said production and purification involves the use of detergent solutions. This challenge exists especially for membrane proteins, in particular for Integral Membrane Proteins such as transport proteins that may be active as drug targets or more generally in drugs pharmacokinetics.
[4] Methods are available in the prior art that improve protein stability, in particular methods that have been applied to Integral Membrane Proteins (IMP). These methods include alanine-Scanning (AS), Directed Evolution (DE) or Rational
Design (RD).
[5] AS is currently the most established method, and relies on screening the thermal stability of single alanine-mutants at each position in the target protein. This feature makes AS a labour-intensive and costly method, and difficult to apply to large IMP (>500 amino acids residues) or multi-protein complexes. Moreover, AS requires the use of radioactive compounds with high affinity (nano molar) for the target protein, which are not available for most membrane proteins.
[6] The DE method relies on a key selection assay for protein expression and stability that requires a small fluorescent compound with high affinity for the protein target, which are not available for most IMP. Moreover, DE in its current stage can only be done using bacterial expression systems, which have been found problematic for most human membrane proteins. [7] The RD approach relies on previous knowledge of atomic-resolution structures of the protein target, which are unknown for most IMP, and requires a great deal of expertise to design the mutagenesis.
[8] The process of the invention is based on the stabilizing effect of consensus mutagenesis, and provides major improvements on protein stability with minimal changes of the amino acid sequence compared to the target protein, and without affecting critical residues for function as illustrated for Integral Membrane Protein function.
[9] As a proof of concept, the inventor applied the method to a family of human neurotransmitter transporters, which are membrane proteins whose biochemical instability has previously precluded protein purification and in vitro studies, besides their importance as drug targets.
[10] Membrane proteins account for ~30% of all open reading frames in the human genome 18 They play essential roles in human physiology and pathology that range from synaptic transmission to viral infection 19 20. Despite the paramount importance of human membrane proteins for life, their molecular mechanisms or function and pharmacology remain largely unknown. This is mainly due to the tremendous experimental challenges associated to their production, purification and analysis by high-resolution structural and functional approaches 21 -24 Clearly, new approaches and efforts are needed to understand the molecular mechanisms of this important class of proteins.
[11] The invention defines a process which is a solution to at least part of the drawbacks of the method disclosed in the art to improve thermal stability of proteins. Accordingly, mutant proteins were designed and obtained that showed improved thermal stability and that could be purified, and proved to be amenable for in vitro structural and functional studies, both of which have been performed successfully. In a particular embodiment, the invention accordingly provides a method to design mutated proteins that harbor improved thermal stability in detergents, wherein said proteins are membrane proteins, especially integral membrane proteins (IMP). The invention also relates to mutant proteins, in particular membrane proteins, especially IMP, designed and recovered accordingly.
[12] Importantly, the mutant protein, in particular mutant IMP, for example mutant transporter, retained the functional and pharmacological properties of the Wild Type human protein, in particular IMP, for example transporter, and shares at least 85% amino acid sequence identity with it, more preferably at least 90% amino acid sequence identity with it, and most preferably at least 95% amino acid sequence identity with it. Therefore, it constitutes an excellent structural and pharmacological model of protein, as illustrated for human protein.
[13] The examples provided therein show that it is the first time that a human IMP could be stabilized for in vitro studies by protein engineering with such a small change in its amino acid sequence and without the need for screening a wide variety of mutants. The method according to the invention requires the production of a reduced number of mutant proteins to be assessed for thermal stability property. Therefore, the method according to the invention is faster and requires far less mutant proteins production. The method herein defined accordingly provides a reliable tool for the production of functional proteins suitable for use in detergent solutions, without the need to produce a wide variety of constructs. Finally, ongoing experiments disclosed in the examples of the invention, show that the method can be successfully applied to other families of IMP, known for being very unstable proteins in detergent solutions, or even other proteins, eukaryotic or prokaryotic, in particular human or bacterial, with similar minimal changes of their amino acid sequence.
[14] Hence, the method that was developed according to the invention should contribute to better understanding of the molecular basis or function and pharmacology of proteins such as IMP, including those of human origin, and to develop efficient drugs that target them.
[15] Moreover, the method can also be applied to water-soluble proteins, in particular to those used in pharmaceutical and non-pharmaceutical industries.
[16] This new approach overcomes major problems associated to the existing methods:
I. It can be applied to any membrane protein or even membrane protein complexes,
II. it can be used with any protein expression system,
III. it does not require specific small compounds (radioactive or fluorescent) with high affinity for the target protein, IV. it relies only on knowledge of the amino acid sequence of the target and not of its three-dimensional structure,
V. it does not require special training skills, and
VI. it is both labour- and cost-effective.
[17] Proteins thermostable in detergent solutions may be useful in various applications, in particular structural analysis and biological analysis. Structural and biological analysis of proteins that requires proteins to be stable in a detergent solution encompass methods such as crystallography (i.e. X-ray crystallography), electron microscopy (i.e. cryo-electron microscopy), protein NMR, X-ray scattering (Small-angle X-ray scattering), circular dichroism, and the like. Methods for the biological analysis or pharmacological analysis encompass ligand-binding screening, affinity/kinetic screening, functional analysis of the proteins, drug targets screening, antagonist/agonist screening, and the like. As an example, production of proteins in detergents for crystallization requires a well- ordered crystal. Disposing stable proteins in detergents remains both challenging and necessary in order to have proteins suitable for study of their properties, in particular of their structural or pharmacological properties.
[18] The invention accordingly relates to a method of designing a protein with modified thermal stability in a detergent solution with respect to the thermal stability of a target protein comprising: a. providing a multiple sequence alignment (MSA) wherein at least a portion comprising at least 50 % of the full-length amino acid sequence of a target protein is aligned, in particular the full-length amino acid sequence of the target protein is aligned, with the amino acid sequence of homologous sequences of said protein present (i) in other species and/or (ii) in protein variants of the same species, using a determined alignment tool; wherein the homologous sequences are each different from the amino acid sequence of the target protein by at least one amino acid residue in their amino acid sequence, in particular wherein each homologous sequence shares at least 50% identity, more preferably at least 70% identity, with the amino acid sequence of the target protein or with the aligned portion thereof; b. defining for at least part of the positions of the amino acid residues in the target protein, in particular for each amino acid position of the aligned amino acid sequences, a consensus amino acid residue wherein the consensus amino acid residue has the following characteristics:
i. a frequency of occurrence among the aligned amino acid sequences which is higher than 20%,
ii. a position in the alignment of the amino acid sequences wherein gaps constitute less than 30% of the entries,
iii. a frequency of occurrence which is higher than 10% with respect to the frequency of occurrence of the residue at the same position in the target amino acid sequence when said frequency is evaluated among the aligned amino acid sequences;
c. providing a mutated amino acid sequence defining a consensus mutant protein of the target sequence by substituting one or more amino acid residues in the amino acid sequence of the target protein by its(their) respective consensus amino acid residue(s) defined for said each amino acid residue according to step b.
In a particular embodiment of the invention, the target protein is a membrane protein, in particular an integral membrane protein.
[19] The invention also relates to a process to produce a protein with modified thermal stability in a detergent solution with respect to the thermal stability of a target protein comprising: a. providing a multiple sequence alignment (MSA) wherein at least a portion comprising at least 50 % of the full-length amino acid sequence of a target protein is aligned, in particular the full-length amino acid sequence of the target protein is aligned, with the amino acid sequence of homologous sequences of said protein present (i) in other species and/or (ii) in protein variants of the same species, using a determined alignment tool; wherein the homologous sequences are each different from the amino acid sequence of the target protein by at least one amino acid residue in their amino acid sequence, in particular wherein each homologous sequence shares at least 50% identity, more preferably at least 70% identity, with the amino acid sequence of the target protein or with the aligned portion thereof;
b. defining for at least part of the positions of the amino acid residues in the target protein, in particular for each amino acid position of the aligned amino acid sequences, a consensus amino acid residue wherein the consensus amino acid residue has the following characteristics:
i. a frequency of occurrence among the aligned amino acid sequences which is higher than 20%,
ii. a position in the alignment of the amino acid sequences wherein gaps constitute less than 30% of the entries, iii. a frequency of occurrence which is higher than 10% with respect to the frequency of occurrence of the residue at the same position in the target amino acid sequence when said frequency is evaluated among the aligned amino acid sequences;
c. providing a mutated amino acid sequence defining a consensus mutant protein of the target sequence by substituting one or more amino acid residues in the amino acid sequence of the target protein by its(their) respective consensus amino acid residue(s) defined for said each amino acid residue according to step b.;
d. obtaining a polynucleotide encoding the mutant protein of step c. for expression in a cellular expression system;
e. producing the mutant protein from the polynucleotide encoding the mutated amino acid sequence provided in step d. in a cellular expression system;
f. evaluating thermal stability in a detergent solution of the mutant protein recovered from the production cells in step e.
In a particular embodiment of the invention, the target protein is a membrane protein, in particular an integral membrane protein.
A portion of an amino acid sequence corresponds to at least 50% of the full-length amino acid sequence of the target protein, more preferably at least 75% of the full length amino acid sequence, more preferably at least 90% of the full-length amino acid sequence. A portion of amino acid sequence may also correspond to a plurality of non-contiguous portions of the full-length amino acid sequence, said plurality representing at least 50% of the full-length amino acid sequence. [20] Thermal stability in detergent solution is a parameter that is essential for the production of thermostable variants of proteins as it enables their purification in detergent solutions while preserving the biological function(s) of the wild type (or target) protein. It also enables production of variants of proteins (by oriented mutations) that are stable for biophysical analysis or structural analysis, including for the determination of their three-dimensional structure (by crystallization or electron microscopy for example) and for the determination of their biological function (ligand-binding assay for example). Thermal stability is accordingly the property of the target protein that is primarily altered when performing the process of the invention, in particular that is increased when providing or selecting a mutated sequence of the target protein. Thermal stability may be measured as disclosed hereafter in a denaturation assay assessing resistance to denaturation of the produced mutant protein. The invention thus provides a process for the identification and for the preparation of thermostable mutant proteins and in particular provides a thermostable variant of a target protein.
[21] In a particular embodiment other properties may additionally be modified in relation or independently of thermal stability of the target protein in accordance with the process of the invention. Such properties include the capability of the protein to efficiently keep their three-dimensional structure in detergent solutions and therefore keep their function. As an example, production crystal that are well- ordered may involve determination of the capability of said crystal to diffract X- rays to high resolution and comparison to a control. It has been shown indeed that there is a measurable correlation between the stability of a protein, in particular a membrane protein in a detergent and the capability of proper crystallization.
[22] The target protein for use in the invention is any protein present in any species.
In particular it is a eukaryotic protein and in particular belongs to animal, such as to vertebrates, especially to human, or it is a prokaryotic protein and belong to virus, bacteria, parasites species, in particular to a pathogenic species. According to a particular embodiment of the invention, the target protein is membrane protein of such organism, especially an IMP and in particular is a viral envelope protein, or a virus membrane receptor on a cell, especially a human cell. According to a particular embodiment, the target protein is a wild-type protein. According to another embodiment the target protein is a mutant, in particular a naturally occurring mutant of a wild-type protein, in particular a mutant selected or designed to alter properties with respect to the wild-type protein, possibly in respect of thermal stability or crystallization capability.
Accordingly, the target protein may have any length determined in number of amino acids residues. Advantageously, the protein has at least 300 amino acid residues, in particular at least 400 or 500 amino acid residues, but the method can also be performed with a target protein having less than 300 amino acid residues. In a particular embodiment of the invention, the protein has at least 1300 amino acid residues. Advantageously, the protein comprises between 300 amino acid residues and 1500 amino acid residues, and more advantageously, the protein comprises between 350 and 1400 amino acid residues. It should be noted that the methods of the prior art are increasingly cumbersome with the length of the amino acid sequence of the target protein. The method disclosed therein allows the efficient production of thermal stable proteins, irrespective of their number of amino acid residues.
For the purpose of the invention, the target protein is one the amino acid sequence of which is known and especially disclosed in databases such as GenBank. Similarly, the amino acid sequences of the homologous sequences of the target protein are known and in particular disclosed in databases such as GenBank.
In a particular embodiment, the amino acid sequences used for the performance of the alignment do not include a defined consensus sequence i.e., an artificial archetypal sequence defined using sequence data obtained from various known natural products but are rather representatives of isolated proteins. According to this embodiment, the target protein and the homologous proteins are wild-type sequences.
In another particular embodiment, especially the multiple sequence alignment (MSA) follows a first round or mutations in the amino acid sequence of the target protein, such as function-improving mutation(s) and/or structure-improving mutation(s). In such a case, the amino acid sequence of the target protein may be an artificial sequence, i.e., a sequence that has been mutated with respect to a naturally occurring protein in order to modify properties of the target protein. In another embodiment of the process of the invention, step c. of providing a mutated amino acid sequence defining a mutant protein of the target is followed by a further step of mutation in the sequence in order to alter other properties of the target protein.
[28] The alignment performed according to the process of the invention is a (MSA) which as such is well known to the person skilled in the art. It may be performed through available tools including a suitable algorithm (such as Clustal W) and software, especially using an alignment software. Many software suitable to perform alignments are available such as JALVIEW software or others listed on https://en.wikipedia.org/wiki/List_of_sequence_alignment_software. In a particular embodiment of the invention, the determined alignment tool used for the MSA is JALVIEW software. https://en.wikipedia.org/wiki/List_of_sequence_alignment_software
Multiple sequence alignment
Figure imgf000010_0001
Figure imgf000011_0001
Figure imgf000012_0001
Figure imgf000013_0001
Figure imgf000014_0001
Figure imgf000015_0001
*Sequence type: protein or nucleotide. **Alignment type: local or global
[29] Homologous sequences of the target protein may be found in species different from the species providing the target sequence. They may additionally encompass or alternatively consist in variants of the target sequence available in the same species. In particular, when the target protein is a human protein, the homologous sequences may originate from other vertebrate species, in particular from other mammal species. They may also encompass variants of the target sequences disclosed in human or in other vertebrate species. A homologous sequence may also be defined as a variant of the target protein having at least 50% identity with the target protein, in particular at least 70% identity, whether wild-type or mutated as disclosed above.
It is preferable but not necessary that each selected homologous sequence for the MSA has a difference with the target sequence such that both sequences share less than 95% sequence identity when aligned with the MSA tool.
Although there is no absolute requirement regarding the number of homologous sequences used in order to perform the MSA, having at least 7 and preferably more than 10 sequences in the alignment or more than 13 or more than 20 is advantageous for the performance of the process of the invention.
In step b. of the process the consensus amino acid residue may be defined for each amino acid position in the amino acid sequence of the target protein (also designated target sequence).
Alternatively the consensus amino acid residue may be defined for at least part of the amino acid residues in the target sequence and in particular for at least 25% or at least 50% or at least 75% or at least 90% of the amino acid positions in the target sequence.
In a particular embodiment of the process, the amino acid residues targeted for substitution by the consensus amino acid residues are contained in secondary structural elements of the target protein, such as in predicted or known (characterized) a-helices or in β-turns. Alternatively or in addition, the amino acid residues targeted for substitution by the consensus amino acid residues are contained in loops, in particular in unstructured loops connecting the other structured regions. In a particular embodiment such secondary structural elements and/or loops are involved in the alignment step performed on portion(s) of the amino acid sequence of the target protein.
When the consensus amino acid residue has been defined for the determined position of the target amino acid sequence, in accordance with steps i.to iii. of the process, a mutated amino acid sequence may be defined that results from the substitution of all amino acid residue(s) of the target sequence for which a consensus residue has been identified. The thus obtained amino acid sequence defines a mutant protein of the target protein. In a particular embodiment of the process the mutated protein has a sequence identity with the target protein which is at least 80%, more preferably at least 85%, more preferably at least 90%, most preferably at least 95%, in order to minimize the effects of the mutations on the properties of the target protein. Identity means to the extent to which two amino acid sequences have the same residue at the same position in an alignment. Identity could be defined as the number of exact matches in the alignment of two sequences (the target protein and a mutated protein), divided by the length of the target protein or the length of the aligned portion of the target protein. An exact match occurs when the target protein and the mutated protein have identical amino acid residues in the same position.
[36] From the identified mutant amino acid sequence, a polynucleotide can be designed, especially by mutation of the polynucleotide encoding the target protein, for the expression of the mutant protein in a cellular expression system.
The polynucleotide encoding the mutant protein having the mutated amino acid sequence may further be mutated for codon optimization in accordance with methods well known from the person skilled in the art for its expression in the selected cellular expression system. This polynucleotide may further be modified to add a sequence that encodes a purification handle at the terminus of the amino acid sequence of the mutant protein.
[37] The invention thus relates to the mutated protein which is produced and whose amino acid sequence consists in the consensus mutated sequence of the target protein.
[38] The invention also relates to the polynucleotide encoding the consensus mutated protein and to cells transformed with such polynucleotides in conditions enabling the expression of the polynucleotide as a mutated protein with respect to the target protein.
[39] According to another aspect of the invention, the process may additionally comprise, after step c. a further step (c(i)) of determination according to an additional parameter of the amino acid residues in the target sequence that may be mutated. In accordance with this embodiment step c(i) is a step of calculating pairs of residues that co-evolve in the target protein and substituting these pairs of residues in the target sequence.
[40] Step c(i) may be carried out using co-evolution software suitable for the determination of amino acid pairs that co-evolve such as
[41] EVcouplings software (website: http://evfold.org/evfold- web/evfold.do; reference: https://www.ncbi.nlm.nih.gov/pubmed/22579045) or others such as [42] GREMLIN (website: http://gremlin.bakerlab.org/index.php) (reference: https://www.ncbi. nlm.nih.gov/pubmed/?term=Assessing+the+utilitv+of+coevoluti on-based+residue-residue+contact+predictions+in+a+seguence- +and+structure-rich+era):
DCA (website: http://dca.rice.edu/portal/dca/)
(reference: https://www.ncbi. nlm.nih. gov/pubmed/?term=Direct- coupling+analvsis+of+residue+coevolution+captures+native+contacts+across+ manv+protein+families):
PSICOV
website: http://bioinfadm in. cs. ucl. ac. uk/downloads/PS ICON//) (reference: https:/ /www.ncbi.nlm.nih.gov/pubmed/?term=PSICOV%3A+precise+structural+contac t+prediction+using+sparse+inverse+covariance+estimation+on+large+multiple+ seguence+alignments)
[43] The additional but optional c(i) step has particular interest in situations where at the end of step c the mutant protein obtained has 80% or less sequence identity with the target sequence. The co-evolution determination brings the advantage of pointing to amino acid residues that co-evolved in the 3-dimensional structure of the protein rather than in its amino acid sequence. In a particular embodiment of the invention, this step c(i) may be performed when the number of homologous sequences available for the MSA is at least 5 times the number of amino acid residues of the target sequence. Because of that the homologous sequences may have to be found in species which are remote from the species of the target sequence, such as in prokaryotic homologues when the target protein is found in a eukaryotic species such as in human, and/or in divergent branches of the family protein of the target protein. When step c(i) is performed the identity of the mutations to be introduced in the target sequence is still based on step c. but the additional step ci enables to lessen the number of residues to be mutated. The obtained mutant sequence may thus be in the range of sequence identity of at least 90%, preferably at least 95% with the target sequence.
[44] Accordingly a variant (i.e. consensus mutant) protein obtained after step c and optionally c(i) may be prepared or produced in a cellular expression system and recovered for testing, in particular for thermal stability testing in detergent solution. [45] In a particular embodiment of the process of the invention, the target protein is a membrane protein, in particular a eukaryotic, such as a mammalian, especially a human, membrane protein. In a particular embodiment, the membrane protein is an integral membrane protein.
[46] In a particular embodiment of the process, the membrane protein is selected among channels, enzymes, and primary or secondary active transporters. Transporters may in particular be of the SLC1 transporters family which are ion- coupled amino acid transporters, and may comprise divergent branche(s) of this family, like sodium-dependent neutral amino acid transporters (ASCT1 -2).
[47] In another particular embodiment of the process of the invention, the target protein is a soluble protein, especially a globular soluble protein.
[48] The study of thermal stability of the mutant protein aims at determining whether mutations have improved, i.e., increased thermal stability in detergent solution. According to the invention, a produced, especially crystallized mutant protein having improved thermal stability shows resistance to denaturation when the melting temperature is raised for at least 2°C, advantageously for at least 3 to 10°C and in a particular embodiment up to 20°C with respect to the melting temperature of the target protein.
[49] The detergent solution is one known from the person skilled in the art in biochemistry, in particular one suitable for use in production, purification or analysis, especially by high-resolution structural and functional approaches. Detergent solutions may in particular be those used for preparation of protein for structure determination or biological function analysis; for example detergent used for X-ray structure or crystallization. For illustration such detergent solutions encompass small micelle detergents, dodecyl-p-D-maltopyranoside (12M), n- dodecyl-N,N-dimethylamine-N-oxide (LDAO), n-nonyl-p-D-maltopyranoside (9M), nonyl-p-D-glucopyranoside (NG), n-decyl-p-D-maltopyranoside. Detergent solutions may in particular be those used for preparation of protein for biophysical analysis and functional analysis using fluorescence spectroscopy (dodecyl-p-D- maltopyranoside, dodecyl-p-D-glucopyranoside, 7-Cyclohexyl-1 -Heptyl-p-D-
Maltoside), as well methods for determination of high-resolution three- dimensional structures in particular X-ray crystallography and single-particle electron microscopy (dodecyl- decyl-, nonyl-, or octyl-p-D-maltopyranoside; dodecyl-, decyl-, nonyl-, or octyl-p-D-glucopyranoside; Neopentyl glycol based detergents like 2,2-dioctylpropane-1 ,3-bis-p-D-glucopyranoside).
[50] Thermal stability of proteins may in general be characterized by the difference between the folded and unfolded states of the protein measured in kcalmol"1. Measurement of stability may encompass thermal denaturation of the protein in a detergent solution and measurement by the increase of the apparent melting temperature of the mutant protein which is then compared to the melting temperature of the target protein by chromatographic observation, in particular by size-exclusion chromatography (SEC-TS).
[51] According to SEC-TS, protein stability may be determined with or without ligand.
A sample of the purified protein is heated and then cooled and centrifuged to remove aggregated proteins and run on analytical HPLC. When the melting temperature is reached and proteins precipitates or aggregates, pic height decreases and void pic height increases.
[52] In a particular embodiment of the process when expressed in a cellular system, the mutant protein is fused to a detectable marker, such as a fluorescent marker, for the evaluation of the melting temperature of the mutant protein.
[53] The expression of the mutant protein may be performed in any available cellular system. When the protein is a eukaryotic protein, it is advantageously produced in a eukaryotic cell.
[54] The process of the invention enables selecting and producing proteins having improved thermal stability with respect to the target protein.
[55] Accordingly, the invention also relates to a process for modifying, in particular for improving thermal stability of a protein comprising carrying out consensus mutagenesis in the amino acid sequence of a target protein using the process defined herein in accordance to its embodiments and recovering the mutant protein having a modified, in particular improved thermal stability.
[56] According to another aspect, the invention relates to a process to identify an amino acid sequence which is mutated with respect to the amino acid sequence of a target protein said process comprising the steps of: a. providing a multiple sequence alignment (MSA) wherein at least a portion comprising at least 50% of the full-length amino acid sequence of a target protein is aligned, in particular wherein the full- length amino acid sequence of a target protein is aligned, with the amino acid sequence of homologous sequences of said protein present (i) in other species and/or (ii) in protein variants of the same species, using a determined alignment tool; wherein the homologous sequences are each different from the amino acid sequence of the target protein by at least one amino acid residue in their amino acid sequence, in particular wherein each homologous sequence shares at least 50% identity, more preferably at least 70% identity, with the amino acid sequence of the target protein or with the aligned portion thereof,
b. defining for at least part of the positions of the amino acid residues in the target protein, in particular for each amino acid position of the aligned amino acid sequences, a consensus amino acid residue wherein the consensus amino acid residue has the following characteristics:
i. a frequency of occurrence among the aligned amino acid sequences which is higher than 20%,
ii. a position in the alignment of the amino acid sequences wherein gaps constitute less than 30% of the entries, iii. a frequency of occurrence which is higher than 10% with respect to the frequency of occurrence of the residue at the same position in the target amino acid sequence when said frequency is evaluated among the aligned amino acid sequences;
c. providing a mutated amino acid sequence defining a mutant protein of the target sequence by substituting one or more amino acid residues in the amino acid sequence of the target protein by its(their) respective consensus amino acid residue(s) defined for said each amino acid residue according to step b.
[57] The invention also relates to a process for the preparation of a protein with modified crystallization capability comprising preparing a protein having modified thermal stability in a detergent solution according to any of the embodiments of the process of the invention. An optional step of further mutation may be performed after step c. and in particular before step d. of the process to prepare the mutant protein to further improve crystallization capability, such optional step of further mutation being in particular a step of mutation in the extracellular region of the target protein.
[58] In a particular embodiment of the invention, the process is performed, wherein the target protein is the Excitatory Amino Acid Transporter 1 (EAAT1 ) and the obtained mutant protein is EAAT1 of sequence SEQ ID No.1 SEQ ID No.2, SEQ ID No.3 or SEQ ID No.4. When the target protein is human EAAT1 , the homologous sequences aligned within the MSA may comprise at least one sequence selected from the group comprising SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10 and SEQ ID No: 1 1 . In a particular embodiment of the invention, when the target protein is human EAAT1 , the homologous sequences alignment within the MSA comprise at least the following sequences: SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10 and SEQ ID No: 1 1 .
[59] The invention thus also concerns a mutant protein of Excitatory Amino Acid Transporter 1 (EAAT1 ) the amino acid sequence of which is SEQ ID No.1 SEQ ID No.2, SEQ ID No.3 or SEQ ID No.4.
[60] In a particular embodiment of the invention, the process is performed, wherein the target protein is NPC1 L1 (Niemann-Pick C1 -Like 1 ), in particular human NPC1 L1 , in particular NPC1 L1 of Accession number Q9UHC9.2 (SEQ ID No: 12). Various isoforms of human NPC1 L1 , or ortholog proteins may be used in step a. of the method as homologous sequences, for example Niemann-Pick C1 - like protein 1 from Canis lupus familiaris (Accession number NP_001091019.1 - SEQ ID No: 14), from Bos Taurus (Accession number XP_588051 .4 - SEQ ID No: 15), from Loxodonta Africana (Accession number XP_003418622.1 - SEQ ID No: 16), from Mus musculus (Accession number Q6T3U4.1 - SEQ ID No: 17), from Monodelphis domestica (Accession number XP_001379744.2 - SEQ ID No: 18), from Dasypus novemcinctus (Accession number XP_004467651 .1 - SEQ ID No: 19), from Xenopus tropicalis (Accession number XP_002932620.2 - SEQ ID No: 20), from Oryzias latipes (Accession number XP_004072748.1 - SEQ 21 ), from Danio rerio (Accession Number XP_009302449.1 - SEQ ID No: 22), from Ornithorhynchus anatinus (Accession number XP_007662878.1 - SEQ ID No:
23) , from Python bivittatus (Accession number XP_007440049.1 - SEQ ID No:
24) , and from Aquila chrysaetos canadensis (Accession number XP_01 1573737.1 - SEQ ID No: 25). At least one of the above cited sequence may be aligned within the MSA when the target protein is human NPC1 L1 . In a particular embodiment of the invention, SEQ ID No: 12 to SEQ ID No: 25 may be aligned within the MSA when the target protein is human NPC1 L1 .
[61] In a particular embodiment of the invention, the process is performed, wherein the target protein is the Niemann-Pick C1 -like1 (NPC1 L1 ) and the obtained mutant protein is NPCL1 of sequence SEQ ID No.13.
[62] The invention thus also concerns a mutant protein of Niemann-Pick C1 -like1
(NPC1 L1 ) the amino acid sequence of which is SEQ ID No.13.
[63] According to another aspect, the invention relates to a liposome or a collection of liposomes containing a mutant protein obtained from carrying out the process according to the invention.
[64] The invention also relates to a cell expressing a mutant protein obtained from the process of the invention.
[65] According to a particular embodiment, the invention relates to a cell or a liposome comprising a mutant protein of Excitatory Amino Acid Transporter 1 (EAAT1 ) the amino acid sequence of which is SEQ ID No.1 SEQ ID No.2, SEQ ID No.3 or
SEQ ID No.4.
[66] According to a particular embodiment, the invention relates to a cell or a liposome comprising a mutant protein of Niemann-Pick C1 -Iike1 (NPC1 L1 ) the amino acid sequence of which is SEQ ID No.13.
[67] Each and every embodiment defined in respect to the process to produce a protein with modified thermal stability in a detergent solution according to the invention applies to the process to modify the thermal stability of a target protein and to the process to identify an amino acid sequence which is mutated with respect to the sequence of a target protein.
[68] Other features and advantages of the invention will be apparent from the Examples below and from the figures.
[69] The process of the invention may be followed by additional steps of mutation in the mutant protein such as mutations that enable the improvement of purification or crystallization of the protein.
[70] Table 1 . Data collection and refinement statistics. EAAT1 cryst EAAT1 cryst-l EAAT1 cryst-l EAAT1 cryst
UCPH10I UCPH10I UCPH101 and
TBOATFB
Bound bound
bound
Data collection*
Space group P63 P63 P63 P63
Cell dimensions
a=b, c (A) 123.27, 89.87 123.1 1 , 89.62 123.32, 89.57 124.33, 90.8
Alpha=beta(°) 90.0, 120.0 90.0, 120.0 90.0, 120.0 90.0, 120.0
Wavelength 0.979 0.976 1.009 0.977
Resolution (A) 45.89-3.25 45.82-3.1 45.87-3.32 46.31 -3.71
(3.34-3.25) (3.18-3.1) (3.41 -3.32) (3.81 -3.71)
Anisotropy
direction8
Resolution where
Overall (A) 3.37 3.1 3.32 3.71 along h, k axis (A) 3.75 3.68 3.85 4.35 along I axis (A) 3.25 3.1 3.32 3.71
Measured 333978 290672 326273 141904 reflections (24261) (21906) (21829) (1 1 176)
Unique reflections 12338 (902) 141 15 (1 032) 11556 (834) 8570 (628)
Completeness (%) 100 (100) 99.9 (99.9) 100 (100) 99.9 (100.0)
Mn (I) half-set 0.99 (0.22) 1 (0.43) 0.99 (0.31) 0.99 (0.373) correlation
l/ (l) 1 1.7 (0.7) 15.8 (0.8) 13 (0.7) 12.1 (0.9)
Emerge 0.20 (6.90) 0.1 (6.71) 0.15 (8.16) 0.14 (3.71)
Redundancy 27.1 (26.9) 20.6 (21.2) 28.2 (26.2) 16.3 (14.3)
Structure
determination
Refinement
Resolution cut-off 45.89-3.25 45.80-3.10 20.00-3.32 25.00-3.71
(A)
No. of Work / Test 9891/475 10725/528 9251/445 6860/684 reflections ftcryst (%) / ftfree (%) 21.9/24.1 21.7/25.9 20.9/25.3 22.7/25.4
No. of protein 3002 2960 2995 3008
atoms
No. of 42 42 10 62
heteroatoms
B factors (A)2
Protein 129.5 1 11.8 137.0 135.5
Heteroatoms 107.3 99.5 125.6 132.7
R.m.s. deviations
from ideal
Bond lengths 0.009 0.01 0.009 0.009
(A)
Bond angles (°) 1.06 1.12 1.05 1.03
One crystal was used to collect diffraction datasets for each structure, except in the EAATl
UCPHioi and TFB-TBOA bound structure, where datasets from three crystals were merged.
5% of reflections were used for calculation of /?free.
*Values in parentheses are for the highest-resolution shell.
$The anisotropy directions where computed with AIMLESS.
[71 ] Legend of the Figures
Some of the figures, to which the present application refers, are in color. The application as filed contains the color print-out of the figures, which can therefore be accessed by inspection of the file of the application at the patent office.
Figure 1 . Size exclusion chromatograms of detergent-solubilized GFP-fusions of EAATI WT and its thermostable variants at different temperatures from clear lysates of HEK293 cells (upper plots). The apparent melting temperature (Tm) was estimated from the GFP-fluorescence value of the peak corresponding to the trimeric protein (lower plots).
Figure 2. Uptake of radioactive L-glutamate by purified ΕΑΑΤ1 τι (left), and ΕΑΑΤ1 τ2 (right) reconstituted in liposomes. Purified EAATI WT did not show significant levels of uptake above the control liposomes without reconstituted transporters (see Fig. 3a). Figure 3. function and architecture of EAAT1 cryst. a-b, Uptake of radioactive L- glutamate by purified EAAT1 , EAATl cryst (cryst), and EAAT1 Cryst-n (cryst-ll) reconstituted in liposomes. Transport was abolished when choline (Ch+) was used as the main cation in the extra- or intra-liposomal solutions (circles) (a). UCPH101 inhibits glutamate transport EAATl cryst (lower plot), but not in EAAT1 Cryst-ii (upper plot) in a concentration dependent manner (b). Plots depict an average of three independent experiments performed with duplicate measurements, and error bars represent s.e.m. c-d, Structure of EAATI cryst trimer viewed from the extracellular solution (c) and from the membrane (d), with the ScaD and TranD as surfaces, and U PCH101 bound between them (spheres), e, EAATI cryst monomer viewed parallel to the membrane. The ScaD domain is represented as surface, and several helices and loops in the TranD have been removed for clarity of display, f, Domain organization diagram of EAATI cryst monomer.
Figure 4. Alignment of human SLC1 transporters. Amino acid sequences of EAAT1 -5, ASCT1 -2 and EAATI cryst are compared. The boundaries of the a-helices (cylinders) in the TranD and the ScaD seen in the EAATI cryst structure are shown. In order to confer crystallizability, the region between TM3 and TM4c (arrows) from ASCT2 was transferred to a thermally stabilized EAAT1 . To further improve crystal formation in the absence of U PCH101 , mutations M231 1 and F235I (circles) were introduced to generate EAAT1 cryst-n. These substitutions are found in EAAT2. Other residues involved in U PCH101 coordination are more conserved (triangles). Sequences were aligned with Jalview.
Figure 5. UCPH101 binding site, a, Lateral view of EAATI cryst monomer from the membrane showing UCPH101 bound between the TranD and ScaD. b-c, UCP H101 coordination and Fo-Fc densities contoured at 2.0o in EAATI cryst (b) and EAAT1 Cryst-ii (c), respectively. Side chains of residues in TM3, TM4c, and TM7 involved in coordination are shown. F369 side chain moves outward in the EAAT1 Cryst-ii unbound state (c).
Figure 6. UCPHI OI -TBOATFB bound EAATI cryst structure, a, The TranD and ScaD of the EAATI cryst monomer are represented in light grey and grey, respectively. The movement of HP2 partly exposes the substrate-binding pocket to the solvent and shows a molecule of TBOATFB bound to it. UCP H101 is also observed in this structure. b, The tip of HP2 moves as much as 9.5 A in the UCPHI OI -TBOATFB-, compared to the
UCPHioi-substrate bound structures, moving the carbonyl oxygen of A420 away from Na2 (black sphere), c, Omit map Fo-Fc(68) density for the TBOATFB molecule is contoured at 2.3o (black mesh), and some of the residues at Van deer Waals or H- bond distance from the compound are represented as sticks.
Figure 7. EAAT1 consensus mutants, a-b, Residues exchanged for consensus amino acids in ΕΑΑΤ1 τι (a) and ΕΑΑΤ1 τ2 (b) are mapped into the structure of the EAATI CRYST (PDB 5LLM) trimer viewed from the extracellular medium (left panel), as well as the scaffold (cyan) and the transport (orange) domains viewed from the membrane. These domains are depicted separately for clarity of display, including two views of the transport domain separated ~180° from each other, showing its interface with the scaffold domain (left) and the membrane (right), respectively. Spheres correspond to the alpha carbon atoms of residues that were exchanged by conservative (grey) and non-conservative (black) consensus mutations, c-d, Radioactive L-glutamate uptake in cells expressing transporters (c), including control cells transfected with a vector lacking EAAT1 genes, and in liposomes with purified reconstituted transporters (d). Yellow circles depict the liposomal bilayer separating sodium- (Na+), potassium- (K+), and choline-based (Ch+) solutions, e, Rate of L-glutamate uptake by purified ΕΑΑΤ1 τι (blue) and ΕΑΑΤ1τ2 (red) reconstituted in liposomes, as a function of L-glutamate concentration. Solid lines indicate Michael is-Menten fits to the data with Km values 30.7 and 18.8 μΜ, and Vmax values 18.3 and 1 2.8 pmol Mg"1 min"1 for ΕΑΑΤ1 τι (blue) and ΕΑΑΤ1 τ2 (red), respectively.
Figure 8. Deuterium exchange at 20 °C. a-c, Deuterium uptake kinetics at 20 °C of examples peptides covering both helical and non-helical regions of EAATI WT (grey), ΕΑΑΤ1 -Π (blue), and ΕΑΑΤ1 τ2 (red), respectively. Solid lines represent double- exponential fits to the data, and dotted lines the expected deuterium kinetics of unfolded and solvent exposed peptides, d-f Deuterium incorporation after 1 h at 20 °C in EAAT1 TI (d), EAAT1 Τ2 (e), and EAATI WT (f) mapped into the structure of the EAAT1 cRYTs (PDB 5LLM) trimer viewed from the extracellular medium (upper panel), as well as the scaffold (ScaD) and the transport (TranD) domains viewed from the membrane (lower panel), respectively. These domains are depicted separately for clarity of display. In the trimeric depiction, arrows point to the interface between protomers. Deuterium incorporation was calculated as an average of three experiments, and normalized to the maximal theoretical incorporation based on the number of backbone amide available for exchange in each peptide. A scale bar representing deuterium incorporation is depicted (d).
Figure 9. Melting of trimeric transporters, a-c, Size-exclusion chromatograms of purified ΕΑΑΤ1 τι (a), ΕΑΑΤτ2 (b), and the chimeric transporter EAAT1 -ScaD-n-TranDT2 (c), respectively, pre-heated at different temperatures. Chromatograms show how the trimeric form of the transporters that elutes at ~3.0 ml melts into lower oligomeric state(s), most likely monomers, that elute at ~ 3.5 ml. d-f Melting curves depicting the change in fractional area of the chromatographic peak corresponding to the trimeric transporters (black symbols), as a function of the temperature pre-pulse in ΕΑΑΤ1τι (d), ΕΑΑΪΤ2 (e), and EAAT1 -ScaD-n-TranDT2 (f), respectively. Solid lines indicate fits of a Hill-like equation (see methods) to the data with TSO-FSEC values 49.3, 38.6 and 45.7 °C, and n values -21.5, -9.3, and -18.3 for EAAT1 T1 , EAAT1 T2, and EAAT1 - ScaD-n-TranDT2, respectively. The total area under the chromatogram at each temperature, normalized to that at 4 °C, is also shown (empty symbols), and remains relatively constant at all temperatures.
Figure 10. Bimodal m/z envelopes, a-b, m/z envelopes of an example peptide covering residues 174-184 of ΕΑΑΤ1 τι (a), ΕΑΑΤτ2 (b), at different pre-pulse temperatures. Solid symbols represent the average of three experiments and error bars represent s.e.m, and are superimposed on the m/z spectrum of a representative experiment. Solid lines represent fits of a double Gaussian equation to the data, as well as their low- and high-m/z components, c-d, melting plots of the low-m/z component of peptides covering residues 174-184 (c) and 187-194 (d) of ΕΑΑΤ1 τι (solid circles), ΕΑΑΪΤ2 (solid triangles). Solid lines indicate fits of a Hill-like equation to the data with T50-HDX-GAUSS values °C, and n values for peptide 174-184 in ΕΑΑΤ1 τι and ΕΑΑΤ1 τ2, respectively, and TSO-HDX-GAUSS values °C, and n values for peptide 187-194 in ΕΑΑΤ1τι and EAAT1 T2, respectively.
Figure 1 1 . Local thermal unfolding, a-b, Deuterium uptake kinetics at different pre- pulse temperatures of an example peptides containing residues 174-184 in ΕΑΑΤ1 τι (a), and ΕΑΑΤ1 τ2 (b), respectively. Solid lines represent double-exponential fits to the data, and dotted lines the expected deuterium kinetics of the unfolded and solvent exposed peptide. Plots in a-b depict an average of three experiments, and error bars represent s.e.m. The inset represents the temperature protocol used to prepare the protein samples, c-d Apparent melting temperatures of the peptides folded state in EAAT1 TI (C), and EAAT1 T2 (d) are mapped into the structure of the EAATI CRYST (PDB 5LLM) trimer viewed from the extracellular medium (upper panel), as well as the scaffold (ScaD) and the transport (TranD) domains viewed from the membrane (lower panel), respectively. These domains are depicted separately for clarity of display. The color code representing the apparent melting temperature values is depicted in a scale bar.
Figure 12. Size exclusion chromatograms of detergent-solubilized GFP-fusions of NPC1 L1WT (front, row 1 ) and NPC1 L1 T1 (back, row 2) from lysates of HEK293 cells. Figure 13. Alignment of ΕΑΑΤ1 τι , ΕΑΑΤ1 τ2 and EAATI WT. Amino acid residues of EAAT1 TI , EAAT1 T2 and EAATI WT are compared. The boundaries of the a-helices (cylinders) in the TranD and the ScaD issued from the structural analysis of the EAAT1 CRYST are shown. ΕΑΑΤ1 τι and EAATI WT share 465 amino acid residues (85 % identity), while ΕΑΑΤ1τ2 and EAATI WT share 513 amino acid residues (95% identity).
Figure 14. Alignment of NPC1 L1WT and NPC1 L1 TL Amino acid residues of NCP1 L1 TI and NCP1 L1WT are compared. NPC1 L1 TI and NPC1 L1WT share 1239 amino acid residues (91 % identity).
Example 1 : THERMAL STABILIZATION OF HUMAN EXCITATORY AMINO ACID TRANSPORTER 1 FOR IN VITRO STRUCTURAL AND FUNCTIONAL STUDIES
This Example describes a process to improve the human Excitatory Amino Acid Transporter 1 (EAAT1 ) thermal stability in detergent solutions, as well as the determination of the X-ray structures of EAAT1 thermostable variants. In particular, amino acid mutations that increase thermal stability have been introduced in EAAT1 that make the transporter amenable for purification in detergent solutions, while preserving the neurotransmitter transport function of the wild type protein. This feature allowed to solve the first three-dimensional structures of thermostable EAAT1 variants and unravel a novel allosteric mechanism of inhibition.
Human members of the solute carrier 1 (SLC1 ) family of transporters take up excitatory neurotransmitters in the brain and amino acids in peripheral organs. Dysregulation of their functions is associated to neurodegenerative disorders and cancer. Here the first crystal structures are presented for a human SLC1 transporter thermostabilized according to the method of the invention, the excitatory amino acid transporter 1 (EAAT1 ), with and without allosteric and competitive inhibitors bound. The structures show novel architectural features of the human transporters, including intra- and extracellular domains with potential roles in transport function, as well as regulation by lipids and post-translational modifications. The coordination of the inhibitor in the structures and the change in the transporter dynamics measured by hydrogen-deuterium exchange mass spectrometry, reveal an allosteric mechanism of inhibition, whereby the transporter is locked in the outward-facing states of the transport cycle. These results provide unprecedented insights into the molecular mechanisms of function and pharmacology of human SLC1 transporters.
SLC1 transporters constitute a large family of ion-coupled amino acid transporters present in all kingdoms of life1. In humans, there are seven SLC1 transporters that share 40-70% amino acid identity (Fig. 4) and have evolved to serve two specialized functions2: in the central nervous system, SLC1 excitatory amino acid transporters (EAAT1 -5) take up the neurotransmitter glutamate into the cell. In peripheral organs, EAATs take up glutamate and aspartate, while neutral amino acid transporters (ASCT1 -2) exchange small amino acids between the extra- and intracellular compartments, contributing to the cellular solute homeostasis.
Glutamate is the most important excitatory transmitter in the mammalian brain and is involved in most aspects of brain physiology, from development to cognition3. Notably, most of the glutamate in the brain is intracellular, and it has to be continuously pumped into the cytoplasm to allow for rounds of transmission and prevent cytotoxicity. This essential neurological function is done by the EAAT1 -5 isoforms expressed at the plasma membrane of astrocytes and neurons. In particular, astroglial EAAT1 and EAAT2 orthologs are highly expressed in the hind- and forebrain, respectively, and are responsible for most of the glutamate uptake in the rodent brain4. EAATs are powerful molecular pumps capable of maintaining up to 104-fold glutamate gradients by using energy stored in sodium, proton and potassium gradients5. Remarkably, their dysregulation has been associated with several neurological diseases, including amyotrophic lateral sclerosis6, ataxia7 8, stroke9, depression10 and glioma11 , making them important drug targets. Moreover, they are also expressed in intestine and kidney, where mutations in EAAT3 have been associated to dicarboxylic aminoaciduria12.
ASCTs are structurally similar to EAATs, and function as sodium-dependent neutral amino acid exchangers at the plasma membrane13. They are highly expressed in intestine, kidney and testis, where they play a key role in maintaining the amino acid cellular homeostasis. Importantly, ASCT2 is up-regulated in several forms of cancer, including melanoma14, lung15, prostate16 and breast cancer17, and it is a key drug target for the treatment and diagnosis of these diseases.
Despite the need for small compounds that selectively and allosterically modulate SLC1 human transporters, most of their pharmacology is based on substrate-analogs that inhibit transport with rather low selectivity among EAAT60 and ASCT61 isoforms, respectively. Notably, the only known selective allosteric modulators of SLC1 transporters are a series of non-competitive EAAT1 -selective inhibitors, of which 2-Amino-4-(4-methoxyphenyl)-7-(naphthalen-1 -yl)-5-oxo-5,6,7,8-tetrahydro-4H- chromene-3-carbonitrile (UCPH101 ) is the best studied62 63. However, its mechanism of action is still poorly understood at the molecular level. Despite the expression of wild type EAAT1 at the cell surface of HEK293 cells where the neurotransmitter function is retained (Fig.7c), purification and reconstitution of these transporters in synthetic liposomes lack this function (Fig.7d). The detergent solutions used for solubilization and purification induces irreversible unfolding events in the transporters that render them inactive. Human wild type EAAT1 is extensively unfolded in detergent-lipid micelles loosing several secondary structural elements important for oligomerization and transport function.
The process used and disclosed herein to prepare stable and functional EAAT1 is general, fast, and cost-effective, and can be applied to other human membrane protein families. Moreover, with small variations the method can be applied to membrane proteins from other species, in particular pathogenic bacteria and viruses, or even to globular soluble proteins of different origin.
The following steps have been carried out:
a) The amino acid sequences of SLC1 transporters from selected vertebrate animals used as target sequence that share less than 95% sequence identity with each other have been aligned using JALVIEW software59. Other softwares for Multiple Sequence Alignment may be used such as Clustal W algorithm, or others that can be identified on are disclosed in above paragraph [028]. The parent (or query) sequence was the amino acid sequence of the human protein.
b) At each position in the alignment, a consensus residue was defined when it met all the below criteria: i) the residue had the highest frequency of occurrence among the aligned sequences, and this frequency is >20%; ii) the residue occupied a position in the alignment in which gaps constitute < 30% of the entries; iii) the residue's frequency of occurrence differed by more >10% from the corresponding frequency of the residue in the target/parent sequence (human sequence: EAATI WT);
c) In a first aspect of the process to increase the stability residues in the target protein were substituted for consensus residues in regions where secondary structural elements (e.g. alpha helices) are predicted, and in the short unstructured loops connecting them. The EAAT1 homologous transporter EAAT1 thermostable 1 (EAAT1 TI) has been generated using this approach and has the following sequence: Sequence ID No.1
MTKSNGEEPKMGGRMERFQQGVSKRTLLAKKKVQNITKEDVKSFLRRNALLLLTVL AVILGVVLGFLLRPYPLSPREVKYFAFPGELLMRMLKMLILPLIVSSLITGLASLDAKA SGRLGMRAWYYMSTTIIAWLGIILVLIIHPGKGTKENMHREGKIVRVTAADAFLDLIR NMFPENLVEACFQQYKTVYEKRSFKVPIQANETLVGAVINNVSEAMETLTRITEELV PVPGSVDGMNVLGLWFSIVFGIALGKMGEQGQLLVDFFNSLNEATMKLVAIIMWYA PLGILFLIAGKIVEMEDLEVLGGQLGMYMVTVIVGLVIHGLIVLPLIYFLITRKNPFVFIA GILQALITALGTSSSSATLPITFKCLEENNGVDKRITRFVLPVGATINMDGTALYEAVA AIFIAQVNNYELDFGQIITISITATAASIGAAGIPQAGLVTMVIVLTAVGLPTDDITLIIAVD WLLDRFRTMVNVLGDALGAGIVEHLSRKELEKQDAELGNSVIEENEMKKPYQLIAQ DNETEKPIDSETKM
To improve the stability of the transporters in detergent solutions, 44 and 33 amino acid residues have been substituted in the transmembrane helices of the ScaD and TranD respectively, for consensus amino acids among SLC1 animal homologs (Fig.7a where the alpha carbon atoms of the exchanged amino acid residues are illustrated as spheres and Fig. 13 where the sequence of the target protein and the sequence of the mutated protein are aligned). Most of the mutated positions localize to hydrophobic regions of the transporter facing either the trimeric interface of the lipid bilayer, and involve conservative substitutions among hydrophobic amino acids (lie, Leu, Val, Met). EAAT1 TI is 85% identical and 92% similar to EAAT
c') According to an optional aspect of the process, an additional step consisted in calculating pairs of residues that strongly co-evolved, using EVcouplings software67 (http://evfold.org/evfold-web/evfold.do) (or others having similar functionality), and substituting these pairs of residues in the target sequence for consensus residues. In a particular embodiment of the process, this additional step is performed so that the substitutions performed in step c) are limited to co-varying residues. The EAAT1 homologous transporter EAAT1 thermostable 2 (ΕΑΑΤ1 τ2) has been generated using this approach and has the following sequence:
Sequence ID No.2
MTKSNGEEPKMGGRMERFQQGVSKRTLLAKKKVQNITKEDVKSYLFRNAFVLLTVT AVILGTILGFTLRPYRMSYREVKYFSFPGELLMRMLKMLVLPLIVSSLITGLAALDSKA SGKMGMRAVVYYMTTTIIAWLGIILVLIIHPGKATKENMHREGKIVRVTAADAFLDLIR NMFPENLVEACFKQFKTNYEKRSFKVPIQANETLVGAVINNVSEAMETLTRITEELVP VPGSVNGVNVLGLWFSIVFGIALGKMGEQGQALREFFDSLNEAIMKLVAVIMWYAP VGILFLIAGKIVEMEDMGVIGGQLAMYMVTVIVGLLIHAVIVLPLIYFLVTRKNPWVFIG GILQALITALGTSSSSATLPITFKCLEENNGVDKRITRFVLPVGATINMDGTALYEAVA AIFIAQVNNFELNFGQIITISITATAASIGAAGIPQAGLVTMVIVLTAVGLPTDDITLIIAVD WFLDRFRTMVNVLGDALGAGIVEHLSRHELKNRDVEMGNSVIEENEMKKPYQLIAQ DNETEKPIDSETKM
In order to decrease the amount of mutagenesis and retain the stability of the purified transporters in detergent solutions, the inventors hypothesized that pairs of co-varying residues predicted to be in close proximity play an important role on the stability of proteins. Accordingly, the consensus mutagenesis was restricted to residues that show strong-covariance within the SLC1 family (Fig.7b where the alpha carbon atoms of the exchanged amino acid residues are illustrated as spheres and Fig. 14 where the sequence of the target protein and the sequence of the mutated protein are aligned). 15 and 14 amino acid residues have been substituted in the transmembrane helices of the ScaD and TranD respectively. ΕΑΑΤ1τ2 is 95% identical and 97% similar to EAAT1 wt.
The homologous transporters ΕΑΑΤ1 τι and ΕΑΑΤ1 τ2 have higher thermostability in detergent solutions and showed an increase in the apparent melting temperature (Tm) of ~13, and ~5 °C, respectively (Fig. 1 ), compared to EAATIWT. The homologous transporters ΕΑΑΤ1 τι and ΕΑΑΤ1 τ2 have conserved transport mechanism (i.e. functional) are when expressed in cells (Fig. 7c). In other words, these mutants are functional. The homologous transporters ΕΑΑΤ1 τι and ΕΑΑΤ1τ2 also have a conserved transport mechanism (i.e. are functional) upon reconstitution of purified transporters in liposomes (Fig. 7d and 7e). ΕΑΑΤ1 τι and ΕΑΑΤ1τ2 also have a good solubility in detergent solutions. As an example, ΕΑΑΤ1 τι shows similar level of neurotransmitter uptake in cells compared to EAATIWT, substantially higher solubilization yields in all detergents tested, and robust glutamate uptake upon purification and reconstitution in synthetic liposomes (Fig. 7d). Moreover, glutamate transport in liposomes was strictly dependent on opposite gradients of sodium and potassium across the bilayer, and the rate of transport was dependent on the concentration of L-glutamate, with a Km (~30μΜ) similar to the one reported for EAATIWT (Fig. 7e). The transport mechanism is therefore conserved for ΕΑΑΤ1 τι and EAAT1 T2. The neurotransmitter experiment shows that the detergent solutions used for solubilization and purification induce unfolding events that render the EAAT1 WT inactive, while ΕΑΑΤ1 τι and ΕΑΑΤ1 τ2 remain functional. Notably, due to the increase in stability the homologous transporters ΕΑΑΤ1 τι and ΕΑΑΤ1 τ2, but not EAATI WT, could be purified as properly folded transporters and reconstituted in liposomes as functional proteins (Fig. 2). In turn, the stable and functional purified transporters could be used in high-resolution structural studies and would be suitable candidates for high- throughput screening of small compounds using in vitro techniques for drug discovery. Moreover, purified ΕΑΑΤ1 τι and ΕΑΑΤ1 τ2 can be used to develop monoclonal molecules, like antibodies and nanobodies that recognize the transporters in the cell surface using three-dimensional epitopes, as opposed to the linear and intracellular epitopes currently used. Mutations based on consensus amino acids of animal SLC1 transporters kinetically trapped EAAT1 in folded and functional states, and allowed the analysis of intermediate unfolded states along their denaturation pathways.
Example 2: STRUCTURE AND ALLOSTERIC INHIBITION MECHANISM OF EXCITATORY AMINO ACID TRANSPORTER 1
In structural terms, most knowledge of the molecular mechanism of transport and pharmacology of SLC1 transporters comes from the prokaryotic homolog GltPh that has been crystallized in the main conformational states of the transport cycle, outward-22 and inward-facing states23 24, as well as in complex with a non-selective and competitive inhibitor of the EAATs25, DL-threo-p-benzyloxyaspartic acid (TBOA). However, the presence of amino acid insertions and deletions, as well as important differences in the transport function and pharmacology of GltPh, make this homolog a limited structural model to understand the molecular mechanism of the human SLC1 proteins.
Here 3.1 -3.3 A X-ray crystal structures of thermostable EAAT1 variants in complex with a substrate (L-aspartate), and the allosteric inhibitor UCPH101 are presented. The structures, and supporting functional data, show new architectural features of the EAATs and ASCTs, and unravel the allosteric mechanism of UCPH101- like inhibitors in atomic detail. Taken together, these structural data can prove useful for the design of novel allosteric compounds with improved selectivity for both EAATs and ASCTs. EAAT1 engineering and crystallization
Purified wild-type EAAT1 shows a poly-disperse size-exclusion chromatogram in detergent solutions, and lacks transport activity upon reconstitution in synthetic liposomes (Fig. 3a). To confer stability to EAAT1 , consensus mutations26 were introduced in the predicted transmembrane helices, and obtained a biochemically stable and functional transporter (Fig. 4). As the protein was still refractory to crystallization, the hypothesis was made that the extracellular region of the transporter between the predicted transmembrane helix 3 (TM3) and the cytoplasmic half of TM4 (TM4c) could preclude crystal growth due to the presence of a long amino acid insertion compared to GltPh. A chimeric transporter substituting this region with the corresponding amino acid sequence from ASCT2 was therefore engineered, which is the shortest in this region among human SLC1 transporters (Fig. 4). The resulting transporter, called EAATI cryst, shares an overall ~75% sequence identity with wild type EAAT1 , and up to ~90% identity at the C-terminal core of the protein, where the transported substrate and coupled ions are expected to bind25 27"31. Importantly, purified EAATI cryst reconstituted in liposomes showed robust glutamate uptake that depended on opposite gradients of sodium and potassium ions across the bilayer (Fig. 3a), and was inhibited by the EAAT1 -selective compound UCPH101 in a concentration- dependent fashion (IC50 of 4.5±0.3 μΜ, Hill coefficient 0.92±0.07) (Fig. 3b). These data show that the transport mechanism and pharmacological selectivity are conserved in EAAT1 cryst-
Notably, EAATI cryst formed crystals in the presence of UCPH101 that diffract X- rays anisotropically and up to 3.25 A resolution, and its inhibitor-bound structure was solved by molecular replacement (see Methods and Table 1 ). As EAATI cryst was refractory to crystallization in the absence of UCPH101 , it was reasoned that mutations in the inhibitor-binding pocket could aid with the crystallization of the transporter. A construct carrying M231 I and F235I mutations (EAAT1 Cryst-ii; Fig. 4), crystallized both in the presence and absence of the inhibitor, and diffracted X-rays up to 3.1 and 3.32 A resolution, respectively (Extended Data Table 1 ). Remarkably, purified EAAT1 Cryst-ii reconstituted in liposomes also showed robust sodium- and potassium-dependent glutamate uptake, while the UCPH101 IC50 increased >30-fold (>131 ±38 μΜ, Hill coefficient 0.92±0.0; Fig. 3a, b), as expected for mutations in the binding pocket of the inhibitor (see below). Domain organization
The structure of EAAT1 cryst shows a symmetric homotrimer in a substrate- and UCPHi 01 -bound outward-facing conformation (Fig. 3c-e). EAATI cryst adopts an overall GltPh-like fold22 23, in which each monomer is composed of two domains: a trimerization or scaffold domain (ScaD), including TM1 -2 and TM4-5; and a transport domain (TranD), including TM3, TM6-8 and re-entrant helical loops 1 -2 (HP1 -2; Fig. 3f). The ScaD forms all inter-subunit contacts through residues in TM2, TM4 and TM5, that include six salt bridges and bury ~3,000 A2 from each subunit. Hence, the three ScaDs form a compact central structure with a propeller-like shape that ensures the trimeric form of the transporter and anchors it to the membrane.
The three TranDs are more peripheral and localize between the blades of the propeller, making protein contacts exclusively with the ScaDs of their own monomer. The TranD-ScaD interface buries ~3,500 A2, including a conserved salt bridge between E256 and K364. This interface is mainly formed by cytoplasmic residues in HP1 , TM7, and TM3 (TranD), and TM2, TM4c, and TM5 (ScaD). However, on the extracellular side additional contacts occurred between HP2 and TM4, through residues that are well conserved among human transporters.
Substrate and ion translocation in SLC1 transporters is thought to occur through large rigid-body movements of the TranD, relative to the static ScaD, that move the cargo in an elevator-like fashion across the membrane23 32. Thus, during the isomerization to the inward-facing state the TranD-ScaD interface changes drastically on the TranD side, and the novel features observed at this interface in EAATI cryst might influence the distinct TranD dynamics in human SLC1 proteins.
Transport domain
One of the most remarkable architectural features of the EAAT1 cryst TranD is at the TM8 level, in which deletions and insertions compared to GltPh reshape this helix and its interactions with neighboring structural elements important for transport. In EAATI cryst, TM8 can be divided into extracellular (TM8a), transmembrane (TM8b), and cytoplasmic (TM8c) helices. The loop connecting TM8a and the C-terminal helix of HP2 (HP2b) is six residues shorter in human SLC1 transporters. Consequently, the extracellular ends of TM8a and HP2b are in close proximity and engage in hydrogen bonding and hydrophobic interactions. HP2 is a key component of the gating machinery that controls the access of substrate and ions to their binding sites in the TranD25 33-35, and its interactions with TM8b likely play an important role in determining HP2 movements. Notably, several single-cysteine mutations at positions along TM8a in EAAT136, and in a rodent EAAT2 ortholog37 impaired glutamate transport, highlighting the significance of this extracellular region for function.
At TM8b level, strong electron density for the substrate (L-aspartate) was found and one of the sodium-binding sites previously observed in the archaeal homologs of SLC1 family (Na2), which was modeled with similar coordination than in their structures25 31. Remarkably, the carboxylate group of D456 (TM8b), which coordinates the alpha-amino group of the substrate, is also at hydrogen bond distance with the hydroxyl group of S343 (HP1 ). Moreover, the guanidinium group of R457 (TM8b) engages in hydrogen bonding with HP1 residue G341 , and possibly L340 and T342 that point their backbone carbonyl oxygen atoms towards TM8b. Residues S343 and R457 are well conserved in human SLC1 transporters, and substitutions at equivalent positions in EAAT1 (S363 and R477)38, and EAAT3 (R445)39 inactivate transport. In addition, the loss-of-function mutation R445W in EAAT3, equivalent to R457 in EAATI cryst, causes human dicarboxylic aminoaciduria due to the lack of aspartate/glutamate reabsorption function in the kidney12. Overall, the functional studies and our structural data converge to suggest that interactions between conserved human residues at HP1 and TM8b are important to the correct folding and function of the transporters.
On the cytoplasmic side, hydrophilic TM8c extends beyond the membrane plane through a hydrophilic helix (TM8c), and makes contact with residues in TM3 and TM7a. Notably, EAAT2 deletion mutants in this region have a deleterious effect on transport function and membrane trafficking40. Accordingly, a deletion of TM8c beyond E501 in EAAT1 (equivalent to E500 in EAAT2 and E481 in EAATI cryst) decreased glutamate uptake rate by ~2-fold. The functional data, and the amino acid conservation in TM8c among EAATs, underscore the pivotal role of this structural motif in protein folding and transport kinetics.
Scaffold domain
The ScaD is less conserved than the TranD in the SLC1 family. In particular,
TM4 is highly divergent (Fig. 4), and shows several unique architectural features in EAATI cryst. On the extracellular side, TM4a forms inter- and intra-monomeric contacts with TM2 and HP2, respectively. Moreover, an amino acid insertion between TM4b and TM4c (TM4b-c loop) that appeared during the evolution of eukaryotic transporters protrudes into the central vestibule of the EAAT1 cryst trimer. The TM4b-c loop forms the center of the propeller, and makes extensive contacts within and between protomers. Due to the lack of electron density it was not possible to model the outermost residues of the TM4b-c loop (Y200-V210), but they are expected to reach out to the bulk solvent, and expose one of the predicted N-glycosylation sites of the transporter (N204). Notably, despite the lack of sequence identity among human SLC1 transporters, all of them contain predicted N-glycosylation sites in the TM4b-c loop, suggesting a common role of this loop in the posttranslational processing of these proteins.
An additional novel feature of the EAAT1 cryst ScaD architecture is the N-terminal extension of TM1 by an amphipathic helix (TM1 a). Positioned nearly parallel to the membrane plane, TM1 a forms the tips of the blades in the propeller (Fig. 3c). Remarkably, it does not form inter- or intra-monomeric contacts, and its position and amphipathic nature suggest that TM1 a somehow interacts with the inner leaflet of the membrane. Indeed, there is a hydrophobic crevice between TM1 a and HP1 a from the same monomer, where strong non-protein electron density in EAAT1 cryst was observed that likely corresponds to bound detergent or lipid molecules. Interestingly, a second hydrophobic crevice is observed between the extracellular part of TM4 and HP2, where there is also strong non-protein electron density. A similar crevice was also noted in a substrate-bound structure of GltPh22.
The lipidic composition of the bilayer regulates the function of SLC1 transporters41 -45. Because TM1 a-HP1 a and TM4-HP2 hydrophobic crevices are at the interface between the TranD and ScaD, where large conformational changes are expected to occur during substrate translocation, they might constitute sites for lipid regulation of transport function.
UCPH101 binding site
The structure of EAAT1 cryst showed strong electron density for UCPH101 in a hydrophobic pocket facing the inner leaflet of the membrane on the interface between the TranD and ScaD (Fig. 3d,e and Fig. 5a, b). This pocket is formed by residues in TM3, TM7 and TM4c, and extends the TranD-ScaD interface by ~ 500 A2.
The chromene skeleton of UCPH101 , the parental group of the UCPH series of compounds, is buried deeply in the domain interface, and coordinated by a direct ring- stacking interaction with F369 (TM7a), as well as hydrophobic interactions with G120 (TM3), V373 (TM7a) and M231 (TM4c) (Fig. 5b). In addition, the amine group of UCPH101 forms a hydrogen bond with the main-chain carbonyl of F369, while its carbonitrile group interacts with Y127 (TM3). The methoxy-phenyl and naphthalene groups are more peripheral and partly facing the hydrocarbon core of the membrane. Yet, the former establishes hydrophobic interactions with V124 (TM3), V373 and M231 , while the latter is mainly coordinated by F235 (TM4c). The majority of the above- mentioned residues have been reported to be important for the inhibition of an EAAT1 rodent ortholog by UCPH101 in cell assays21. Furthermore, the EAAT1 cryst double mutant M231 1-F235I (EAAT1 Cryst-n) showed >30-fold increase in UCPH101 IC50 compared to EAAT1 cryst in proteo-liposomes (Fig. 3b). Hence, there is an excellent agreement between the crystallographic and functional data.
Several mechanistically-relevant observations can be made regarding the UCPH101 binding pocket in EAAT1 cryst: i) it is over 15 A away from the substrate and sodium binding sites, suggesting that UCPH101 does not preclude substrate binding, as expected for a non-competitive allosteric inhibitor; ii) it faces the inner leaflet of the membrane, implying that UCPH101 accesses its binding site from the lipidic, and not the aqueous phase, when applied extracellularly; iii) it is fully contained in a single subunit, in agreement with the lack of cooperativity observed in proteo-liposome (Fig. 3b), and cell assays21 ; iv) a comparison of the EAAT1 -5 sequences suggests that the main determinants of UCPH101 selectivity for EAAT1 are in TM4c, where M231 and F235 are the only coordinating residues that differ between EAAT1 and all other EAATs (Fig. 4). Consistently, the equivalent residues in EAAT2 are isoleucine, and the double mutant EAAT1 Cryst-n, containing M231 I and F235I, shows a >30-fold increase in the UCPH101 IC50 compared to EAAT1 cry st- UCPH 101-unbound state
To better understand the conformational changes of the transporter induced by UCPH101 , determination of the structure of the UCPHioi-unbound state was contemplated. As mentioned above, EAAT1 cryst was refractory to crystallization in the absence of the compound and instead, the structure of EAAT1 Cryst-ii UCPHioi-unbound state was solved. For comparison, the structure of the EAAT1 Cryst-ii UCPHioi-bound state was determined, using a large excess of the compound in the crystallization conditions (see Methods). The structure of EAAT1 Cryst-n in the UCPHioi-bound state is nearly identical to that of the EAATI cryst with the exception of the mutated 1231 and I235 side chains, and a ~2 A movement of UCPH101 methoxy-phenyl and naphthalene groups away from them (Fig. 5c). Interestingly, the EAAT1 Cryst-ii UCPHioi-unbound structure has an overall similar conformation to the UCPHioi-bound state, but shows notable differences. First, the UCPH101 binding pocket contains no excess electron density, and the side chain of F369 moves outward by as much as 1.9 A, partly occupying the volume for UCPH101 chromene group (Fig. 5c). Second, there is a small rigid-body movement of the entire TranD that is shifted by as much as 0.7 A, compared to the UCPH101 -bound structures. Interestingly, this conformational change shows the EAATI cryst TranD is able to undergo rigid-body movements relative to the ScaD, and highlights the importance of such movements for the function of the human transporters, as it has been shown for the prokaryotic homolog23 24
The structural changes observed in the UCPHioi-unbound structure unambiguously demonstrate that the assigned binding pocket of UCPH101 is correct, and that within the restricted environment of the crystal lattice, UCPH101 induces both local and global conformational changes of the transporter that optimize its coordination in an outward-facing state.
Transport domain dynamics
The coordination of UCPH101 in the crystal structures, wedged between the
TranD (TM3 and TM7a) and the ScaD (TM4c), as well as the effect of the M231 1-F235I mutations on the UCPH101 potency strongly suggest that UCPH101 inhibits transport by trapping the transporter in an outward-facing state. Consistently, the rigid-body movements of the TranD to isomerize into the inward-facing state would separate the coordinating residues in the TranD from those in the ScaD, and disrupt the UCPH101 coordination. Hence, under equilibrium conditions where the transporters are sampling outward- and inward-facing states, the expected effect of UCPH101 binding is to shift the equilibrium in favor of the outward-facing state.
To gain insights into the effects of UCPH101 binding to the transporters at equilibrium, the detergent solubilized EAATI cryst by hydrogen-deuterium exchange mass spectrometry (HDX-MS) was probed. HDX-MS measures the rate of exchange of backbone amide hydrogen atoms that depends on solvent accessibility and hydrogen bonding, and provides valuable information on the dynamics and conformational changes of proteins46 47.
The HDX behavior of the EAAT1 cryst was compared in the presence and absence of UCPH101. Overall, the deuterium uptake pattern of EAAT1 cryst shows dynamic structural elements in both the TranD and ScaD, and reveals the unstructured and solvent-exposed nature of several regions that were not resolved in the crystal structures, including the TM3-TM4a (peptide 153-173) and TM4b-c (peptide 200-208) loops, as well as the N- (peptide 1 -28) and C- termini (peptides 490-522).
Binding of UCPH101 significantly decreased deuterium uptake in several areas of the TranD including its binding pocket (residues 1 12-123 and 370-374), and the surrounding area (residues 354-369), while it left the uptake in the ScaD unchanged. It also decreased deuterium uptake in distant residues (336-349 and 420-430) at the tips of HP1 and HP2 involved in substrate coordination and occlusion, suggesting that UCPH101 induces conformational changes in the transporter upon binding. To gain insights into the nature of those conformational changes, the TranD areas in which UCPH101 decreased uptake was first compared with those buried at the interface with ScaD in the EAAT1 cryst structure, and found that they correlate remarkably well. Second, a model of the EAAT1 cryst inward-facing state was built, based on a recently solved structure of GltPh24, to assess the changes in solvent accessibility in a possible transition between inward- and outward-facing states. Indeed, the comparison between the structure and the model shows that the UCPHioi-modified areas detected by HDX-MS transit as rigid bodies from being solvent-exposed, in the inward-facing state, to buried at the TranD-ScaD interface, in the outward-facing state. Such conformational change is expected to decrease the dynamics of alpha helices and/or the solvent accessibility of the loops in the UCPHioi-modified areas and thus, is consistent with the observed decrease in deuterium uptake. Overall, the HDX-MS and structural analysis support the stabilization of the outward-facing state, at the expense of the inward-facing state(s), induced by UCPH101.
UCPH101- and TBOAiFB-bound state
The distant position of UCPH101 from the substrate and the HP2, a structural element that controls extracellular access to the binding site25, suggests that the UCPH101 -bound transporters could undergo the conformational changes required to exchange the substrate with the extracellular solution. To test this, the crystal structure of EAAT1 cryst in complex with both UCPH101 and (2S,3S)-3-[3-[4- (trifluoromethyl)benzoylamino]benzyloxy]aspartate (TBOATFB), a potent and nonselective TBOA derivative48, was solved at 3.7 A resolution (Table 1 ).
Overall, the UCPHI OI -TBOATFB bound structure is similar to that of the UCPHioi-bound state, with the exception of HP2 that adopts an "open" conformation and packs against the TM4b-c loop, disrupting the coordination of the Na2 (Fig. 6a, b). These conformational changes resemble those previously observed in the structure of the GltPh-TBOA complex, and are in excellent agreement with the proposed competitive inhibitory mechanism of TBOA-like compounds25.
In the substrate-binding site, excess electron density was observed for the bulky
TBOATFB (Fig. 6c), but due to lack of resolution, it was not possible to unambiguously orient the compound. In order to fit the TBOATFB molecule into the density, the TBOA moiety of TBOATFB was initially positioned using the TBOA-bound GltPh structure as a guide. In this position, it remained stable during several successive cycles of refinement that yielded a reasonable fit into the electron density. The additional benzoylamino and trifluoromethyl groups of TBOATFB localized in a hydrophobic cavity mainly formed by residues in HP1 b and TM7a, and possibly by residues in TM2 and TM4c. Interestingly, the interactions of these groups with the transporter could explain the ~1500-fold increase in inhibitory potency of TBOATFB, compared to TBOA, that has been observed in EAAT1 transport assays48.
Indeed, it was also observed that UCPH101 bound in this structure with an identical coordination than in the substrate-bound state. Therefore, despite the lower resolution of the TBOATFB bound structure, it shows that UCPH101 binding at its allosteric site does not preclude the movements of HP2 involved in substrate and sodium binding from the extracellular solution.
Inhibitory mechanisms of EAAT1
The structures of EAAT1 cryst reveal new architectural features of human SLC1 transporters, and the first non-competitive inhibitory mechanism of this family of proteins in molecular detail. UCPH101 is an EAAT1 -selective inhibitor with a bipartite coordination by residues in both the TranD and ScaD, and the downward rigid-body movements of the TranD during transport disrupt such coordination. This implies that upon binding, UCPH101 "glues" the TranD to the ScaD in the outward-facing states, and precludes the translocation reaction of the transport cycle, but not the substrate binding/unbinding reactions from the extracellular solution.
The inhibitory mechanism of UCPH 101 contrasts with that of substrate-analog competitive inhibitors like TBOA. The binding pocket of TBOA-like compounds overlap to some extent with that of the substrate25, and some of these molecules can bind the transporter from both the extra- and intracellular aqueous solutions49 50. Therefore, they inhibit transport by precluding substrate binding on either side of the membrane. Moreover, TBOA-like compounds are not selective among glutamate transporters due to the high amino acid conservation in the substrate-binding sites.
The mechanistic differences observed in UCPH101 over other known inhibitors make it an extremely valuable pharmacological tool, to isolate and study the conformational changes that EAAT1 undergoes upon substrate and ion binding. Remarkably, the UCPH101 allosteric binding site observed in EAAT1 cryst highlights a cavity that can facilitate the design of selective compounds for other human SLC1 transporters, and possibly the long-sought positive modulators of glutamate uptake.
To sum up the results relative to the EAATCRYST, the structure shows that the transporter is a homo-trimer, in which each protomer contains two structural and functional domains, the scaffold (ScaD) and transport (TranD) domains, previously observed in the structures of a glutamate transporter prokaryotic homologue. The ScaD forms the inter-subunit interface through extensive contacts between transmembrane helices TM2 and TM4 on the extracellular half of the membrane, as well as between TM4 and TM5 on the cytoplasmic side. The remaining alpha helical regions of the protein, including TM3 and TM6-8, as well as two re-entrant helical loops (HP1 -2) fold into the TranD that encages the substrate and the coupled ions and translocates them across the membrane through rigid-body movements in an "elevator-like" fashion.
Example 3: STRUCTURAL COMPARISON BETWEEN EAAT1WT, EAAT1Ti AND EAAT1 T2
EAATI WT is inactive in transport assays, while ΕΑΑΤ1 τι and ΕΑΑΤ1 τ2 are active. The detergent solutions used for solubilization and purification may induce unfolding events that render the wild type transporter inactive. The structural differences between the two mutants and the wild type transporter in detergent solutions have therefore been compared. Hydrogen-deuterium exchange behavior of the three proteins using a protease-induced fragmentation approach linked to mass spectrometry (HDX-MS) was used. As previously explained, HDX-MS measures the rate of deuterium exchange of the hydrogen amide in the protein backbone, which depends strongly on the presence of secondary structure due to the engagement of the amide group in hydrogen bonding as well as access to the aqueous solvent. Therefore, HDX-MS provides valuable information on protein folding, stability and dynamics.
EAAT1 TI and ΕΑΑΤ1 τ2 showed similar overall HDX patterns when assayed at 20 °C in detergent solutions, with sequence coverage of ~70 % and unimodal isotopic envelopes across all peptides measured. Analysis of the HDX kinetics (Fig. 8 a-c) revealed that most peptides covering the alpha-helical regions of the TranD and ScaD take up deuterium at rates between 10~2 to 10~5 s~1 , which are several orders of magnitude slower than predicted for unstructured peptides with the same sequence (102 s"1). Moreover, peptides partly covering at least TM4-5, as well as TM8 and HP2, were completely protected and do not show any deuterium uptake. In contrast, peptides covering predicted unstructured regions like the N- and C-termini, as well as extracellular loops between TM3-TM4a and TM4b-TM4c respectively, which were not resolved in the EAATI CRYST structure, showed saturating levels of deuterium uptake already at 10s, the shortest time measured in the experiments, consistent with a solvent accessible backbone in these regions being fully solvent accessible and lacking secondary structure. To sum up, the wild transporter should have secondary structure but it loses it in detergent and therefore loses its function, while the mutated proteins ΕΑΑΤ1τι and ΕΑΑΤ1 τ2 keep secondary structure. Importantly, the HDX patterns of EAAT1 τι and EAAT1 Τ2 are similar to that of EAAT1 CRYST and EAAT1 CRYST- ii, which are also functional transporters when reconstituted in liposomes.
The HDX behavior of EAATI WT was strikingly different form the above-mentioned mutants and showed an overall dramatic increase in deuterium uptake (Fig. 8 d-f). This increase in backbone amide HDX is most prominent in two regions of the transporter: first, the cytoplasmic half of the TranD, where several peptides covering TM3 (peptide 1 12-123), TM7 (peptides 390-399 and 397-404) and HP1 (peptide 357-369), including the substrate and ion binding sites revealed maximal deuterium uptake at 10s, as observed in the unstructured N- and C-termini (Fig. 8 a-c). The lack of backbone protection implies that the native helical structure of these key regions for transport function is lost upon solubilization and purification of the protein; second, several regions of the ScaD, particularly the extracellular helices of TM4 form extensive inter- subunit contact. Although deuterium uptake in these regions still reveals some level of backbone protection, these results suggest that the ScaD might undergo partial unfolding events that could compromise the oligomeric state of the transporter.
All together the HDX results show that ΕΑΑΤ1 τι and ΕΑΑΤ1 τ2 retain the native fold observed in the structure of EAATI CRYST, while the EAATIWT undergoes partial and extensive unfolding events in both the TranD and ScaD in detergent solutions. Consistently, purified ΕΑΑΤ1 τι and ΕΑΑΤ1 τ2, but not EAATI WT, showed robust sodium- and potassium-dependent neurotransmitter transport function, when reconstituted in synthetic liposomes. The above results prove that the consensus, as well as the consensus-covariance approaches, can be used as semi-rational approaches to produce a stabilized version of the human EAAT1 for biophysical analysis of its molecular mechanisms.
ΕΑΑΤ1 -Π and ΕΑΑΤ1 τ2 showed an overall similar sequence of events during thermal inactivation, despite the difference in amino acid sequence, in which the loss of quaternary structure is an essential early event in the denaturation pathway. Several lines of evidence support this conclusion: the overall kinetic stability of the ScaD is lower than that of the TranD, and mutations in the ScaD determined the stability of trimeric form of the transporters, as shown by the chimera EAAT1 -ScaD-n-TranDT2; The lowest local apparent melting (T50-HDX) observed in both ΕΑΑΤ1τι and ΕΑΑΤ1τ2 map to an extensive area of the trimeric interface formed by TM4a-c and TM2 from neighboring subunits, respectively. This means that the first temperature-induced unfolding events of the transporters occur in TM4a-c and TM2 and therefore, that they constitute important rate-limiting step(s) during thermal inactivation. There is excellent quantitative agreement between the T50s of local unfolding at the trimeric interface (T50-HDX in TM4a-c and TM2), and those that characterize the loss of trimeric state (T50-FSEC). This agreement holds for both EAAT1 Ti and EAAT1 T2 despite the ~ 10 °C difference in the T50 values between the transporters. The above results strongly argue that the loss of secondary structure in TM4a-c and TM2 causes the dissociation of the trimers.
Unfolding events occur at higher temperatures in the TranD than in the ScaD. Clear unfolding has been observed for important helical regions for substrate and ion binding (TM3, TM7-8, and HP1 -2) in EAAT1 Ti at temperatures as high as 65 °C, while the TranD of ΕΑΑΤ1 τ2 showed no major changes in deuterium uptake at 55 °C. This contrasts with the loss of function observed in ΕΑΑΤ1 τ2 pre-heated at 55 °C and suggests that thermal inactivation is due to the loss of trimeric state, and does not require extensive unfolding events of the TranD. This could be explained if dynamic regions of the transporter required for transport (e.g. HP2) were constrained by lipid or detergent molecules in the monomeric form of the transporters, but not in the trimeric one. In fact, decreased in deuterium uptake at the level of HP2 in both ΕΑΑΤ1 τι and EAAT1 T2 at high temperatures, is consistent with this line of thought.
Early unfolding events at the trimeric interface determine the lifetime of the transporters and opens the interesting possibility to target this region by mutagenesis, or even small compounds that could extend the half-life of the transporters and possibly act as the long-sought glutamate transport activators.
Example 4: THERMAL STABILITY OF EAAT1Ti AND EAAT1T2
The thermal denaturation of both mutants was assessed. Thermal denaturation is in general an irreversible process in detergent-solubilized membrane proteins. Applying temperature protocols that generate partial denaturation of the proteins offers the possibility to capture kinetic intermediates of the unfolding pathway. Size Exclusion Chromatography has first been used to analyze the effect of twenty-minute temperature pre-pulses on purified ΕΑΑΤ1 τι and ΕΑΑΤ1 τ2 in detergent solutions, at a constant protein concentration, in order to quantify the irreversible effect of temperature on protein solubility, aggregation, and oligomeric state compared to reference transporters that were not pre-heated.
ΕΑΑΤ1 -Π and ΕΑΑΤ1 τ2 reference samples maintained at 4 °C eluted as symmetric and monodisperse peaks that correspond to the trimeric form of the transporters (~3 ml elution volume), and remained stable up to 35 °C and 25 °C respectively (Fig. 9a, 9b and Fig. 9c,9d respectively). At higher temperatures the trimers melt into a lower oligomeric state that elutes at ~3.5 ml, without any signs of aggregation or insolubility, judging by the lack of high molecular-weight peaks and the constant area under the chromatograms at all temperatures respectively. The apparent melting temperatures (TSO-SEC) of ΕΑΑΤ1 τι and ΕΑΑΤ1 τ2 trimeric states were 49.06 ± 0.02 and 38.4 ± 0.3 °C respectively, demonstrating that the quaternary structure of ΕΑΑΤ1 -Π is kinetically more thermo-stable than that of ΕΑΑΤ1 τ2 (Fig.9b and 9d) Interestingly, the appearance of a single low molecular-weight peak in the chromatographic profiles at high temperatures argues that the transporters lose their quaternary structure by melting into monomers, in a cooperative process that does not involve the formation of stable dimers, and that those monomers remain soluble at pre- pulses of up to 65 °C.
In order to probe the transport function of the low molecular-weight species,
EAAT1 T2 samples pre-heated at 55 °C in liposomes were reconstituted and compared to the reference samples maintained at 4 °C. Indeed, the large decrease in neurotransmitter uptake observed in the pre-heated samples shows that the transport function of the "low molecular-weight" species has been largely impaired, and suggests that the monomeric form of the transporter is not functional.
The loss of trimeric state and transport function argues that early events during the thermal inactivation of the transporters involve the formation of partly unfolded monomers, and that the interface between the subunits plays an important role in the thermal stability of the transporters. To gain further insights into the role of the trimeric interface, a chimeric transporter with the ScaD of ΕΑΑΤ1 τι and the TranD of ΕΑΑΤ1 τ2 (EAAT1 -ScaD-n-TranDT2) was built. Notably, the melting curve of EAAT1 -ScaD-n- TranDT2 approached that of EAAT1Ti (T50-SEC of 45.2 ± 1 .0 °C) showing that key determinants of the transporter thermal stability localize to the trimeric interface (Fig. 9e,f).
LOCAL THERMAL UNFOLDING OF EAAT1Ti AND EAAT1T2
The chromatographic and functional analysis clearly established that the temperature pre-pulse protocols irreversibly generate intermediates of the thermal unfolding pathway. To gain structural insights into the unfolded state of these intermediates, pre-heated transporters were analyzed by HDX-MS using the untreated transporters as reference. Since the sample throughput by HDX-MS is limited, analysis was focused on ΕΑΑΤ1 τι and ΕΑΑΤ1 τ2 to the corresponding apparent melting temperatures observed by SEC (TSO-SEC) as well as selected temperatures below and above it.
The overall HDX pattern of ΕΑΑΤ1 τι and ΕΑΑΤ1 τ2 was similar when the transporters were pre-heated at nearly their TSO-SEC, 40 °C and 50 °C respectively. Both transporters showed a significant increase in deuterium uptake in peptides covering TM4a-b (e.g. peptide 174-184) and the extracellular part of TM2 (e.g. peptide 81 -87) indicating partial loss of secondary structure in these helices. Additional increase in deuterium uptake was observed in the cytoplasmic parts of TM2 and TM3 (e.g. peptides 105-1 1 1 and 1 12-1 15 in EAAT1 Ti), as well as TM4c (peptide 230-246 in EAAT1 T2), while HDX remained unchanged in the TranD at large. These results map the initial thermal unfolding events to the trimeric interface of the transporters and highlight the key role of this region in thermal stability.
EAAT1 TI and ΕΑΑΤ1 τ2 were also probed at pre-pulse temperatures above the
T50-SEC at which complete loss of trimeric state was observed based on SEC, 65 °C and 55 °C respectively. Under these conditions, both transporters showed a dramatic increase in HDX at the trimeric interface (TM4a-c and external TM2) that went beyond the one observed at their respective TSO-SEC. The HDX profile of the TranD differed greatly between ΕΑΑΤ1 τι and ΕΑΑΤ1 τ2: in the latter, the deuterium uptake remained unchanged compared to the reference condition, with the exception of a slight decrease in uptake observed in HP2; ΕΑΑΤ1 τι showed large HDX changes in most peptides covering the TranD including increased deuterium uptake in HP1 (e.g. peptide 361 -369) and TM7-TM8 (e.g. peptides 390-399 and 483-495 respectively). These results show that the TranD of both ΕΑΑΤ1 τι and EAAT1 i2 are more thermo-stable than their ScaD, and only undergo extensive unfolding events at pre-pulse temperatures overs 55 °C.
CORRELATION BETWEEN LOCAL UNFOLDING AND LOSS OF QUATERNARY STRUCTURE
The thermal denaturation experiments show that increasing temperatures induce on one side loss of the trimeric state, as observed by SEC, and on the other side local unfolding events that initially map to the extracellular part of the trimeric interface between TM4a-b and TM2, as observed by HDX-MS. The important question then raises regarding if there is a quantitative correlation between the two events that could establish a causative link between the local unfolding events and the loss of quaternary structure. To shed light on this problem, the apparent melting temperatures of the individual peptides obtained by HDX (TSO-HDX) were estimated and compared to the corresponding TSO-SEC, the parameter that characterizes melting of the trimeric state.
The peptides covering the extracellular helices TM4a-b (e.g. 174-184 and 187- 194) showed clear bimodal isotopic envelops in the m/z spectra, while all other peptides were unimodal. The bimodal envelopes were most apparent at pre-pulse temperatures close to the TSO-SEC (Fig. 10a and 10b), in which the amplitudes of the high- and low-mass components were similar. In contrast, at temperatures above and below the Tso-sEc the envelopes were nearly unimodal and dominated by the high- and low-mass components respectively. Using Gaussian fitting to calculate fraction of the low-mass components as a function of temperature, and estimated the apparent melting temperature of the peptides at the inter-subunit interface (TSO-HDX-GAUSS), peptides 174-184 and 187-194 in ΕΑΑΤ1 τι showed TSO-HDX-GAUSS values of 49,5 °C and 48,8 °C respectively, while those of EAAT1 T2 were 36,8 °C and 42,4 °C respectively (Fig. 10c). These values are in excellent agreement with the T50-SEC values observed for EAAT1 TI (49,0 °C) and EAAT1 Τ2 (38,4 °C), strongly suggesting a causative correlation between the unfolding of the TM4a-b helices and the loss of quaternary structure.
In order to estimate the apparent melting temperature of all peptides that showed temperature-dependent HDX changes, and not just those that showed bimodal isotopic envelopes, a kinetic analysis of the deuterium uptake data has been carried out. The deuterium uptake kinetics of all peptides is well described by three components: an initial burst, which corresponds to the uptake measured at 10 s; a slow component with uptake rates < 10~3 s~1 , and an intermediate component with rates ~ 10"1-10"2 s"1 (Fig. 1 1 a and 1 1 b). The deuterium uptake of the peptides covering the helical regions of the transporter is dominated by the slow component under reference conditions (e.g. >70% of deuterium uptake in peptide 174-184, Fig. 1 1 a) and therefore, the decrease in the fractional amplitude of this component at increasing temperatures was used as a proxy for the loss of native folded state and to extract the associated apparent melting temperature (TSO-HDX-KINET) (Fig. 1 1 c and 1 1 d).The peptides covering the extracellular part of TM2, as well as TM4a-c showed the lowest TSO-HDX-KINET values that ranged between 50.6-52.8 °C and 40.6-42.1 °C for EAAT1 T1 and EAAT1 T2, respectively. These values are in excellent quantitative agreement with the TSO-SEC obtained for the loss of the trimeric state, and strongly support the idea that unfolding of TM4 and TM2 are key initial events that determine the trimeric stability and functional state of the transporters. Interestingly, the T50-HDX-KINET values of peptides covering the TranD of ΕΑΑΤ1 τι ranged between 52.8 °C in the cytoplasmic end of TM3, to 56-58 °C in HP1 , TM7, as well as TM8, demonstrating that the regions involved in substrate and ion coordination unfolds at later stages in the thermal denaturation pathway of the transporters. Conclusion
Mutagenesis based on consensus amino acids may allow the engineering of membrane protein with enhanced stability. The two approaches disclosed therein (substituting one or more amino acid residues in the amino acid sequence of the target protein by its(their) respective consensus amino acid residue(s) or substituting one or more amino acid residues in the amino acid sequence of the target protein by substitution, wherein said amino acid residues are selected among residues which are present in pairs of residues that show co-evolution in the target protein) allow the production of two stabilized human homo-trimeric membrane proteins. The consensus approach is especially dedicated for protein families that lack high selective pressure on their stability determinants. The loss of stability observed in a full-consensus sequence of highly thermostable soluble proteins supports the above. Furthermore, non-specialized ancestral proteins are expected to be more stable than the extant specialized structural homologs; the gain of stability has been observed in several laboratories during resurrection of ancestral soluble proteins.
The consensus approach may be adapted to include the condition of co- variance. Conserved amino acid interactions in the SLC1 family are important to the stability of the fold and therefore, restricting the consensus mutagenesis to conserved interacting positions may improve the protein stability. The determination of pairs of position that show high co-variance in a curated PFAM alignment of the SLC1 family, as a proxy for conserved amino acid interactions, and application of the same consensus amino acid exchanges than in the consensus mutagenesis to those positions only permit to reduce the overall number of mutations. Indeed, the second method decreases the number of consensus mutations from 77 to 29, and yield a transporter that shares ~95% sequence identity with wild type EAAT1 . The proteins produced according to this method remains folded and functional in detergent solutions, and even forms low-diffracting 3D crystals, demonstrating a better stability than the wild type protein. Example 5: THERMAL STABILIZATION OF HUMAN Niemann-Pick C1 -like1 PROTEIN
The consensus approach has been applied to another human integral membrane protein family. Niemann-Pick C1 -Like1 (NPCL1 ) is a cholesterol binding protein and is responsible for cholesterol re-adsorption in the apical plasma membrane of intestinal and hepatic epithelia. Notably, some of the current FDA-approved cholesterol lowering drugs, like ezetimibe, inhibit NPC1 L1 transport function making this protein an important drug target. Wild type human NPC1 L1 (NPC1 L1WT) contains 13 transmembrane helices and three large cytoplasmic domains, and although it expresses well in HEK293 cells, it is barely soluble in detergent solutions. The amino acid sequence of N PC1 L1WT includes 1 ,359 residues (SEQ ID No: 12), and it makes impractical and costly the implementation of methods like amino acid scanning mutagenesis to improve its stability. To overcome this problem, the method of the invention has been applied and a mutant construct called NPC1 L1 TI has been designed.
The following steps have been carried out:
a) The amino acid sequences of wild type human NPC1 L1 from selected vertebrate animals used as target sequence that share less than 95% sequence identity with each other have been aligned using JALVIEW software59. Other softwares for Multiple Sequence Alignment may be used such as Clustal W algorithm, or others that can be identified on are disclosed in above paragraph [028]. The parent (or query) sequence was the amino acid sequence of the human protein.
b) At each position in the alignment, a consensus residue was defined when it met all the below criteria: i) the residue had the highest frequency of occurrence among the aligned sequences, and this frequency is >20%; ii) the residue occupied a position in the alignment in which gaps constitute < 30% of the entries; iii) the residue's frequency of occurrence differed by more >10% from the corresponding frequency of the residue in the target/parent sequence (human sequence: NPC1 L1WT);
c) In a first aspect of the process to increase the stability residues in the target protein were substituted for consensus residues in regions where secondary structural elements (e.g. alpha helices) are predicted, and in the short unstructured loops connecting them. The NPC1 L1 homologous transporter NPC1 L1 thermostable T1 (NPC1 L1 TI) has been generated using this approach and has the following sequence:
Sequence ID No.13:
MAEAGLRGWLLWALLLHLAQSEPYTPIHQAGYCAFYDECGKNPELSGSLIPLSNVS CLSNTPARKVTGDHLILLQRICPRLYTGPTTYACCSLKQLVSLELSLSLSKALLTRCP ACAENFANLHCHNTCSPNQSLFINVTRVAQLGAGQLPAWAYEAFYQRSFAEQAYD
SCSRVRIPAAATLAVGTMCGVYGSALCNAQRWLNFQGDTSNGLAPLDITFHLLEPG
QALGSGIQPLNGEVWRCNESQGDGSAACSCQDCAASCPVIAQPPALDSTFRLGQM
PGSLVLIIILCSVFVLLFAFLVGSRVASCRGKDKAKDPKKGTSCSDKLSLSTHTLLGRL
FQSWGTWVASWPLTVLAVSVIVWALAGGLAFIELTTDPVELWSAPNSQARQEKAF
HDQHFGPFFRTNQVILTAPNRPSYRYDSLLLGPKNFSGILSLDLLLELLELQERLRHL
QVWSPEEQRNISLQDICYAPLNPHNASLSDCCVNSLLQYFQNNRTNLLLTANQTLM
GQTSQVDWRDHFLYCANSPLTFKDGTALALSCMADYGAPVFPFLAVGGYKGKDYS
EAEALIMTFSLNNYPAGDPRLAQAKLWEEAFLEEMRAFQRRTAGNFQVTFMAERSL
EDEINRTTAEDLPIFAISYLVIFLYISLALGSYSSWSRVLVDSKATLGLGGVAVVLGAV
LASMGFFSYLGVPSSLVILQVVPFLVLAVGADNIFIFVLEYQRLPRRPGEPREVHIGR
ALGRVAPSMLLCSLSEAICFFLGALTPMPAVRTFALTAGLAVILDFLLQMSAFVALLSL
DSRRQEASRLDVCCCVKAQKLPPPKQGEGLLLRFFRKFYAPFLLHRVTRGVVLLLF
LALFGVSLYFMCHISVGLDQELALPKDSYLLDYFLFLNRYFEVGAPVYFVTTGGYNF
SSEAGMNAICSSAGCDNFSLTQKIQYATEFPEQSYLAIPASSWVDDFIDWLTPSSCC
RLYAFGPNKDEFCPSTVNSLNCLKKCMSITLGPVRPSVEQFHKYLPWFLNDRPNIK
CPKGGLAAYDTSVNLSSDGQILDTVAILSPRLEYSGTISAHCNLYLLDSASRFMAYHK
PLKNSQDYTEALRAARELAANITADLRKVPGTDPAFEVFPYTITNVFYEQYLTIVPEG
LFMLALCLVPTFAVCCLLLGMDLRSGLLNLFSIIMILVDTVGFMALWGISYNAVSLINL
VTAVGISVEFVSHITRSFAISTKPTRLERAKEATISMGSAVFAGVAMTNLPGILVLGLA
KAQLIQIFFFRLNLLITLLGLLHGLVFLPVILSYLGPDVNQALVLEQKRAEEAVAAVME
ASCPNHPSRVSTADNIYVNHSFEHPAKGAGAISSSLPNNGRQF
Alignment between NPC1 L1 TI and NPC1 L1WT is illustrated on Fig. 14. NPC1 L1 TI shares ~ 91 % amino acid sequence identity with NPC1 L1WT (Sequence 12). NPC1 L1 TI expresses at similar levels than NPC1 L1WT in HEK293 cells, but shows a strong improvement in detergent solubility giving rise to a major dominant peak in the size exclusion chromatogram of clear lysates (Fig. 12). These results argue that the consensus approach is a universal method that can be applied to challenging targets with limited phylogenetic information on the protein target.
The consensus-based approaches illustrated by ΕΑΑΤ1 τι , ΕΑΑΤ1 τ2 and NPC1 L1 TI can be generalized to other membrane proteins to obtain stable and functional proteins for structural, as well as biophysical analysis. The methods of the invention allow reduction of the number of constructs to be screened or tested by nearly two orders of magnitude compared to current method like alanine screening and direct evolution, since only a few synthetic genes containing the consensus mutations have to be tested. Furthermore, the methods of the invention allow an easy and eventually automated construct design; based on simple phylogenic analysis of protein sequences and algorithms available online through web-based servers, the calculation and of amino acid consensus sequences is simple.
METHODS
Construct optimization
Fluorescence-detection size-exclusion chromatography (FSEC)51 was used to screen solubilization conditions and EAAT1 variants fused to enhanced green fluorescent protein (eGFP). EAAT1 N-terminal fusions solubilized in dodecanoyl sucrose (DDS, Anatrace) were found to have good solubility and mono-dispersity by FSEC in clear lysates. However, EAAT1 loses its transport activity and chromatographic monodispersity upon purification. To increase its stability, consensus mutagenesis26 was used according to the method of the invention as described in Example 1 , and EAAT1 variants were screened with different consensus mutations in the predicted transmembrane helices by FSEC. The apparent melting temperature (Tm) of the most stable EAAT1 construct was >20 °C over that of the wild-type EAAT1 , but the mutated transporter was still refractory to crystallization. It The extracellular region between TM3-4c was changed for the shorter TM3-4c sequence from ASCT2 (Fig.4). In addition, the two predicted N-glycosylation sites of the transporter (N155T and N204T mutations) were mutated. The obtained mutant showed improved crystallizability.
The transporter obtained this way, called EAAT1 cryst, retained the functional and pharmacological properties of the EAATI WT upon purification and has the following sequence:
Sequence-3
MTKSNGEEPKMGGRMERFQQGVSKRTLLAKKKVQNITKEDVKSFLRRNALLLLTVL A VI LG VVLGFLLRPYPLSPREVKYFAFPGELLMRMLKMLILPLI VSSLITGLA SLDAKA SGRLGMRA VVYYMSTTIIA VVL GIIL VLIIHPGAA SAA I TA S VGA A GSA ENAPSKE VLD SFLDLARNIFPSNL VSAAFRS YS TTYEER Tl TG TR VKVP VGQE VEGMNIL GL VVFSMV FGFALGKMGEQGQLL VDFFNSLNEA TMKL VAIIMWYAPLGILFLIA GKI VEMEDLEVL GGQLGMYMVTVIVGLVIHGLIVLPLIYFLITRKNPFVFIAGILQALITALGTSSSSATLPIT FKCLEENNGVDKRITRFVLPVGA TINMDGTALYEA VAAIFIAQVNNYELDFGQIITISIT A TAA SIGAA GIPQA GL VTMVI VLTA VGLPTDDITLIIA VDWLLDRFRTMVNVLGDALGA
Gl VEHLSRKELEKQDAELGNS VIEENEMKKPYQLIA QDNETEKPIDSETKM Expression and purification
All constructs were introduced into pcDNA3.1 (+) (Invitrogen) with N-terminal Strep-tag II affinity tag followed by eGFP and PreScission protease cleavage site, and expressed in HEK293F cells (ATCC, mycoplasma test negative) grown in Ex-Cell®293 medium (Sigma) and supplemented with 4mM L-glutamine (Sigma) and 5 g/ml Phenol red (Sigma-Aldrich) to densities of 2.5 x 106 cells ml"1. Cells were transiently transfected in Freestyle™293 medium (Invitrogen) using poly-ethylenimine (PEI) (Tebu-bio) at a density of 2.5 x 106 cells ml"1 , diluted with an equivalent volume of Ex- Cell®293 6 hours post-transfection, and treated with 2.2 mM valproic acid (Sigma) 12 hours after dilution of the cultures. Cells were collected at ~48 h post-transfection.
Initial screens of constructs and detergent solubilization buffers were done in small-scale (5-10 ml), and cells were collected, mechanically disrupted with a douncer and solubilized in 50 mM HEPES/Tris-base, pH 7.4, 200 mM NaCI buffer supplemented with 1 mM L-asp, 1 mM EDTA, 1 mM Phenylmethylsulfonyl fluoride (PMSF), 1 mM Tris(2-carboxyethyl)phosphine (TCEP), 1 :200 (v/v) dilution of mammalian protease inhibitor cocktail (Sigma), 10 % glycerol, 2% detergent and 0.4% cholesterol hemisuccinate (CHS) (Anatrace). After 1 -hour incubation at 4 °C, clear lysates were obtained by ultracentrifugation (247,000 g for 45 min). A high-throughput auto-sampler was used to inject the lysates in a SRT SEC-500 column (Sepax Technologies) equilibrated in 50 mM HEPES/Tris-base, pH 7.4, 200 mM NaCI buffer supplemented with 1 mM L-asp, 1 mM (TCEP), 5 % glycerol, 3 x CMC (Critical Micelle Concentration) detergent, and ~0.01 % CHS, in line with fluorescence detection (Photon technology international) for FSEC analysis.
Large-scale expression was done in 2-4 I cultures with cells collected in 50 mM HEPES/Tris-base, pH 7.4, 50 mM NaCI buffer supplemented with 1 mM L-asp, 1 mM EDTA, 1 mM PMSF, 1 mM TCEP, and 1 :200 (v/v) dilution of mammalian protease inhibitor cocktail (Sigma), and disrupted in a cell homogenizer (EmulsiFlex-C5, Avestin) after 3 runs at 15,000 Psi. The resulting homogenate was clarified by centrifugation (4,500 g, 0.5 h) and the crude membranes were collected by ultracentrifugation (186,000 g for 1 .5 h). Membranes were washed once with the above-mentioned buffer and finally homogenized with a douncer in a buffer containing 50 mM HEPES/Tris-base, pH 7.4, 200 mM NaCI, 1 mM L-asp, 1 mM EDTA, 1 mM TCEP, and 10% Glycerol, snap-frozen in liquid N2 and stored at -80°C at 0.5 g of membranes ml"1. Membrane solubilization was done by thawing out and supplementing the membrane homogenate with 2% DDS, 0.4% CHS, and 25 μΜ UCPHioi (Abeam). After 1 -hour incubation, the insoluble material was removed by ultracentrifugation (186,000g for 1 h), and Strep-Tactin® sepharose® resin (GE Healthcare) was added to the supernatant and rotated for 2 h. Resin was washed with 25 column volumes of 50 mM HEPES/Tris-base, pH 7.4, 200 mM NaCI, 1 mM L-asp, 1 mM TCEP, 5% Glycerol, 0.05% DDS, 0.01 % CHS and 25 μΜ UCPHioi , and the protein was eluted with the same buffer supplemented with 2.5 mM D-desthiobiotin.
The eluted eGFP-transporter fusion was concentrated to 1 -2 mg ml"1 using 100- kDa cutoff membranes (Millipore), and digested with His-tagged PreScission protease overnight at 4 °C. The protease was removed by reverse Ni-NTA (Qiagen) affinity chromatography, and the flow through containing the transporter was concentrated to 500 μΙ, ultra-centrifuged (86,900 g, 20 min), and applied to a Superose 6 10/300 gel filtration column (GE Healthcare) equilibrated with 50 mM HEPES/Tris-base, pH 7.4, 200 mM NaCI, 1 mM L-asp, 1 mM TCEP, 5% Glycerol, 0.25% decanoyl sucrose (DS, Sigma), 0.05% CHS and 100 μΜ UCPHioi . To obtain the UCPH101 unbound structure, the protocol was identical, but the allosteric inhibitor was omitted from all buffers. To obtain the UCP HI OI -TBOATFB bound structure, the protein sample was supplemented with 3 mM TBOATFB (Tocris) before the injection in the gel filtration column equilibrated with 50 mM HEPES/Tris-base, pH 7.4, 200 mM NaCI, 1 mM TCEP, 5% Glycerol, 0.25% decanoyl sucrose (DS, Sigma), 0.05% CHS, 300 μΜ TBOATFB and 100 μΜ UCPHioi . Protein samples after the solubilization step were kept on ice or at 4 °C at all times.
Crystallization and structure determination
Purified protein was concentrated to 3.5-4.0 mg ml"1 and 1 mM UCP H 101 was added in experiments with the inhibitor-bound transporters. Initial vapor diffusion crystallization screens were done by mixing 300 nl of protein and reservoir solution in sitting drops, dispensed by a Mosquito® robot (TTP labtech) in 96-well Greiner plates. The purified transporters form three-dimensional crystals in several conditions containing low molecular weight polyethylene glycols. The best-diffracting crystals were obtained after manual optimization using 1.6 μΙ hanging drops at 4 °C, obtained by mixing equal volumes of protein supplemented with 0.2% n-Octyl-b-D- glucopyranoside (BOG, Anatrace) and 0.04% CHS, and reservoir solutions containing 100 mM Tris, pH 8.2, 50 mM CaCI2, 50 mM BaCI2, and 28-30% PEG 400. Crystals appeared after 24-48 h and reached their maximum size after a week. Crystals were flash-frozen in liquid nitrogen before X-ray diffraction data collection without any further cryo protection.
X-ray diffraction data were collected at beamlines PROXIMA-1 at the SOLEIL synchrotron (St Aubin, France) and at beamlines at the European Synchrotron Radiation Facility (Grenoble, France). In general, 2-3 data sets from single crystals were collected, and indexed, integrated, scaled and merged using XDS package52. Due to the anisotropic nature of the diffraction data, the DEBYE and STARANISO programs were applied to scale it using the STARANISO server (http://staraniso.globalphasing.org/). The software performs an anisotropic cut-off of merged intensity data with a Bayesian estimation of the structure amplitudes, and applies an anisotropic correction to the data. Table 1 shows the refinement statistics for the full sets of reflections truncated at the best high-resolution along h, k or I axis, values given by AIMLESS53, before the anisotropic corrections computed by the STARANISO software. The corrected anisotropic amplitudes were then used for molecular replacement in PHASER54, using the scaffold and transport domains of GltPh (PDB code 2NWL) as independent search models. The initial electron density maps were clearly interpretable, and the final model was obtained through rounds of manual building in COOT55 and refinement in Buster56, until reaching good crystallographic statistics and stereochemistry. The model contains one EAAT1 cryst monomer per asymmetric unit and most of the EAAT1 cryst polypeptide (residues 37- 487), with the exception of some residues in the extracellular loops between TM3-4a, TM4b-4c, TM5-6 and TM7b-HP2a. Sequence assignment was aided by anomalous difference Fourier maps from diffraction data collected with low energy X-rays (1.77 A) to highlight the sulfur atoms of methionine and cysteine residues. EAAT1 Cryst-n, as well as the EAATI cryst UCPHIOI -TBOATFB bound structures were solved by the same approach above mentioned, but using the EAAT1 cryst TranD and ScaD as independent search models for molecular replacement.
The stereochemical properties of the final models were analyzed with the Molprobity server (http://molprobity.biochem.duke.edu/). At least 95% of the residues in all models are in the Ramachandran favored region. Protein interfaces were analyzed with the PISA server (http://www.ebi.ac.uk/msd-srv/prot_int/pistart.html). Structural alignments were done with Superpose in the CCP4 suite. All structural figures were prepared with PyMOL Molecular Graphics System, Schrodinger, LLC. Radioactive substrate transport assays
Unilamellar liposomes were made at 9: 1 molar ratio of 1 -palmitoyl-2-oleoyl-sn- glycero-3-phosphocholine (Avanti Polar Lipids) and CHS, in a buffer containing 50 mM HEPES/Tris-base, pH 7.4, 200 mM NaCI and 1 mM L-asp. The transporters were purified as described above, but excluding the reverse chromatography step after protease cleavage, and using a Superose 6 10/300 column equilibrated with 50 mM HEPES/Tris-base, pH 7.4, 200 mM NaCI, 1 mM L-asp, 0.5 mM TCEP, 0.0632% DDS, 0.01264% CHS, and 5% glycerol.
To reconstitute the protein, liposomes were first mixed with DDS at a 1 :2 (w/w) lipid-to-detergent ratio for 1 h, and then the purified transporters were added at a 1 :40 (w/w) protein-to-lipid ratio. Detergent removal was done at 4° C using SM-2 biobeads (BioRad) at 100 mg ml"1. The internal solution of the liposomes was exchanged using 10 freeze-thaw cycles in the appropriate buffer. After extrusion through 400-nm polycarbonate membranes (Avanti Polar Lipids), the proteoliposomes were concentrated by ultracentrifugation (150,000 g for 30 min at 4°C) and resuspended at 20 mg of lipids ml"1 , for immediate use.
Substrate transport was assayed at 37°C. The uptake reaction was initiated by diluting the proteo-liposomes 10-fold into a buffer containing 50 mM HEPES/Tris-base, pH 7.4, 200 mM NaCI, 50 μΜ L-glutamate, and 5 μΜ [14C]-L-glutamate (PerkinElmer), and 2.5% glycerol. After 30 min, 200-μΙ aliquots were diluted 5-fold into ice-cold quench buffer (50 mM HEPES/Tris-base, pH 7.4, 200 mM ChCI, and 2.5% glycerol), followed by immediate filtration and wash on nitrocellulose 0.22-μηΊ filters (Millipore). Radioactivity was quantified by liquid scintillation using a Tri-Carb 31 10TR counter (PerkinElmer). For the UCPH101 titrations, proteo-liposomes were both pre-incubated for 20 min at room temperature, and assayed in the presence of UCPH101. Background radioactivity was estimated from protein-free liposomes, and subtracted from the uptake data. Data was fitted to a Hill equation of the form:
F=F- + deltaF-/(1 +(IC5o/[UCPHioi])n)
Where F~ is the final level of inhibition, deltaF~ is the final amplitude of the UCPH101 effect, and n is the Hill coefficient.
To titrate the rate of L-glutamate transport by EAAT1 cryst, proteo-liposomes were assayed in the presence of 0, 5, 50 or 200 μΜ L-glutamate supplemented with 1 , 5, 5, or 5 μΜ [14C]-L-glutamate, respectively. At each substrate concentration, the initial rate of transport was calculated by a linear fit to 120 s and 180 s uptake measurements with origin fixed at zero. Background radioactivity was estimated from protein-free liposomes, and subtracted from the uptake data.
For the cell-based transport uptake, cells were collected 36 h post-transfection, and washed three times and resuspended at a density of 50 x 106 cells ml"1 in 1 1 mM HEPES/Tris-base, pH 7.4, 140 mM ChCI, 4.7 mM KCI, 2.5 mM CaCI2, 1.2 mM MgCI2, and 10 mM D-glucose, for immediate use. The uptake assay was performed similarly to the one described for the proteo-liposomes, but using a reaction buffer containing 1 1 mM HEPES/Tris-base, pH 7.4, 140 mM NaCI, 4.7 mM KCI, 2.5 mM CaCI2, 1 .2 mM MgC , 10 mM D-glucose, 50 μΜ L-glutamate, and 5 μΜ [14C]-L-glutamate, and 0.8- m nitrocellulose filters. Background radioactivity was estimated from cells transfected with empty vector, and subtracted from the uptake data.
Hydrogen-deuterium exchange mass spectrometry
HDX-MS experiments were performed with transporters purified as described in the proteo-liposome section, and using a Superose 6 5/150 gel filtration column equilibrated with 50 mM HEPES/Tris-base, 200 mM NaCI, pH7.4, 1 mM L-asp, 0.5 mM TCEP, 0.0632% DDS, 0.01264% CHS, and 5% glycerol.
The purified EAAT1 cryst was incubated in ice for 30 min with 2.2% DMSO at a monomer concentration of 5.2 μΜ, in the presence and absence of 102 μΜ UCPHioi , respectively. Prior to labeling, 10 μΙ_ of the unbound and UCPHioi-bound EAAT1 cryst solution was equilibrated for 10 min at room temperature. Deuterium exchange was initiated by adding 40 μΙ_ of D20 buffer (50 mM HEPES, pH 7.3, 200 mM NaCI, 1 mM L-asp, 5% glycerol, 0.0632% DDS, 0.01264% CHS, 0.5 mM TCEP) supplemented or not with 101 .2 μΜ UCPHioi . Assuming that the KD and IC50 values of UCPH101 are similar, ~96% of the transporters would remain bound during deuterium labeling. Aliquots of 10.4 pmols of protein were removed at defined deuterium exchange time points (from 10 sec to 60 min) and quenched upon mixing with an ice-cold acidic solution (0.75% formic acid, 5% glycerol) to decrease the pH to 2.6. Quenched samples were immediately snap-frozen in liquid N2 and stored at -80°C until analysis.
Prior to mass analysis, quenched samples were rapidly thawed and immediately injected into a cooled nanoACQUITY UPLC® HDX system (Waters corp.) maintained at 0°C. 8.6-pmol protein samples were on-line digested for 2 min at 20 °C using an in- house packed immobilized pepsin cartridge (2.0 x 20 mm, 66 μΙ_ bed volume). The resulting peptides were trapped and desalted onto a C18 Trap column (VanGuard BEH 1 .7 μίτι, 2.1 x 5 mm, Waters corp.) at a flow rate of 100 μΙ_Ληίη of 0.15% formic acid, and then separated in 10 min by a linear gradient of acetonitrile from 5 to 40% at 40 ML/min using an ACQUITY UPLC® BEH C18 analytical column (1 .7 Mm, 1 x 100 mm, Waters corp.). After each run, the pepsin cartridge was manually cleaned with two consecutive washes of 1 % formic acid, 5% acetonitrile, 1.5 M guanidinium chloride, pH 2.5. Blank injections were performed between each run to confirm the absence of carry-over.
Mass spectra were acquired in resolution and positive mode on a Synapt G2-Si HDMS mass spectrometer (Waters corp.) equipped with a standard electrospray ionization source, as described previously57. Peptides were identified from undeuterated protein samples acquired in MSE mode by database searching in ProteinLynX Global Server 3.0 (Waters corp.). Each fragmentation spectrum was manually inspected for assignment validation. Deuterium uptake values were calculated for each peptide using DynamX 3.0 (Waters corp.). Only one unique charge state was considered per peptide and no adjustment was made for back-exchange. HDX-MS results are reported as relative deuterium uptake values expressed in mass unit or fractional exchange57. A statistical analysis was performed with MEMHDX58 using a False Discovery Rate of 1 %. References
Slotboom, D. J., Konings, W. N. & Lolkema, J. S. Structural features of the glutamate transporter family. Microbiol Mol Biol Rev 63, 293-307 (1999).
Kanai, Y. & Hediger, M. A. The glutamate and neutral amino acid transporter family: physiological and pharmacological implications. Eur J Pharmacol 479, 237-247 (2003).
Danbolt, N. C. Glutamate uptake. Prog Neurobiol 65, 1 -105 (2001 ).
Lehre, K. P. & Danbolt, N. C. The number of glutamate transporter subtype molecules at glutamatergic synapses: chemical and stereological quantification in young adult rat brain. J Neurosci 18, 8751 -8757 (1998).
Zerangue, N. & Kavanaugh, M. P. Flux coupling in a neuronal glutamate transporter. Nature 383, 634-637, doi: 10.1038/383634a0 (1996). Rothstein, J. D., Van Kammen, M., Levey, A. I., Martin, L. J. & Kuncl, R. W. Selective loss of glial glutamate transporter GLT-1 in amyotrophic lateral sclerosis. Ann Neurol 38, 73-84, doi: 10.1002/ana.4103801 14 (1995).
Winter, N., Kovermann, P. & Fahlke, C. A point mutation associated with episodic ataxia 6 increases glutamate transporter anion currents. Brain 135, 3416-3425, doi: 10.1093/brain/aws255 (2012).
Choi, K. D. et al. Late-onset episodic ataxia associated with SLC1A3 mutation. J Hum Genet, doi: 10.1038/jhg.2016.137 (2016).
Chao, X. D., Fei, F. & Fei, Z. The role of excitatory amino acid transporters in cerebral ischemia. Neurochem Res 35, 1224-1230, doi: 10.1007/s1 1064-010- 0178-3 (2010).
Pile, A., Wieronska, J. M. & Skolnick, P. Glutamate-based antidepressants: preclinical psychopharmacology. Biol Psychiatry 73, 1 125-1 132, doi: 10.1016/j.biopsych.2013.01 .021 (2013).
Robert, S. M. & Sontheimer, H. Glutamate transporters in the biology of malignant gliomas. Cell Mol Life Sci 71 , 1839-1854, doi: 10.1007/s00018-013- 1521 -z (2014).
Bailey, C. G. et al. Loss-of-function mutations in the glutamate transporter SLC1A1 cause human dicarboxylic aminoaciduria. J Clin Invest 121 , 446-453, doi: 10.1 172/JCI44474 (201 1 ).
Zerangue, N. & Kavanaugh, M. P. ASCT-1 is a neutral amino acid exchanger with chloride channel activity. J Biol Chem 271 , 27991 -27994 (1996).
Wang, Q. et al. Targeting glutamine transport to suppress melanoma cell growth. Int J Cancer 135, 1060-1071 , doi: 10.1002/ijc.28749 (2014).
Shimizu, K. et al. ASC amino-acid transporter 2 (ASCT2) as a novel prognostic marker in non-small cell lung cancer. Br J Cancer 110, 2030-2039, doi: 10.1038/bjc.2014.88 (2014).
Wang, Q. et al. Targeting ASCT2-mediated glutamine uptake blocks prostate cancer growth and tumour development. J Pathol 236, 278-289, doi: 10.1002/path.4518 (2015).
van Geldermalsen, M. et al. ASCT2/SLC1A5 controls glutamine uptake and tumour growth in triple-negative basal-like breast cancer. Oncogene 35, 3201 - 3208, doi: 10.1038/onc.2015.381 (2016). Shimamoto, K. Glutamate transporter blockers for elucidation of the function of excitatory neurotransmission systems. Chem Rec 8, 182-199, doi: 10.1002/tcr.20145 (2008).
Grewer, C. & Grabsch, E. New inhibitors for the neutral amino acid transporter ASCT2 reveal its Na+-dependent anion leak. J Physiol 557, 747-759, doi: 10.1 1 13/jphysiol.2004.062521 (2004).
Jensen, A. A. et al. Discovery of the first selective inhibitor of excitatory amino acid transporter subtype 1 . J Med Chem 52, 912-915, doi: 10.1021 /jm8013458 (2009).
Abrahamsen, B. et al. Allosteric modulation of an excitatory amino acid transporter: the subtype-selective inhibitor UCPH-101 exerts sustained inhibition of EAAT1 through an intramonomeric site in the trimerization domain. J Neurosci 33, 1068-1087, doi: 10.1523/JNEUROSCI.3396-12.2013 (2013). Yernool, D., Boudker, O., Jin, Y. & Gouaux, E. Structure of a glutamate transporter homologue from Pyrococcus horikoshii. Nature 431 , 81 1 -818, doi: 10.1038/nature03018 (2004).
Reyes, N., Ginter, C. & Boudker, O. Transport mechanism of a bacterial homologue of glutamate transporters. Nature 462, 880-885, doi: 10.1038/nature08616 (2009).
Akyuz, N. et al. Transport domain unlocking sets the uptake rate of an aspartate transporter. Nature 518, 68-73, doi: 10.1038/nature14158 (2015).
Boudker, O., Ryan, R. M. , Yernool, D. , Shimamoto, K. & Gouaux, E. Coupling substrate and ion binding to extracellular gate of a sodium-dependent aspartate transporter. Nature 445, 387-393, doi:10.1038/nature05455 (2007).
Steipe, B., Schiller, B., Pluckthun, A. & Steinbacher, S. Sequence statistics reliably predict stabilizing mutations in a protein domain. J Mol Biol 240, 188-
192, doi: 10.1006/jmbi.1994.1434 (1994).
Zhang, Y., Bendahan, A., Zarbiv, R., Kavanaugh, M. P. & Kanner, B. I. Molecular determinant of ion selectivity of a (Na+ + K+)-coupled rat brain glutamate transporter. Proc Natl Acad Sci U S A 95, 751 -755 (1998).
Seal, R. P. & Amara, S. G. A reentrant loop domain in the glutamate carrier EAAT1 participates in substrate binding and translocation. Neuron 21 , 1487- 1498 (1998). Tao, Z. et al. Mechanism of cation binding to the glutamate transporter EAAC1 probed with mutation of the conserved amino acid residue Thr101 . J Biol Chem
285, 17725-17733, doi: 10.1074/jbc.M1 10.121798 (2010).
Larsson, H. P. et al. Evidence for a third sodium-binding site in glutamate transporters suggests an ion/substrate coupling model. Proc Natl Acad Sci U S
A 107, 13912-13917, doi: 10.1073/pnas.1006289107 (2010).
Guskov, A., Jensen, S. , Faustino, I. , Marrink, S. J. & Slotboom, D. J. Coupled binding mechanism of three sodium ions and aspartate in the glutamate transporter homologue GltTk. Nat Commun 7, 13420, doi: 10.1038/ncomms13420 (2016).
Crisman, T. J., Qu, S., Kanner, B. I. & Forrest, L. R. Inward-facing conformation of glutamate transporters as revealed by their inverted-topology structural repeats. Proc Natl Acad Sci U S A 106, 20752-20757, doi: 10.1073/pnas.0908570106 (2009).
Focke, P. J., Moenne-Loccoz, P. & Larsson, H. P. Opposite movement of the external gate of a glutamate transporter homolog upon binding cotransported sodium compared with substrate. J Neurosci 31 , 6255-6262, doi: 10.1523/JNEUROSCI.6096-10.201 1 (201 1 ).
Brocke, L, Bendahan, A., Grunewald, M. & Kanner, B. I. Proximity of two oppositely oriented reentrant loops in the glutamate transporter GLT-1 identified by paired cysteine mutagenesis. J Biol Chem 277, 3985-3992, doi: 10.1074/jbc.M107735200 (2002).
Qu, S. & Kanner, B. I. Substrates and non-transportable analogues induce structural rearrangements at the extracellular entrance of the glial glutamate transporter GLT-1 /EAAT2. J Biol Chem 283, 26391 -26400, doi: 10.1074/j be. M 802401200 (2008).
Seal, R. P., Leighton, B. H. & Amara, S. G. A model for the topology of excitatory amino acid transporters determined by the extracellular accessibility of substituted cysteines. Neuron 25, 695-706 (2000).
Grunewald, M., Bendahan, A. & Kanner, B. I. Biotinylation of single cysteine mutants of the glutamate transporter GLT-1 from rat brain reveals its unusual topology. Neuron 21 , 623-632 (1998).
Ryan, R. M., Kortt, N. C, Sirivanta, T. & Vandenberg, R. J. The position of an arginine residue influences substrate affinity and K+ coupling in the human glutamate transporter, EAAT1 . J Neurochem 114, 565-575, doi: 10.1 1 1 1 /j .1 1 - 4159.2010.06796.x (2010).
Borre, L. & Kanner, B. I. Arginine 445 controls the coupling between glutamate and cations in the neuronal transporter EAAC-1 . J Biol Chem 279, 2513-2519, doi: 10.1074/jbc.M31 1446200 (2004).
Leinenweber, A., Machtens, J. P., Begemann, B. & Fahlke, C. Regulation of glial glutamate transporters by C-terminal domains. J Biol Chem 286, 1927- 1937, doi: 10.1074/jbc.M1 10.153486 (201 1 ).
Shouffani, A. & Kanner, B. I. Cholesterol is required for the reconstruction of the sodium- and chloride-coupled, gamma-aminobutyric acid transporter from rat brain. J Biol Chem 265, 6002-6008 (1990).
Butchbach, M. E., Tian, G., Guo, H. & Lin, C. L. Association of excitatory amino acid transporters, especially EAAT2, with cholesterol-rich lipid raft microdomains: importance for excitatory amino acid transporter localization and function. J Biol Chem 279, 34388-34396, doi: 10.1074/jbc.M403938200 (2004). Mcllwain, B. C, Vandenberg, R. J. & Ryan, R. M. Transport rates of a glutamate transporter homologue are influenced by the lipid bilayer. J Biol Chem 290, 9780-9788, doi: 10.1074/jbc.M1 14.630590 (2015).
Fairman, W. A., Sonders, M. S., Murdoch, G. H. & Amara, S. G. Arachidonic acid elicits a substrate-gated proton current associated with the glutamate transporter EAAT4. Nat Neurosci 1 , 105-1 13, doi: 10.1038/355 (1998).
Raunser, S. et al. Heterologously expressed GLT-1 associates in approximately 200-nm protein-lipid islands. Biophys J 91 , 3718-3726, doi: 10.1529/biophysj.106.086900 (2006).
Wales, T. E. & Engen, J. R. Hydrogen exchange mass spectrometry for the analysis of protein dynamics. Mass Spectrom Rev 25, 158-170, doi: 10.1002/mas.20064 (2006).
Konermann, L, Pan, J. & Liu, Y. H. Hydrogen exchange mass spectrometry for studying protein structure and dynamics. Chem Soc Rev 40, 1224-1234, doi: 10.1039/c0cs001 13a (201 1 ).
Shimamoto, K. et al. Characterization of novel L-threo-beta-benzyloxyaspartate derivatives, potent blockers of the glutamate transporters. Mol Pharmacol 65, 1008-1015, doi: 10.1 124/mol.65.4.1008 (2004). Watzke, N. & Grewer, C. The anion conductance of the glutamate transporter EAAC1 depends on the direction of glutamate transport. FEBS Lett 503, 121 - 125 (2001 ).
Reyes, N., Oh, S. & Boudker, O. Binding thermodynamics of a glutamate transporter homolog. Nat Struct Mol Biol 20, 634-640, doi: 10.1038/nsmb.2548 (2013).
Kawate, T. & Gouaux, E. Fluorescence-detection size-exclusion chromatography for precrystallization screening of integral membrane proteins. Structure 14, 673-681 , doi: 10.1016/j.str.2006.01 .013 (2006).
Kabsch, W. Xds. Acta Crystallogr D Biol Crystallogr 66, 125-132, doi: 10.1 107/S0907444909047337 (2010).
Evans, P. R. & Murshudov, G. N. How good are my data and what is the resolution? Acta Crystallogr D Biol Crystallogr 69, 1204-1214, doi: 10.1 107/S0907444913000061 (2013).
McCoy, A. J. et al. Phaser crystallographic software. J Appl Crystallogr 40, 658- 674, doi: 10.1 107/S0021889807021206 (2007).
Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr D Biol Crystallogr 66, 486-501 , doi: 10.1 107/S0907444910007493 (2010).
Blanc, E. et al. Refinement of severely incomplete structures with maximum likelihood in BUSTER-TNT. Acta Crystallogr D Biol Crystallogr 60, 2210-2221 , doi: 10.1 107/S0907444904016427 (2004).
O'Brien, D. P. et al. Structural models of intrinsically disordered and calcium- bound folded states of a protein adapted for secretion. Sci Rep 5, 14223, doi: 10.1038/srep14223 (2015).
Hourdel, V. et al. MEMHDX: An interactive tool to expedite the statistical validation and visualization of large HDX-MS datasets. Bioinformatics, doi: 10.1093/bioinformatics/btw420 (2016).
Waterhouse, A. M., Procter, J. B., Martin, D. M., Clamp, M. & Barton, G. J. Jalview Version 2-a multiple sequence alignment editor and analysis workbench. Bioinformatics 25, 1 189-1 191 , doi: 10.1093/bioinformatics/btp033 (2009). Wallin, E. & von Heijne, G. Genome-wide analysis of integral membrane proteins from eubacterial, archaean, and eukaryotic organisms. Protein Sci 7, 1029-1038, doi:10.1002/pro.5560070420 (1998).
Li, M. et al. Selecting optimum eukaryotic integral membrane proteins for structure determination by rapid expression and solubilization screening. J Mol Biol 385, 820-830, doi: 10.1016/j.jmb.2008.1 1 .021 (2009).
Lin, L, Yee, S. W., Kim, R. B. & Giacomini, K. M. SLC transporters as therapeutic targets: emerging opportunities. Nat Rev Drug Discov 14, 543-560, doi: 10.1038/nrd4626 (2015).
Tate, C. G. & Schertler, G. F. Engineering G protein-coupled receptors to facilitate their structure determination. Curr Opin Struct Biol 19, 386-395, doi: 10.1016/j.sbi.2009.07.004 (2009).
Bill, R. M. et al. Overcoming barriers to membrane protein structure determination. Nat Biotechnol 29, 335-340, doi: 10.1038/nbt.1833 (201 1 ).
Schlinkmann, K. M. et al. Maximizing detergent stability and functional expression of a GPCR by exhaustive recombination and evolution. J Mol Biol 422, 414-428, doi: 10.1016/j.jmb.2012.05.039 (2012).
Scott, D. J., Kummer, L., Tremmel, D. & Pluckthun, A. Stabilizing membrane proteins through protein engineering. Curr Opin Chem Biol 17, 427-435, doi: 10.1016/j.cbpa.2013.04.002 (2013).
Hopf, T. A. et al. Three-dimensional structures of membrane proteins from genomic sequencing. Cell 149, 1607-1621 , doi: 10.1016/j. cell.2012.04.012 (2012).
Minichino A et al. The properties of (2Fo-Fc) and (Fo-Fc) electron density maps at medium-to-high resolutions Acta Crys D59, 843-849 (2003)

Claims

Claims
A process to produce a protein with modified thermal stability in a detergent solution with respect to the thermal stability of a target protein comprising: a. providing a multiple sequence alignment (MSA) wherein at least a portion comprising at least 50 % of the full-length amino acid sequence of a target protein is aligned, in particular the full-length amino acid sequence of the target protein is aligned, with the amino acid sequence of homologous sequences of said protein present (i) in other species and/or (ii) in protein variants of the same species, using a determined alignment tool; wherein the homologous sequences are each different from the amino acid sequence of the target protein by at least one amino acid residue in their amino acid sequence, in particular wherein each homologous sequence shares at least 50% identity, more preferably at least 70% identity, with the amino acid sequence of the target protein or with the aligned portion thereof;
b. defining for at least part of the positions of the amino acid residues in the target protein, in particular for each amino acid position of the aligned amino acid sequences, a consensus amino acid residue wherein the consensus amino acid residue has the following characteristics:
i. a frequency of occurrence among the aligned amino acid sequences which is higher than 20%,
ii. a position in the alignment of the amino acid sequences wherein gaps constitute less than 30% of the entries,
iii. a frequency of occurrence which is higher than 10% with respect to the frequency of occurrence of the residue at the same position in the target amino acid sequence when said frequency is evaluated among the aligned amino acid sequences;
c. providing a mutated amino acid sequence defining a mutant protein of the target sequence by substituting one or more amino acid residues in the amino acid sequence of the target protein by its(their) respective consensus amino acid residue(s) defined for said each amino acid residue according to step b.; d. obtaining a polynucleotide encoding the mutant protein of step c. for expression in a cellular expression system;
e. producing the mutant protein from the mutated amino acid sequence provided in step c in a cellular expression system;
f. evaluating thermal stability in a detergent solution of the mutant protein recovered from the production cells in step e.
A process to design a protein with modified thermal stability in a detergent solution with respect to the thermal stability of a target protein comprising: a. providing a multiple sequence alignment (MSA) wherein at least a portion comprising at least 50 % of the full-length amino acid sequence of a target protein is aligned, in particular the full-length amino acid sequence of the target protein is aligned, with the amino acid sequence of homologous sequences of said protein present (i) in other species and/or (ii) in protein variants of the same species, using a determined alignment tool; wherein the homologous sequences are each different from the amino acid sequence of the target protein by at least one amino acid residue in their amino acid sequence, in particular wherein each homologous sequence shares at least 50% identity, more preferably at least 70% identity, with the amino acid sequence of the target protein or with the aligned portion thereof;
b. defining for at least part of the positions of the amino acid residues in the target protein, in particular for each amino acid position of the aligned amino acid sequences, a consensus amino acid residue wherein the consensus amino acid residue has the following characteristics:
i. a frequency of occurrence among the aligned amino acid sequences which is higher than 20%,
ii. a position in the alignment of the amino acid sequences wherein gaps constitute less than 30% of the entries,
iii. a frequency of occurrence which is higher than 10% with respect to the frequency of occurrence of the residue at the same position in the target amino acid sequence when said frequency is evaluated among the aligned amino acid sequences; c. providing a mutated amino acid sequence defining a consensus mutant protein of the target sequence by substituting one or more amino acid residues in the amino acid sequence of the target protein by its(their) respective consensus amino acid residue(s) defined for said each amino acid residue according to step b.
3. A process according to claim 1 or 2, wherein the amino acid residues targeted for substitution are selected among residues located in predicted or characterized secondary structural element(s) of the target protein and/or in loops structures.
4. A process according to any one of claims 1 to 3, wherein the amino acid residues targeted for substitution are selected among residues which are present in pairs of residues that show co-evolution in the target protein.
5. A process according to any one of claims 1 to 4, wherein the amino acid residues targeted for substitution are contained in a characterized or a predicted alpha-helice structure of the target protein.
6. A process according to any one of claims 1 to 5, wherein the protein is selected from integral membrane proteins, membrane protein complexes, water-soluble proteins.
7. A process according to any one of claims 1 to 6, wherein the target protein is selected in the group of membrane proteins, integral membrane proteins, soluble proteins, globular soluble proteins.
8. A process according to any one of claims 1 to 7, wherein the target protein is selected in the group of membrane proteins, integral membrane proteins, soluble proteins, globular soluble proteins, wherein said protein originates from a pathogenic bacterium, virus or parasite.
9. A process according to any one of claims 1 to 8, wherein the target protein is selected in the group of membrane proteins and is a eukaryotic membrane protein, in particular a mammalian membrane protein, especially a human membrane protein, and in particular a human integral membrane protein.
10. A process according to any one of claims 1 to 9, wherein the membrane protein is selected among channels, enzymes, and primary or secondary active transporters.
11. A process according to any one of claims 1 to 10, wherein the thermal stability in detergent solution of the mutant protein is evaluated by size-exclusion chromatography of detergent-solubilized protein expressed in step e. and optionally compared to the thermal stability of the target protein in the same conditions.
12. A process according to any one of claims 1 to 1 1 , wherein the mutant protein is fused to a detectable marker, such as a fluorescent marker, for the evaluation of the melting temperature of the mutant protein.
13. A process to improve thermal stability of a target protein in detergent solution comprising carrying out the process of any one of claims 1 to 12 and selecting the mutant protein(s) having improved thermal stability with respect to the target protein.
14. A process according to any one of claims 1 to 13, wherein the mutant protein has a sequence identity with the target protein which is at least 80%, more preferably at least 85%, in particular the mutant protein has a sequence similarity with SEQ ID No: 1 1 or SEQ ID No: 12 which is at least 85%.
15. A process according to any one of claims 1 to 13, wherein the number of homologous sequences used in the MSA is at least 7, and more preferably at least 10.
16. A process for the preparation of a protein with modified crystallization capability comprising preparing a protein having modified thermal stability in a detergent solution according to any one of claims 1 to 15, wherein an optional step of mutation is performed after step c. and in particular before step d. to further improve crystallization capability, such optional step of mutation being in particular a step of mutation in the extracellular region of the target protein.
17. A process according to any one of claims 1 to 16, wherein the target protein is the Excitatory Amino Acid Transporter 1 (EAAT1 ) and the obtained mutant protein is EAAT1 of sequence SEQ ID No.1 SEQ ID No.2, SEQ ID No.3 or SEQ ID No.4.
18. A process according to any one of claims 1 to 16, wherein the target protein is the Niemann-Pick C1 -Likel (NPCL1 ) and the obtained mutant is NPCL1 of SEQ ID No.13.
19. A process according to any one of claims 1 to 18, wherein the determined alignment tool is JALVIEW software.
20. A mutant protein of Excitatory Amino Acid Transporter 1 (EAAT1 ) the amino acid sequence of which is SEQ ID No.1 SEQ ID No.2, SEQ ID No.3 or SEQ ID No.4.
21. A mutant protein of Niemann-Pick C1 -Likel (NPCL1 ) the amino acid sequence of which is SEQ ID No. 13.
22. A liposome or a collection of liposomes containing a mutant protein obtained from the process according to any one of claims 1 to 19.
23. A cell expressing a mutant protein obtained from the process according to any one of claims 1 to 19.
24. A cell or a liposome comprising a mutant protein of Excitatory Amino Acid Transporter 1 (EAAT1 ) the amino acid sequence of which is SEQ ID No.1 SEQ ID No.2, SEQ ID No.3 or SEQ ID No.4.
25. A cell or a liposome comprising a mutant protein of Niemann-Pick C1-like1 (NPCL1 ) the amino acid sequence of which is SEQ ID No. 13.
PCT/IB2018/000194 2017-02-16 2018-02-15 Method for the identification of mutated proteins having modified thermal stability WO2018150260A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762459721P 2017-02-16 2017-02-16
US62/459,721 2017-02-16

Publications (1)

Publication Number Publication Date
WO2018150260A1 true WO2018150260A1 (en) 2018-08-23

Family

ID=61617049

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2018/000194 WO2018150260A1 (en) 2017-02-16 2018-02-15 Method for the identification of mutated proteins having modified thermal stability

Country Status (1)

Country Link
WO (1) WO2018150260A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113537409A (en) * 2021-09-13 2021-10-22 烟台双塔食品股份有限公司 Pea protein data feature extraction method

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001047956A2 (en) * 1999-12-23 2001-07-05 Genencor International, Inc. Method for obtaining proteins having improved functional characteristics
WO2009071914A2 (en) * 2007-12-08 2009-06-11 Heptares Therapeutics Limited Mutant proteins and methods for producing them

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001047956A2 (en) * 1999-12-23 2001-07-05 Genencor International, Inc. Method for obtaining proteins having improved functional characteristics
WO2009071914A2 (en) * 2007-12-08 2009-06-11 Heptares Therapeutics Limited Mutant proteins and methods for producing them

Non-Patent Citations (74)

* Cited by examiner, † Cited by third party
Title
ABRAHAMSEN, B. ET AL.: "Allosteric modulation of an excitatory amino acid transporter: the subtype-selective inhibitor UCPH-101 exerts sustained inhibition of EAAT1 through an intramonomeric site in the trimerization domain", J NEUROSCI, vol. 33, 2013, pages 1068 - 1087
AKYUZ, N. ET AL.: "Transport domain unlocking sets the uptake rate of an aspartate transporter", NATURE, vol. 518, 2015, pages 68 - 73
BAILEY, C. G. ET AL.: "Loss-of-function mutations in the glutamate transporter SLC1A1 cause human dicarboxylic aminoaciduria", J CLIN INVEST, vol. 121, 2011, pages 446 - 453
BILL, R. M. ET AL.: "Overcoming barriers to membrane protein structure determination", NAT BIOTECHNOL, vol. 29, 2011, pages 335 - 340, XP055049258, DOI: doi:10.1038/nbt.1833
BLANC, E. ET AL.: "Refinement of severely incomplete structures with maximum likelihood in BUSTER-TNT", ACTA CRYSTALLOGR D BIOL CRYSTALLOGR, vol. 60, 2004, pages 2210 - 2221
BORRE, L.; KANNER, B. I.: "Arginine 445 controls the coupling between glutamate and cations in the neuronal transporter EAAC-1", J BIOL CHEM, vol. 279, 2004, pages 2513 - 2519
BOUDKER, O.; RYAN, R. M.; YERNOOL, D.; SHIMAMOTO, K.; GOUAUX, E.: "Coupling substrate and ion binding to extracellular gate of a sodium-dependent aspartate transporter", NATURE, vol. 445, 2007, pages 387 - 393
BROCKE, L.; BENDAHAN, A.; GRUNEWALD, M.; KANNER, B. I.: "Proximity of two oppositely oriented reentrant loops in the glutamate transporter GLT-1 identified by paired cysteine mutagenesis", J BIOL CHEM, vol. 277, 2002, pages 3985 - 3992
BUTCHBACH, M. E.; TIAN, G.; GUO, H.; LIN, C. L.: "Association of excitatory amino acid transporters, especially EAAT2, with cholesterol-rich lipid raft microdomains: importance for excitatory amino acid transporter localization and function", J BIOL CHEM, vol. 279, 2004, pages 34388 - 34396
CHAO, X. D.; FEI, F.; FEI, Z.: "The role of excitatory amino acid transporters in cerebral ischemia", NEUROCHEM RES, vol. 35, 2010, pages 1224 - 1230, XP019827370
CHOI, K. D. ET AL.: "Late-onset episodic ataxia associated with SLC1A3 mutation", J HUM GENET, 2016
CRISMAN, T. J.; QU, S., KANNER; B. I. & FORREST: "L. R. Inward-facing conformation of glutamate transporters as revealed by their inverted-topology structural repeats", PROC NATL ACAD SCI U S A, vol. 106, 2009, pages 20752 - 20757
DANBOLT, N. C.: "Glutamate uptake", PROG NEUROBIOL, vol. 65, 2001, pages 1 - 105
DANIEL J SCOTT ET AL: "Stabilizing membrane proteins through protein engineering", CURRENT OPINION IN CHEMICAL BIOLOGY, vol. 17, no. 3, 1 June 2013 (2013-06-01), pages 427 - 435, XP055105083, ISSN: 1367-5931, DOI: 10.1016/j.cbpa.2013.04.002 *
EMSLEY, P.; LOHKAMP, B.; SCOTT, W. G.; COWTAN, K.: "Features and development of Coot", ACTA CRYSTALLOGR D BIOL CRYSTALLOGR, vol. 66, 2010, pages 486 - 501
ERICKSON ROBERT P: "Current controversies in Niemann-Pick C1 disease: steroids or gangliosides; neurons or neurons and glia", JOURNAL OF APPLIED GENETICS: AN INTERNATIONAL JOURNAL OF GENETICS AND BREEDING, SPRINGER, GERMANY, vol. 54, no. 2, 5 January 2013 (2013-01-05), pages 215 - 224, XP035367200, ISSN: 1234-1983, [retrieved on 20130105], DOI: 10.1007/S13353-012-0130-0 *
EVANS, P. R.; MURSHUDOV, G. N.: "How good are my data and what is the resolution?", ACTA CRYSTALLOGR D BIOL CRYSTALLOGR, vol. 69, 2013, pages 1204 - 1214
FAIRMAN, W. A.; SONDERS, M. S.; MURDOCH, G. H.; AMARA, S. G.: "Arachidonic acid elicits a substrate-gated proton current associated with the glutamate transporter EAAT4", NAT NEUROSCI, vol. 1, 1998, pages 105 - 113
FOCKE, P. J.; MOENNE-LOCCOZ, P.; LARSSON, H. P.: "Opposite movement of the external gate of a glutamate transporter homolog upon binding cotransported sodium compared with substrate", J NEUROSCI, vol. 31, 2011, pages 6255 - 6262
GREWER, C.; GRABSCH, E.: "New inhibitors for the neutral amino acid transporter ASCT2 reveal its Na+-dependent anion leak", J PHYSIOL, vol. 557, 2004, pages 747 - 759
GRUNEWALD, M.; BENDAHAN, A.; KANNER, B. I.: "Biotinylation of single cysteine mutants of the glutamate transporter GLT-1 from rat brain reveals its unusual topology", NEURON, vol. 21, 1998, pages 623 - 632
GUSKOV, A.; JENSEN, S.; FAUSTINO, I.; MARRINK, S. J.; SLOTBOOM, D. J.: "Coupled binding mechanism of three sodium ions and aspartate in the glutamate transporter homologue GltTk", NAT COMMUN, vol. 7, 2016, pages 13420
HOPF, T. A. ET AL.: "Three-dimensional structures of membrane proteins from genomic sequencing", CELL, vol. 149, 2012, pages 1607 - 1621, XP028497259, DOI: doi:10.1016/j.cell.2012.04.012
HOURDEL, V. ET AL.: "MEMHDX: An interactive tool to expedite the statistical validation and visualization of large HDX-MS datasets", BIOINFORMATICS, 2016
JENSEN, A. A. ET AL.: "Discovery of the first selective inhibitor of excitatory amino acid transporter subtype 1", J MED CHEM, vol. 52, 2009, pages 912 - 915, XP002572172, DOI: doi:10.1021/jm8013458
KABSCH, W. XDS, ACTA CRYSTALLOGR D BIOL CRYSTALLOGR, vol. 66, 2010, pages 125 - 132
KANAI, Y.; HEDIGER, M. A.: "The glutamate and neutral amino acid transporter family: physiological and pharmacological implications", EUR J PHARMACOL, vol. 479, 2003, pages 237 - 247
KAWATE, T.; GOUAUX, E.: "Fluorescence-detection size-exclusion chromatography for precrystallization screening of integral membrane proteins", STRUCTURE, vol. 14, 2006, pages 673 - 681, XP025133935, DOI: doi:10.1016/j.str.2006.01.013
KONERMANN, L.; PAN, J.; LIU, Y. H.: "Hydrogen exchange mass spectrometry for studying protein structure and dynamics", CHEM SOC REV, vol. 40, 2011, pages 1224 - 1234
LARSSON, H. P. ET AL.: "Evidence for a third sodium-binding site in glutamate transporters suggests an ion/substrate coupling model", PROC NATL ACAD SCI U S A, vol. 107, 2010, pages 13912 - 13917
LEHRE, K. P.; DANBOLT, N. C.: "The number of glutamate transporter subtype molecules at glutamatergic synapses: chemical and stereological quantification in young adult rat brain", J NEUROSCI, vol. 18, 1998, pages 8751 - 8757
LEINENWEBER, A.; MACHTENS, J. P.; BEGEMANN, B.; FAHLKE, C.: "Regulation of glial glutamate transporters by C-terminal domains", J BIOL CHEM, vol. 286, 2011, pages 1927 - 1937
LI, M. ET AL.: "Selecting optimum eukaryotic integral membrane proteins for structure determination by rapid expression and solubilization screening", J MOL BIOL, vol. 385, 2009, pages 820 - 830, XP025940913, DOI: doi:10.1016/j.jmb.2008.11.021
LIN, L.; YEE, S. W.; KIM, R. B.; GIACOMINI, K. M.: "SLC transporters as therapeutic targets: emerging opportunities", NAT REV DRUG DISCOV, vol. 14, 2015, pages 543 - 560
MCCOY, A. J. ET AL.: "Phaser crystallographic software", J APPL CRYSTALLOGR, vol. 40, 2007, pages 658 - 674
MCILWAIN, B. C.; VANDENBERG, R. J.; RYAN, R. M.: "Transport rates of a glutamate transporter homologue are influenced by the lipid bilayer", J BIOL CHEM, vol. 290, 2015, pages 9780 - 9788
MICHAEL ANBAR ET AL: "ABSTRACT", APPLIED AND ENVIRONMENTAL MICROBIOLOGY, vol. 78, no. 9, 1 May 2012 (2012-05-01), US, pages 3458 - 3464, XP055466446, ISSN: 0099-2240, DOI: 10.1128/AEM.07985-11 *
MINICHINO A ET AL.: "The properties of (2Fo-Fc) and (Fo-Fc) electron density maps at medium-to-high resolutions", ACTA CRYS, vol. D59, 2003, pages 843 - 849
O'BRIEN, D. P. ET AL.: "Structural models of intrinsically disordered and calcium-bound folded states of a protein adapted for secretion", SCI REP, vol. 5, 2015, pages 14223
PILC, A.; WIERONSKA, J. M.; SKOLNICK, P.: "Glutamate-based antidepressants: preclinical psychopharmacology", BIOL PSYCHIATRY, vol. 73, 2013, pages 1125 - 1132, XP028557116, DOI: doi:10.1016/j.biopsych.2013.01.021
QU, S.; KANNER, B. I.: "Substrates and non-transportable analogues induce structural rearrangements at the extracellular entrance of the glial glutamate transporter GLT-1/EAAT2", J BIOL CHEM, vol. 283, 2008, pages 26391 - 26400
RAUNSER, S. ET AL.: "Heterologously expressed GLT-1 associates in approximately 200-nm protein-lipid islands", BIOPHYS J, vol. 91, 2006, pages 3718 - 3726
REYES, N.; GINTER, C.; BOUDKER, O.: "Transport mechanism of a bacterial homologue of glutamate transporters", NATURE, vol. 462, 2009, pages 880 - 885
REYES, N.; OH, S.; BOUDKER, O.: "Binding thermodynamics of a glutamate transporter homolog", NAT STRUCT MOL BIOL, vol. 20, 2013, pages 634 - 640
ROBERT, S. M.; SONTHEIMER, H.: "Glutamate transporters in the biology of malignant gliomas", CELL MOL LIFE SCI, vol. 71, 2014, pages 1839 - 1854, XP035317754, DOI: doi:10.1007/s00018-013-1521-z
ROTHSTEIN, J. D.; VAN KAMMEN, M.; LEVEY, A. I.; MARTIN, L. J.; KUNCL, R. W.: "Selective loss of glial glutamate transporter GLT-1 in amyotrophic lateral sclerosis", ANN NEUROL, vol. 38, 1995, pages 73 - 84
RYAN, R. M.; KORTT, N. C.; SIRIVANTA, T.; VANDENBERG, R. J.: "The position of an arginine residue influences substrate affinity and K+ coupling in the human glutamate transporter, EAAT1", J NEUROCHEM, vol. 114, 2010, pages 565 - 575
SCHLINKMANN, K. M. ET AL.: "Maximizing detergent stability and functional expression of a GPCR by exhaustive recombination and evolution", J MOL BIOL, vol. 422, 2012, pages 414 - 428, XP028934665, DOI: doi:10.1016/j.jmb.2012.05.039
SCOTT, D. J.; KUMMER, L.; TREMMEL, D.; PLUCKTHUN, A.: "Stabilizing membrane proteins through protein engineering", CURR OPIN CHEM BIOL, vol. 17, 2013, pages 427 - 435, XP055105083, DOI: doi:10.1016/j.cbpa.2013.04.002
SEAL, R. P.; AMARA, S. G.: "A reentrant loop domain in the glutamate carrier EAAT1 participates in substrate binding and translocation", NEURON, vol. 21, 1998, pages 1487 - 1498
SEAL, R. P.; LEIGHTON, B. H.; AMARA, S. G.: "A model for the topology of excitatory amino acid transporters determined by the extracellular accessibility of substituted cysteines", NEURON, vol. 25, 2000, pages 695 - 706
SHIMAMOTO, K. ET AL.: "Characterization of novel L-threo-beta-benzyloxyaspartate derivatives, potent blockers of the glutamate transporters", MOL PHARMACOL, vol. 65, 2004, pages 1008 - 1015, XP055426844, DOI: doi:10.1124/mol.65.4.1008
SHIMAMOTO, K.: "Glutamate transporter blockers for elucidation of the function of excitatory neurotransmission systems", CHEM REC, vol. 8, 2008, pages 182 - 199
SHIMIZU, K. ET AL.: "ASC amino-acid transporter 2 (ASCT2) as a novel prognostic marker in non-small cell lung cancer", BR J CANCER, vol. 110, 2014, pages 2030 - 2039
SHOUFFANI, A.; KANNER, B. I.: "Cholesterol is required for the reconstruction of the sodium- and chloride-coupled, gamma-aminobutyric acid transporter from rat brain", J BIOL CHEM, vol. 265, 1990, pages 6002 - 6008
SLOTBOOM, D. J.; KONINGS, W. N.; LOLKEMA, J. S.: "Structural features of the glutamate transporter family", MICROBIOL MOL BIOL REV, vol. 63, 1999, pages 293 - 307
STEIPE, B.; SCHILLER, B.; PLUCKTHUN, A.; STEINBACHER, S.: "Sequence statistics reliably predict stabilizing mutations in a protein domain", J MOL BIOL, vol. 240, 1994, pages 188 - 192, XP024008330, DOI: doi:10.1006/jmbi.1994.1434
TAO, Z. ET AL.: "Mechanism of cation binding to the glutamate transporter EAAC1 probed with mutation of the conserved amino acid residue Thr101", J BIOL CHEM, vol. 285, 2010, pages 17725 - 17733
TATE, C. G.; SCHERTLER, G. F.: "Engineering G protein-coupled receptors to facilitate their structure determination", CURR OPIN STRUCT BIOL, vol. 19, 2009, pages 386 - 395, XP026541926, DOI: doi:10.1016/j.sbi.2009.07.004
VAN GELDERMALSEN, M. ET AL.: "ASCT2/SLC1A5 controls glutamine uptake and tumour growth in triple-negative basal-like breast cancer", ONCOGENE, vol. 35, 2016, pages 3201 - 3208
WALES, T. E.; ENGEN, J. R.: "Hydrogen exchange mass spectrometry for the analysis of protein dynamics", MASS SPECTROM REV, vol. 25, 2006, pages 158 - 170, XP002408800, DOI: doi:10.1002/mas.20064
WALLIN, E.; VON HEIJNE, G.: "Genome-wide analysis of integral membrane proteins from eubacterial, archaean, and eukaryotic organisms", PROTEIN SCI, vol. 7, 1998, pages 1029 - 1038, XP001053440
WANG, Q. ET AL.: "Targeting ASCT2-mediated glutamine uptake blocks prostate cancer growth and tumour development", J PATHOL, vol. 236, 2015, pages 278 - 289
WANG, Q. ET AL.: "Targeting glutamine transport to suppress melanoma cell growth", INT J CANCER, vol. 135, 2014, pages 1060 - 1071
WATERHOUSE, A. M.; PROCTER, J. B.; MARTIN, D. M.; CLAMP, M.; BARTON, G. J.: "Jalview Version 2--a multiple sequence alignment editor and analysis workbench", BIOINFORMATICS, vol. 25, 2009, pages 1189 - 1191
WATZKE, N.; GREWER, C.: "The anion conductance of the glutamate transporter EAAC1 depends on the direction of glutamate transport", FEBS LETT, vol. 503, 2001, pages 121 - 125, XP004300872, DOI: doi:10.1016/S0014-5793(01)02715-6
WINTER, N.; KOVERMANN, P.; FAHLKE, C.: "A point mutation associated with episodic ataxia 6 increases glutamate transporter anion currents", BRAIN, vol. 135, 2012, pages 3416 - 3425
XIAO-DONG CHAO ET AL: "The Role of Excitatory Amino Acid Transporters in Cerebral Ischemia", NEUROCHEMICAL RESEARCH, KLUWER ACADEMIC PUBLISHERS-PLENUM PUBLISHERS, NE, vol. 35, no. 8, 4 May 2010 (2010-05-04), pages 1224 - 1230, XP019827370, ISSN: 1573-6903 *
Y. ITO ET AL: "Advanced evolutionary molecular engineering to produce thermostable cellulase by using a small but efficient library", PROTEIN ENGINEERING, DESIGN AND SELECTION, vol. 26, no. 1, 22 October 2012 (2012-10-22), GB, pages 73 - 79, XP055466444, ISSN: 1741-0126, DOI: 10.1093/protein/gzs072 *
YERNOOL D ET AL: "Structure of Glutamate Transporter homologue from Pyrococcus Horikoshii", NATURE, MACMILLAN JOURNALS LTD., ETC.|, vol. 431, 14 October 2004 (2004-10-14), pages 811 - 818, XP003015959, ISSN: 0028-0836, DOI: 10.1038/NATURE03018 *
YERNOOL, D.; BOUDKER, O.; JIN, Y.; GOUAUX, E.: "Structure of a glutamate transporter homologue from Pyrococcus horikoshii", NATURE, vol. 431, 2004, pages 811 - 818, XP003015959, DOI: doi:10.1038/nature03018
ZERANGUE, N.; KAVANAUGH, M. P.: "ASCT-1 is a neutral amino acid exchanger with chloride channel activity", J BIOL CHEM, vol. 271, 1996, pages 27991 - 27994
ZERANGUE, N.; KAVANAUGH, M. P.: "Flux coupling in a neuronal glutamate transporter", NATURE, vol. 383, 1996, pages 634 - 637
ZHANG, Y.; BENDAHAN, A.; ZARBIV, R.; KAVANAUGH, M. P.; KANNER, B. I.: "Molecular determinant of ion selectivity of a (Na+ + K+)-coupled rat brain glutamate transporter", PROC NATL ACAD SCI U S A, vol. 95, 1998, pages 751 - 755

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113537409A (en) * 2021-09-13 2021-10-22 烟台双塔食品股份有限公司 Pea protein data feature extraction method

Similar Documents

Publication Publication Date Title
Canul-Tec et al. Structure and allosteric inhibition of excitatory amino acid transporter 1
Kovacs et al. Analysis of the role of the C-terminal tail in the regulation of the epidermal growth factor receptor
Kang et al. Crystal structure of rhodopsin bound to arrestin by femtosecond X-ray laser
Geng et al. Structural mechanism of ligand activation in human calcium-sensing receptor
Goult et al. Structural studies on full-length talin1 reveal a compact auto-inhibited dimer: implications for talin activation
Garnham et al. Multivalent microtubule recognition by tubulin tyrosine ligase-like family glutamylases
Zhao et al. Crystal structure of the E. coli peptide transporter YbgH
Gazda et al. The myosin chaperone UNC-45 is organized in tandem modules to support myofilament formation in C. elegans
Dong et al. Structure and mechanism of the human NHE1-CHP1 complex
Huang et al. A molecular mechanism for the generation of ligand-dependent differential outputs by the epidermal growth factor receptor
Custódio et al. Structural comparison of GLUT1 to GLUT3 reveal transport regulation mechanism in sugar porter family
Matsuoka et al. Structure, mechanism and lipid-mediated remodeling of the mammalian Na+/H+ exchanger NHA2
Mönkemeyer et al. Chaperone function of Hgh1 in the biogenesis of eukaryotic elongation factor 2
Xu et al. Structural insights into sodium transport by the oxaloacetate decarboxylase sodium pump
Sun et al. Mechanistic insights into an atypical interaction between ATG8 and SH3P2 in Arabidopsis thaliana
Ho et al. The topology, structure and PE interaction of LITAF underpin a Charcot-Marie-Tooth disease type 1C
Sjøgaard-Frich et al. Dynamic Na+/H+ exchanger 1 (NHE1)–calmodulin complexes of varying stoichiometry and structure regulate Ca2+-dependent NHE1 activation
Covaceuszach et al. Structural flexibility of human α‐dystroglycan
WO2018150260A1 (en) Method for the identification of mutated proteins having modified thermal stability
Pinotsis et al. Evidence for a dimeric assembly of two titin/telethonin complexes induced by the telethonin C-terminus
Penmatsa et al. X-ray structure of the dopamine transporter in complex with tricyclic antidepressant
Rimsa et al. High-resolution structure of the M14-type cytosolic carboxypeptidase from Burkholderia cenocepacia refined exploiting PDB_REDO strategies
Mahbub et al. Structural insights into regulation of CNNM-TRPM7 divalent cation uptake by the small GTPase ARL15
Jirku et al. The characterization of a novel S100A1 binding site in the N-terminus of TRPM1
Schacherl et al. Crystallographic and biochemical characterization of the dimeric architecture of site-2 protease

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18710132

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18710132

Country of ref document: EP

Kind code of ref document: A1