460
Views
13
CrossRef citations to date
0
Altmetric
Review

Computational re-design of protein structures to improve solubility

ORCID Icon & ORCID Icon
Pages 1077-1088 | Received 14 Feb 2019, Accepted 25 Jun 2019, Published online: 08 Jul 2019

References

  • Aggarwal S. What’s fueling the biotech engine — 2007. Nat Biotechnol. 2008;26(11):1227–1234.
  • Lagassé HAD, Alexaki A, Simhadri VL, et al. Recent advances in (therapeutic protein) drug development. F1000Res. 2017. DOI:10.12688/f1000research.9970.1
  • Usmani SS, Bedi G, Samuel JS, et al. THPdb: database of FDA-approved peptide and protein therapeutics. PLoS One. 2017;12:e0181748.
  • Bandaranayake AD, Almo SC. Recent advances in mammalian protein production. FEBS Lett. 2014;588:253–260.
  • Butler M, Meneses-Acosta A. Recent advances in technology supporting biopharmaceutical production from mammalian cells. Appl Microbiol Biotechnol. 2012;96:885–894.
  • Food and Drug Administration. Purple Book: Lists of Licensed Biological Products with Reference Product Exclusivity and Biosimilarity or Interchangeability Evaluations. 2016;2016.
  • Hamrang Z, Rattray NJW, Pluen A. Proteins behaving badly: emerging technologies in profiling biopharmaceutical aggregation. Trends Biotechnol. 2013;31:448–458.
  • Tobin PH, Richards DH, Callender RA, et al. Protein engineering: a new frontier for biological therapeutics. Curr Drug Metab. 2014;15:743–756.
  • Lutz S. Beyond directed evolution–semi-rational protein engineering and design. Curr Opin Biotechnol. 2010;21:734–743.
  • Matasci M, Hacker DL, Baldi L, et al. Recombinant therapeutic protein production in cultivated mammalian cells: current status and future prospects. Drug Discov Today Technol. 2008;5:e37–e42.
  • Dintzis RZ, Okajima M, Middleton MH, et al. The immunogenicity of soluble haptenated polymers is determined by molecular mass and hapten valence. J Immunol. 1989;143:1239–1244.
  • Bachmann MF, Rohrer UH, Kündig TM, et al. The influence of antigen organization on B cell responsiveness. Science. 1993;262:1448–1451.
  • Seong S-Y, Matzinger P. Hydrophobicity: an ancient damage-associated molecular pattern that initiates innate immune responses. Nat Rev Immunol. 2004;4:469–478.
  • Semba H, Ichige E, Imanaka T, et al. Efficient production of active form recombinant cassava hydroxynitrile lyase using escherichia coli in low-temperature culture. In: Fett-Neto A, editor. Methods in molecular biology. Clifton, N.J.: Humana Press;2010. p. 133–144.
  • Yang Q, Xu J, Li M, et al. High-level expression of a soluble snake venom enzyme, gloshedobin, in E. coli in the presence of metal ions. Biotechnol Lett. 2003;25:607–610.
  • García-Fruitós E, Martínez-Alonso M, Gonzàlez-Montalbán N, et al. Divergent genetic control of protein solubility and conformational quality in Escherichia coli. J Mol Biol. 2007;374:195–205.
  • Tokuriki N, Tawfik DS. Chaperonin overexpression promotes genetic variation and enzyme evolution. Nature. 2009;459:668–673.
  • Casi G, Neri D. Antibody–drug conjugates: basic concepts, examples and future perspectives. J Control Release. 2012;161:422–428.
  • Levin D, Golding B, Strome SE, et al. Fc fusion as a platform technology: potential for modulating immunogenicity. Trends Biotechnol. 2015;33:27–34.
  • Rath T, Baker K, Dumont JA, et al. Fc-fusion proteins and FcRn: structural insights for longer-lasting and more effective therapeutics. Crit Rev Biotechnol. 2015;35:235–254.
  • Andersen JT, Pehrson R, Tolmachev V, et al. Extending half-life by indirect targeting of the neonatal Fc receptor (FcRn) using a minimal albumin binding domain. J Biol Chem. 2011;286:5234–5241.
  • Turecek PL, Bossard MJ, Schoetens F, et al. PEGylation of biopharmaceuticals: a review of chemistry and nonclinical safety information of approved drugs. J Pharm Sci. 2016;105:460–475.
  • Sørensen HP, Mortensen KK. Soluble expression of recombinant proteins in the cytoplasm of Escherichia coli. Microb Cell Fact. 2005;4:1.
  • Waldo GS. Genetic screens and directed evolution for protein solubility. Curr Opin Chem Biol. 2003;7:33–38.
  • Aharoni A, Gaidukov L, Yagur S, et al. Directed evolution of mammalian paraoxonases PON1 and PON3 for bacterial expression and catalytic specialization. Proc Natl Acad Sci U S A. 2004;101:482–487.
  • Koepf E, Richert M, Braunschweig B, et al. Impact of formulation pH on physicochemical protein characteristics at the liquid-air interface. Int J Pharm. 2018;541:234–245.
  • Cleland JL, Powell MF, Shire SJ. The development of stable protein formulations: a close look at protein aggregation, deamidation, and oxidation. Crit Rev Ther Drug Carrier Syst. 1993;10:307–377.
  • Shire SJ, Shahrokh Z, Liu J. Challenges in the development of high protein concentration formulations. J Pharm Sci. 2004;93:1390–1402.
  • Wakankar AA, Borchardt RT. Formulation considerations for proteins susceptible to asparagine deamidation and aspartate isomerization. J Pharm Sci. 2006;95:2321–2336.
  • Frokjaer S, Otzen DE. Protein drug stability: a formulation challenge. Nat Rev Drug Discov. 2005;4:298–306.
  • Roberts CJ. Protein aggregation and its impact on product quality. Curr Opin Biotechnol. 2014;30:211–217.
  • Roberts CJ. Therapeutic protein aggregation: mechanisms, design, and control. Trends Biotechnol. 2014;32:372–380.
  • Chiti F, Calamai M, Taddei N, et al. Studies of the aggregation of mutant proteins in vitro provide insights into the genetics of amyloid diseases. Proc Natl Acad Sci. 2002;99:16419–16426.
  • Ventura S, Zurdo J, Narayanan S, et al. Short amino acid stretches can mediate amyloid formation in globular proteins: the Src homology 3 (SH3) case. Proc Natl Acad Sci U S A. 2004;101:7258–7263.
  • Pawar AP, DuBay KF, Zurdo J, et al. Prediction of “Aggregation-prone” and “Aggregation-susceptible” regions in proteins associated with neurodegenerative diseases. J Mol Biol. 2005;350:379–392.
  • Chiti F, Stefani M, Taddei N, et al. Rationalization of the effects of mutations on peptide and protein aggregation rates. Nature. 2003;424:805–808.
  • DuBay KF, Pawar AP, Chiti F, et al. Prediction of the absolute aggregation rates of amyloidogenic polypeptide chains. J Mol Biol. 2004;341:1317–1326.
  • Tartaglia GG, Cavalli A, Pellarin R, et al. Prediction of aggregation rate and aggregation-prone segments in polypeptide sequences. Protein Sci. 2005;14:2723–2734.
  • Galzitskaya OV, Garbuzynskiy SO, Lobanov MY. Prediction of amyloidogenic and disordered regions in protein chains. PLoS Comput Biol. 2006;2:e177.
  • Zibaee S, Makin OS, Goedert M, et al. A simple algorithm locates beta-strands in the amyloid fibril core of alpha-synuclein, Abeta, and tau using the amino acid sequence alone. Protein Sci. 2007;16:906–918.
  • Tian J, Wu N, Guo J, et al. Prediction of amyloid fibril-forming segments based on a support vector machine. BMC Bioinformatics. 2009;10:S45.
  • Yoon S, Welsh WJ. Detecting hidden sequence propensity for amyloid fibril formation. Protein Sci. 2004;13:2149–2160.
  • Yoon S, Welsh WJ. Rapid assessment of contact-dependent secondary structure propensity: relevance to amyloidogenic sequences. Proteins Struct Funct Bioinforma. 2005;60:110–117.
  • Yoon S, Welsh WJ, Jung H, et al. Do: CSSP2: an improved method for predicting contact-dependent secondary structure propensity. Comput Biol Chem. 2007;31:373–377.
  • Garbuzynskiy SO, Lobanov MY, Galzitskaya OV:. FoldAmyloid: a method of prediction of amyloidogenic regions from protein sequence. Bioinformatics. 2010;26:326–332.
  • Zhang Z, Chen H, Lai L. Identification of amyloid fibril-forming segments based on structure and residue-based statistical potential. Bioinformatics. 2007;23:2218–2225.
  • Bryan AW, Menke M, Cowen LJ, et al. BETASCAN: probable beta-amyloids identified by pairwise probabilistic analysis. PLoS Comput Biol. 2009;5:e1000333.
  • Maurer-Stroh S, Debulpaep M, Kuemmerer N, et al. Exploring the sequence determinants of amyloid structure using position-specific scoring matrices. Nat Methods. 2010;7:237–242.
  • Bryan AW, O’Donnell CW, Menke M, et al. STITCHER: dynamic assembly of likely amyloid and prion β-structures from secondary structure predictions. Proteins Struct Funct Bioinforma. 2012;80:410–420.
  • Gasior P, Kotulska M. FISH Amyloid - a new method for finding amyloidogenic segments in proteins based on site specific co-occurrence of aminoacids. BMC Bioinformatics. 2014;15:54.
  • Thangakani AM, Kumar S, Nagarajan R, et al. GAP: towards almost 100 percent prediction for β-strand-mediated aggregating peptides with distinct morphologies. Bioinformatics. 2014;30:1983–1990.
  • Família C, Dennison SR, Quintas A, et al. Prediction of peptide and protein propensity for amyloid formation. PLoS One. 2015;10:e0134679.
  • Ahmed AB, Znassi N, Château M-T, et al. A structure-based approach to predict predisposition to amyloidosis. Alzheimer’s Dement. 2015;11:681–690.
  • Trovato A, Chiti F, Maritan A, et al. Insight into the structure of amyloid fibrils from the analysis of globular proteins. PLoS Comput Biol. 2006;2:e170.
  • Walsh I, Seno F, Tosatto SCE, et al. PASTA 2.0: an improved server for protein aggregation prediction. Nucleic Acids Res. 2014;42:W301–W307.
  • Hebditch M, Carballo-Amador MA, Charonis S, et al. Protein–sol: a web tool for predicting protein solubility from sequence. Bioinformatics. 2017;33:3098–3100.
  • Frousios KK, Iconomidou VA, Karletidi C-M, et al. Amyloidogenic determinants are usually not buried. BMC Struct Biol. 2009;9:44.
  • Emily M, Talvas A, Delamarche C. MetAmyl: a METa-predictor for AMYLoid proteins. PLoS One. 2013;8:e79722.
  • Magnan CN, Randall A, Baldi P. SOLpro: accurate sequence-based prediction of protein solubility. Bioinformatics. 2009;25:2200–2207.
  • Smialowski P, Doose G, Torkler P, et al. PROSO II–a new method for protein solubility prediction. Febs J. 2012;279:2192–2200.
  • Zambrano R, Jamroz M, Szczasiuk A, et al. AGGRESCAN3D (A3D): server for prediction of aggregation properties of protein structures. Nucleic Acids Res. 2015;43:W306–W313.
  • Castillo V, Ventura S. Amyloidogenic regions and interaction surfaces overlap in globular proteins related to conformational diseases. PLoS Comput Biol. 2009;5(8):e1000476.
  • Kumar S, Singh SK, Wang X, et al. Coupling of aggregation and immunogenicity in biotherapeutics: T- and B-cell immune epitopes may contain aggregation-prone regions. Pharm Res. 2011;28:949–961.
  • Idicula-Thomas S, Balaji PV:. Understanding the relationship between the primary structure of proteins and its propensity to be soluble on overexpression in Escherichia coli. Protein Sci. 2005;14:582–592.
  • Chennamsetty N, Voynov V, Kayser V, et al. Design of therapeutic proteins with enhanced stability. Proc Natl Acad Sci U S A. 2009;106:11937–11942.
  • Sormanni P, Aprile FA, Vendruscolo M. The CamSol method of rational design of protein mutants with enhanced solubility. J Mol Biol. 2015;427:478–490.
  • Van Durme J, De Baets G, Van Der Kant R, et al. Solubis: a webserver to reduce protein aggregation through mutation. Protein Eng Des Sel. 2016;29:285–289.
  • Sankar K, Krystek SR, Carl SM, et al. AggScore: prediction of aggregation-prone regions in proteins based on the distribution of surface patches. Proteins Struct Funct Bioinforma. 2018;86:1147–1156.
  • Chennamsetty N, Helk B, Voynov V, et al. Aggregation-Prone Motifs in Human Immunoglobulin G. J Mol Biol. 2009;391:404–413.
  • Klimov DK, Thirumalai D. Dissecting the assembly of Abeta16-22 amyloid peptides into antiparallel beta sheets. Structure. 2003;11:295–307.
  • Gsponer J, Haberthür U, Caflisch A. The role of side-chain interactions in the early steps of aggregation: molecular dynamics simulations of an amyloid-forming peptide from the yeast prion Sup35. Proc Natl Acad Sci U S A. 2003;100:5154–5159.
  • Noon WH, Kong Y, Ma J. Molecular dynamics analysis of a buckyball-antibody complex. Proc Natl Acad Sci U S A. 2002;99(Suppl 2):6466–6470.
  • Black SD, Mould DR. Development of hydrophobicity parameters to analyze proteins which bear post- or cotranslational modifications. Anal Biochem. 1991;193:72–82.
  • Bommana R, Chai Q, Schöneich C, et al. Understanding the increased aggregation propensity of a light-exposed IgG1 monoclonal antibody using hydrogen exchange mass spectrometry, biophysical characterization, and structural analysis. J Pharm Sci. 2018;107:1498–1511.
  • Tiwari MK, Kepp KP. Modeling the aggregation propensity and toxicity of Amyloid-β variants. J Alzheimer’s Dis. 2015;47:215–229.
  • Chennamsetty N, Voynov V, Kayser V, et al. Prediction of protein binding regions. Proteins. 2011;79:888–897.
  • Ranjan S, Chung WK, Hofele R, et al. Investigation of cathepsin D–mAb interactions using a combined experimental and computational tool set. Biotechnol Bioeng. 2019. DOI:10.1002/bit.26968
  • Lauer TM, Agrawal NJ, Chennamsetty N, et al. Developability index: a rapid in silico tool for the screening of antibody aggregation propensity. J Pharm Sci. 2012;101:102–115.
  • Bas DC, Rogers DM, Jensen JH. Very fast prediction and rationalization of pKa values for protein-ligand complexes. Proteins Struct Funct Bioinforma. 2008;73:765–783.
  • Li H, Robertson AD, Jensen JH. Very fast empirical prediction and rationalization of protein pKa values. Proteins. 2005;61:704–721.
  • Voynov V, Chennamsetty N, Kayser V, et al. Predictive tools for stabilization of therapeutic proteins. MAbs. date unknown;1:580–582.
  • Tartaglia GG, Vendruscolo M. The Zyggregator method for predicting protein aggregation propensities. Chem Soc Rev. 2008;37:1395–1401.
  • Agostini F, Vendruscolo M, Tartaglia GG. Sequence-based prediction of protein solubility. J Mol Biol. 2012;421:237–241.
  • Wimley WC, White SH. Experimentally determined hydrophobicity scale for proteins at membrane interfaces. Nat Struct Biol. 1996;3:842–848.
  • Sormanni P, Vendruscolo M. Protein solubility predictions using the CamSol method in the study of protein homeostasis. Cold Spring Harb Perspect Biol. 2019. DOI:10.1101/cshperspect.a033845
  • Camilloni C, Sala BM, Sormanni P, et al. Rational design of mutations that change the aggregation rate of a protein while maintaining its native structure and stability. Sci Rep. 2016;6:25559.
  • Haglund E, Nguyen L, Schafer NP, et al. Uncovering the molecular mechanisms behind disease-associated leptin variants. J Biol Chem. 2018;293:12919–12933.
  • Sormanni P, Amery L, Ekizoglou S, et al. Rapid and accurate in silico solubility screening of a monoclonal antibody library. Sci Rep. 2017;7:8200.
  • Shan L, Mody N, Sormani P, et al. Developability assessment of engineered monoclonal antibody variants with a complex self-association behavior using complementary analytical and in silico tools. Mol Pharm. 2018;15:5697–5710.
  • De Baets G, Van Durme J, van der Kant R, et al. Solubis: optimize your protein: fig. 1. Bioinformatics. 2015;31:2580–2582.
  • Fernandez-Escamilla A-M, Rousseau F, Schymkowitz J, et al. Prediction of sequence-dependent and mutational effects on the aggregation of peptides and proteins. Nat Biotechnol. 2004;22:1302–1306.
  • Richardson JS, Richardson DC. Natural beta-sheet proteins use negative design to avoid edge-to-edge aggregation. Proc Natl Acad Sci U S A. 2002;99:2754–2759.
  • Rousseau F, Schymkowitz J, Serrano L. Protein aggregation and amyloidosis: confusion of the kinds? Curr Opin Struct Biol. 2006;16:118–126.
  • Monsellier E, Chiti F. Prevention of amyloid-like aggregation as a driving force of protein evolution. EMBO Rep. 2007;8:737–742.
  • Serrano L, Schymkowitz J, Borg J, et al. The FoldX web server: an online force field. Nucl Acids Res. 2005;33:W382–388.
  • van der Kant R, van Durme J, Rousseau F, et al. SolubiS: optimizing protein solubility by minimal point mutations. Methods Mol Biol. 2019;1873:317–333.
  • Ganesan A, Siekierska A, Beerten J, et al. Structural hot spots for the solubility of globular proteins. Nat Commun. 2016;7:10816.
  • van der Kant R, Karow-Zwick AR, Van Durme J, et al. Prediction and reduction of the aggregation of monoclonal antibodies. J Mol Biol. 2017;429:1244–1261.
  • Musil M, Konegger H, Hon J, et al. Computational design of stable and soluble biocatalysts. ACS Catal. 2019;9:1033–1054.
  • Krieger E, Koraimann G, Vriend G. Increasing the precision of comparative models with YASARA NOVA–a self-parameterizing force field. Proteins. 2002;47:393–402.
  • Conchillo-Solé O, de Groot NS, Avilés FX, et al. AGGRESCAN: a server for the prediction and evaluation of “hot spots” of aggregation in polypeptides. BMC Bioinformatics. 2007;8:65.
  • de Groot NS, Aviles FX, Vendrell J, et al. Mutagenesis of the central hydrophobic cluster in Abeta42 Alzheimer’s peptide. Side-chain properties correlate with aggregation propensities. Febs J. 2006;273:658–668.
  • Lee B, Richards FM. The interpretation of protein structures: estimation of static accessibility. J Mol Biol. 1971;55:379–400.
  • Gil-Garcia M, Bañó-Polo M, Varejão N, et al. Combining structural aggregation propensity and stability predictions to redesign protein solubility. Mol Pharm. 2018;15:3846–3859.
  • Jamroz M, Kolinski A, Kmiecik S. CABS-flex: server for fast simulation of protein structure fluctuations. Nucleic Acids Res. 2013;41:W427–31.
  • Guerois R, Nielsen JE, Serrano L. Predicting changes in the stability of proteins and protein complexes: a study of more than 1000 mutations. J Mol Biol. 2002;320:369–387.
  • Kmiecik S, Gront D, Kolinski M, et al. Coarse-grained protein models and their applications. Chem Rev. 2016;116:7898–7936.
  • Pujols J, Peña-Díaz S, Ventura S. AGGRESCAN3D: toward the Prediction of the aggregation propensities of protein structures. In: Gore M, Jagtap U, editors. Methods in molecular biology. Clifton, N.J.: Humana Press; 2018. p. 427–443.
  • Patel P, Parmar K, Vyas VK, et al. Combined in silico approaches for the identification of novel inhibitors of human islet amyloid polypeptide (hIAPP) fibrillation. J Mol Graph Model. 2017;77:295–310.
  • Žerovnik E. Putative alternative functions of human stefin B (cystatin B): binding to amyloid-beta, membranes, and copper. J Mol Recognit. 2017;30:e2562.
  • Bhandare VV, Ramaswamy A. The proteinopathy of D169G and K263E mutants at the RNA Recognition Motif (RRM) domain of tar DNA-binding protein (tdp43) causing neurological disorders: A computational study. J Biomol Struct Dyn. 2018;36:1075–1093.
  • Lei H, Smith PE. The role of the unfolded state in hairpin stability. Biophys J. 2003;85:3513–3520.
  • Manning MC, Chou DK, Murphy BM, et al. Stability of protein pharmaceuticals: an update. Pharm Res. 2010;27:544–575.
  • O’Brien CJ, Blanco MA, Costanzo JA, et al. Modulating non-native aggregation and electrostatic protein-protein interactions with computationally designed single-point mutations. Protein Eng Des Sel. 2016;29:231–243.
  • Costanzo JA, O’Brien CJ, Tiller K, et al. Conformational stability as a design target to control protein aggregation. Protein Eng Des Sel. 2014;27:157–167.
  • Lawrence MS, Phillips KJ, Liu DR. Supercharging proteins can impart unusual resilience. J Am Chem Soc. 2007;129:10110–10112.
  • Chen Y, Dokholyan NV. Natural selection against protein aggregation on self-interacting and essential proteins in yeast, fly, and worm. Mol Biol Evol. 2008;25:1530–1533.
  • Ciryam P, Tartaglia GG, Morimoto RI, et al. Widespread aggregation and neurodegenerative diseases are associated with supersaturated proteins. Cell Rep. 2013;5:781–790.
  • Wang X, Das TK, Singh SK, et al. Potential aggregation prone regions in biotherapeutics: A survey of commercial monoclonal antibodies. MAbs. 2009;1:254–267.
  • Rabia LA, Desai AA, Jhajj HS, et al. Understanding and overcoming trade-offs between antibody affinity, specificity, stability and solubility. Biochem Eng J. 2018;137:365–374.
  • Goldenzweig A, Goldsmith M, Hill SE, et al. Automated structure- and sequence-based design of proteins for high bacterial expression and stability. Mol Cell. 2016;63:337–346.
  • Keasar C, McGuffin LJ, Wallner B, et al. An analysis and evaluation of the WeFold collaborative for protein structure prediction and its pipelines in CASP11 and CASP12. Sci Rep. 2018;8:9939.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.