Periplasmic Expression of 4/7 α-Conotoxin TxIA Analogs in E. coli Favors Ribbon Isomer Formation – Suggestion of a Binding Mode at the α7 nAChR

Peptides derived from animal venoms provide important research tools for biochemical and pharmacological characterization of receptors, ion channels, and transporters. Some venom peptides have been developed into drugs (such as the synthetic ω-conotoxin MVIIA, ziconotide) and several are currently undergoing clinical trials for various clinical indications. Challenges in the development of peptides include their usually limited supply from natural sources, cost-intensive chemical synthesis, and potentially complicated stereoselective disulfide-bond formation in the case of disulfide-rich peptides. In particular, if extended structure–function analysis is performed or incorporation of stable isotopes for NMR studies is required, the comparatively low yields and high costs of synthesized peptides might constitute a limiting factor. Here we investigated the expression of the 4/7 α-conotoxin TxIA, a potent blocker at α3β2 and α7 nicotinic acetylcholine receptors (nAChRs), and three analogs in the form of maltose binding protein fusion proteins in Escherichia coli. Upon purification via nickel affinity chromatography and release of the toxins by protease cleavage, HPLC analysis revealed one major peak with the correct mass for all peptides. The final yield was 1–2 mg of recombinant peptide per liter of bacterial culture. Two-electrode voltage clamp analysis on oocyte-expressed nAChR subtypes demonstrated the functionality of these peptides but also revealed a 30 to 100-fold potency decrease of expressed TxIA compared to chemically synthesized TxIA. NMR spectroscopy analysis of TxIA and two of its analogs confirmed that the decreased activity was due to an alternative disulfide linkage rather than the missing C-terminal amidation, a post-translational modification that is common in α-conotoxins. All peptides preferentially formed in the ribbon conformation rather than the native globular conformation. Interestingly, in the case of the α7 nAChR, but not the α3β2 subtype, the loss of potency could be rescued by an R5D substitution. In conclusion, we demonstrate efficient expression of functional but alternatively folded ribbon TxIA variants in E. coli and provide the first structure–function analysis for a ribbon 4/7-α-conotoxin at α7 and α3β2 nAChRs. Computational analysis based on these data provide evidence for a ribbon α-conotoxin binding mode that might be exploited to design ligands with optimized selectivity.

[1]  U. Maskos,et al.  Role of the Nicotinic Acetylcholine Receptor Chrna5 Gene Mutation in Chronic Obstructive Pulmonary Disease , 2020 .

[2]  D. Craik,et al.  Stoichiometry dependent inhibition of rat &agr;3&bgr;4 nicotinic acetylcholine receptor by the ribbon isomer of &agr;‐conotoxin AuIB , 2018, Biochemical pharmacology.

[3]  C. Enjalbal,et al.  Synthesis, Structure and Biological Activity of CIA and CIB, Two α-Conotoxins from the Predation-Evoked Venom of Conus catus , 2018, Toxins.

[4]  R. M. Walsh,et al.  Structural principles of distinct assemblies of the human alpha 4 beta 2 nicotinic receptor. , 2018 .

[5]  R. M. Walsh,et al.  Structural principles of distinct assemblies of the human α4β2 nicotinic receptor , 2018, Nature.

[6]  J. McIntosh,et al.  Nicotinic acetylcholine receptors in neuropathic and inflammatory pain , 2018, FEBS letters.

[7]  K. Lindorff-Larsen,et al.  How well do force fields capture the strength of salt bridges in proteins? , 2018, bioRxiv.

[8]  Jinpeng Yu,et al.  Expression in Escherichia coli of fusion protein comprising α‐conotoxin TxIB and preservation of selectivity to nicotinic acetylcholine receptors in the purified product , 2018, Chemical biology & drug design.

[9]  S. Dutertre,et al.  α-Conotoxins to explore the molecular, physiological and pathophysiological functions of neuronal nicotinic acetylcholine receptors , 2017, Neuroscience Letters.

[10]  A. Nicke,et al.  Nicotinic acetylcholine receptor inhibitors derived from snake and snail venoms , 2017, Neuropharmacology.

[11]  Richard Bonneau,et al.  Discovery of peptide ligands through docking and virtual screening at nicotinic acetylcholine receptor homology models , 2017, Proceedings of the National Academy of Sciences.

[12]  Mengsen Li,et al.  Residues Responsible for the Selectivity of α-Conotoxins for Ac-AChBP or nAChRs , 2016, Marine drugs.

[13]  D. Craik,et al.  Structure-Activity Studies of Cysteine-Rich α-Conotoxins that Inhibit High-Voltage-Activated Calcium Channels via GABA(B) Receptor Activation Reveal a Minimal Functional Motif. , 2016, Angewandte Chemie.

[14]  M. Zieliński,et al.  Expression of recombinant human bifunctional peptidylglycine α-amidating monooxygenase in CHO cells and its use for insulin analogue modification. , 2016, Protein expression and purification.

[15]  Jinpeng Yu,et al.  Recombinant Expression and Characterization of α-Conotoxin LvIA in Escherichia coli , 2016, Marine drugs.

[16]  S. Jayakar,et al.  Orthosteric and Allosteric Ligands of Nicotinic Acetylcholine Receptors for Smoking Cessation , 2015, Front. Mol. Neurosci..

[17]  M. Christie,et al.  Conotoxin Interactions with α9α10-nAChRs: Is the α9α10-Nicotinic Acetylcholine Receptor an Important Therapeutic Target for Pain Management? , 2015, Toxins.

[18]  Uwe Maskos,et al.  Role of the nicotinic acetylcholine receptor in Alzheimer's disease pathology and treatment , 2015, Neuropharmacology.

[19]  J. Yakel,et al.  Nicotinic ACh receptors as therapeutic targets in CNS disorders. , 2015, Trends in pharmacological sciences.

[20]  M. Antoniewicz Methods and advances in metabolic flux analysis: a mini-review , 2015, Journal of Industrial Microbiology & Biotechnology.

[21]  D. Craik,et al.  Discovery, synthesis, and structure-activity relationships of conotoxins. , 2014, Chemical reviews.

[22]  D. Craik,et al.  A novel α4/7‐conotoxin LvIA from Conus lividus that selectively blocks α3β2 vs. α6/α3β2β3 nicotinic acetylcholine receptors , 2014, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[23]  Jinpeng Yu,et al.  Influence of Disulfide Connectivity on Structure and Bioactivity of α-Conotoxin TxIA , 2014, Molecules.

[24]  M. Yandell,et al.  Characterization of the peptidylglycine α-amidating monooxygenase (PAM) from the venom ducts of neogastropods, Conus bullatus and Conus geographus. , 2013, Toxicon : official journal of the International Society on Toxinology.

[25]  A. Bax,et al.  Protein backbone and sidechain torsion angles predicted from NMR chemical shifts using artificial neural networks , 2013, Journal of Biomolecular NMR.

[26]  G. King,et al.  Production of Recombinant Disulfide-Rich Venom Peptides for Structural and Functional Analysis via Expression in the Periplasm of E. coli , 2013, PloS one.

[27]  Brian D. Weitzner,et al.  Serverification of Molecular Modeling Applications: The Rosetta Online Server That Includes Everyone (ROSIE) , 2013, PloS one.

[28]  A. M. Phillips,et al.  Oxidative folding and preparation of α‐conotoxins for use in high‐throughput structure–activity relationship studies , 2013, Journal of peptide science : an official publication of the European Peptide Society.

[29]  H. Grubmüller,et al.  Efficient Binding of 4/7 α-Conotoxins to Nicotinic α4β2 Receptors Is Prevented by Arg185 and Pro195 in the α4 Subunit , 2012, Molecular Pharmacology.

[30]  Holger Gohlke,et al.  MMPBSA.py: An Efficient Program for End-State Free Energy Calculations. , 2012, Journal of chemical theory and computation.

[31]  A. Steiner,et al.  Modulation of Conotoxin Structure and Function Is Achieved through a Multienzyme Complex in the Venom Glands of Cone Snails* , 2012, The Journal of Biological Chemistry.

[32]  G. King,et al.  Functional Expression in Escherichia coli of the Disulfide-Rich Sea Anemone Peptide APETx2, a Potent Blocker of Acid-Sensing Ion Channel 3 , 2012, Marine drugs.

[33]  Vladimir B. Bajic,et al.  Conotoxins that Confer Therapeutic Possibilities , 2012, Marine drugs.

[34]  D. Yoshikami,et al.  Selective Purification of Recombinant Neuroactive Peptides Using the Flagellar Type III Secretion System , 2012, mBio.

[35]  Quentin Kaas,et al.  Delineation of the unbinding pathway of α-conotoxin ImI from the α7 nicotinic acetylcholine receptor. , 2012, The journal of physical chemistry. B.

[36]  David J. Craik,et al.  ConoServer: updated content, knowledge, and discovery tools in the conopeptide database , 2011, Nucleic Acids Res..

[37]  David J. Craik,et al.  Blockade of Neuronal α7-nAChR by α-Conotoxin ImI Explained by Computational Scanning and Energy Calculations , 2011, PLoS Comput. Biol..

[38]  D. Baker,et al.  Alternate states of proteins revealed by detailed energy landscape mapping. , 2011, Journal of molecular biology.

[39]  Nir London,et al.  Sub‐angstrom modeling of complexes between flexible peptides and globular proteins , 2010, Proteins.

[40]  D. Craik,et al.  Conopeptide characterization and classifications: an analysis using ConoServer. , 2010, Toxicon : official journal of the International Society on Toxinology.

[41]  David John Adams,et al.  α-Conotoxin AuIB Isomers Exhibit Distinct Inhibitory Mechanisms and Differential Sensitivity to Stoichiometry of α3β4 Nicotinic Acetylcholine Receptors* , 2010, The Journal of Biological Chemistry.

[42]  R. Dror,et al.  Improved side-chain torsion potentials for the Amber ff99SB protein force field , 2010, Proteins.

[43]  D. Bertrand,et al.  Interaction of α‐conotoxin ImII and its analogs with nicotinic receptors and acetylcholine‐binding proteins: additional binding sites on Torpedo receptor , 2009, Journal of neurochemistry.

[44]  A. Bax,et al.  TALOS+: a hybrid method for predicting protein backbone torsion angles from NMR chemical shifts , 2009, Journal of biomolecular NMR.

[45]  J. McIntosh,et al.  Alpha-conotoxins as pharmacological probes of nicotinic acetylcholine receptors , 2009, Acta Pharmacologica Sinica.

[46]  D. Craik,et al.  Structure of α-conotoxin BuIA: influences of disulfide connectivity on structural dynamics , 2007, BMC Structural Biology.

[47]  M. Parrinello,et al.  Canonical sampling through velocity rescaling. , 2007, The Journal of chemical physics.

[48]  A. Sali,et al.  Statistical potential for assessment and prediction of protein structures , 2006, Protein science : a publication of the Protein Society.

[49]  R. Kini,et al.  Effect of C-Terminal Amidation on Folding and Disulfide-Pairing of α-Conotoxin ImI† , 2005 .

[50]  R. Lewis,et al.  β2 Subunit Contribution to 4/7 α-Conotoxin Binding to the Nicotinic Acetylcholine Receptor* , 2005, Journal of Biological Chemistry.

[51]  Wayne Boucher,et al.  The CCPN data model for NMR spectroscopy: Development of a software pipeline , 2005, Proteins.

[52]  K. Gayler,et al.  Drugs from the sea: conopeptides as potential therapeutics. , 2004, Current medicinal chemistry.

[53]  A. Gomes,et al.  Determinants of Potency on α-Conotoxin MII, a Peptide Antagonist of Neuronal Nicotinic Receptors , 2004 .

[54]  David John Adams,et al.  Chemical and functional identification and characterization of novel sulfated alpha-conotoxins from the cone snail Conus anemone. , 2004, Journal of medicinal chemistry.

[55]  D. Craik,et al.  Isolation, Structure, and Activity of GID, a Novel α4/7-Conotoxin with an Extended N-terminal Sequence* , 2003, The Journal of Biological Chemistry.

[56]  C. Dominguez,et al.  HADDOCK: a protein-protein docking approach based on biochemical or biophysical information. , 2003, Journal of the American Chemical Society.

[57]  D. Craik,et al.  A New Level of Conotoxin Diversity, a Non-native Disulfide Bond Connectivity in α-Conotoxin AuIB Reduces Structural Definition but Increases Biological Activity* , 2002, The Journal of Biological Chemistry.

[58]  Dirk Labudde,et al.  A software tool for the prediction of Xaa-Pro peptide bond conformations in proteins based on 13C chemical shift statistics , 2002, Journal of biomolecular NMR.

[59]  D. Yoshikami,et al.  α-Conotoxin GIC from Conus geographus, a Novel Peptide Antagonist of Nicotinic Acetylcholine Receptors* , 2002, The Journal of Biological Chemistry.

[60]  T. Copeland,et al.  The P1' specificity of tobacco etch virus protease. , 2002, Biochemical and biophysical research communications.

[61]  D. Craik,et al.  Two new classes of conopeptides inhibit the α1-adrenoceptor and noradrenaline transporter , 2001, Nature Neuroscience.

[62]  D. Yoshikami,et al.  Isolation and Characterization of a Novel ConusPeptide with Apparent Antinociceptive Activity* , 2000, The Journal of Biological Chemistry.

[63]  H. Nagasawa,et al.  2,2′-Bispyridyl disulfide rapidly induces intramolecular disulfide bonds in peptides , 1999, Peptides.

[64]  R J Read,et al.  Crystallography & NMR system: A new software suite for macromolecular structure determination. , 1998, Acta crystallographica. Section D, Biological crystallography.

[65]  D. Craik,et al.  Structure determination of the three disulfide bond isomers of alpha-conotoxin GI: a model for the role of disulfide bonds in structural stability. , 1998, Journal of molecular biology.

[66]  K. Wüthrich,et al.  Torsion angle dynamics for NMR structure calculation with the new program DYANA. , 1997, Journal of molecular biology.

[67]  D. Yoshikami,et al.  A New -Conotoxin Which Targets 32 Nicotinic Acetylcholine Receptors (*) , 1996, The Journal of Biological Chemistry.

[68]  M. Billeter,et al.  MOLMOL: a program for display and analysis of macromolecular structures. , 1996, Journal of molecular graphics.

[69]  R. Hodges,et al.  1H, 13C and 15N random coil NMR chemical shifts of the common amino acids. I. Investigations of nearest-neighbor effects , 1995, Journal of biomolecular NMR.

[70]  A. J. Shaka,et al.  Water Suppression That Works. Excitation Sculpting Using Arbitrary Wave-Forms and Pulsed-Field Gradients , 1995 .

[71]  T. Blundell,et al.  Comparative protein modelling by satisfaction of spatial restraints. , 1993, Journal of molecular biology.

[72]  F. Richards,et al.  Relationship between nuclear magnetic resonance chemical shift and protein secondary structure. , 1991, Journal of molecular biology.

[73]  P. Wright,et al.  Sensitivity improvement in proton-detected two-dimensional heteronuclear correlation NMR spectroscopy , 1991 .

[74]  K. Wüthrich NMR of proteins and nucleic acids , 1988 .

[75]  Richard R. Ernst,et al.  Coherence transfer by isotropic mixing: Application to proton correlation spectroscopy , 1983 .

[76]  Richard R. Ernst,et al.  Investigation of exchange processes by two‐dimensional NMR spectroscopy , 1979 .

[77]  E. G. Sherry,et al.  STRUCTURE DETERMINATION I , 1960 .

[78]  T. Sixma,et al.  AChBP-targeted α-conotoxin correlates distinct binding orientations with nAChR subtype selectivity , 2007, The EMBO Journal.

[79]  Christopher J. Williams,et al.  MolProbity: More and better reference data for improved all‐atom structure validation , 2018, Protein science : a publication of the Protein Society.

[80]  D. Craik,et al.  Stoichiometry dependent inhibition of rat α3β4 nicotinic acetylcholine receptor by the ribbon isomer of α-conotoxin AuIB , 2018 .

[81]  Jack Snoeyink,et al.  Scientific benchmarks for guiding macromolecular energy function improvement. , 2013, Methods in enzymology.

[82]  S. Wonnacott,et al.  Nicotinic ACh Receptors , 2007 .

[83]  M. Nilges,et al.  Influence of non-bonded parameters on the quality of NMR structures: A new force field for NMR structure calculation , 1999, Journal of biomolecular NMR.

[84]  F. Barrantes The Nicotinic Acetylcholine Receptor , 1998, Biotechnology Intelligence Unit.

[85]  O. Pongs,et al.  A vector for the synthesis of cRNAs encoding Myc epitope-tagged proteins in Xenopus laevis oocytes. , 1995, Gene.

[86]  B D Sykes,et al.  1H, 13C and 15N random coil NMR chemical shifts of the common amino acids. I. Investigations of nearest-neighbor effects , 1995, Journal of biomolecular NMR.

[87]  A Karlin,et al.  Nicotinic acetylcholine receptors. , 1977 .

[88]  Miron Livny,et al.  RECOORD: A recalculated coordinate database of 500+ proteins from the PDB using restraints from the BioMagResBank , 2005, Proteins.