Improving recombinant protein production using TISIGNER.com

Planning experiments using accurate prediction algorithms could mitigate failures in recombinant protein production. We have developed TISIGNER.com with the aim of addressing the technical challenges in recombinant protein production. We offer two web services, TIsigner (Translation Initiation coding region designer) and SoDoPE (Soluble Domain for Protein Expression), which are specialised in prediction/optimisation of recombinant protein expression and solubility, respectively. Importantly, TIsigner and SoDoPE are linked, which allows users to switch between the tools when optimising their genes of interest.

[1]  P. Gardner,et al.  Solubility-Weighted Index: fast and accurate prediction of protein solubility , 2020, bioRxiv.

[2]  Marianne Rooman,et al.  Computational analysis of the amino acid interactions that promote or decrease protein solubility , 2018, Scientific Reports.

[3]  J. Plotkin,et al.  Synonymous but not the same: the causes and consequences of codon bias , 2011, Nature Reviews Genetics.

[4]  Adam Paul Arkin,et al.  Evaluation of 244,000 synthetic sequences reveals design principles to optimize translation in Escherichia coli , 2018, Nature Biotechnology.

[5]  Peter F. Stadler,et al.  Local RNA base pairing probabilities in large sequences , 2006, Bioinform..

[6]  Germán L. Rosano,et al.  Recombinant protein expression in Escherichia coli: advances and challenges , 2014, Front. Microbiol..

[7]  G. Waldo,et al.  Genetic screens and directed evolution for protein solubility. , 2003, Current opinion in chemical biology.

[8]  Reinhard Wolf,et al.  Coding-Sequence Determinants of Gene Expression in Escherichia coli , 2009 .

[9]  Tamir Tuller,et al.  Modelling the Efficiency of Codon–tRNA Interactions Based on Codon Usage Bias , 2014, DNA research : an international journal for rapid publication of reports on genes and genomes.

[10]  Stephan H. Bernhart,et al.  RNA Accessibility in cubic time , 2011, Algorithms for Molecular Biology.

[11]  P. Gardner,et al.  Highly accessible translation initiation sites are predictive of successful heterologous protein expression , 2019, bioRxiv.

[12]  M. Moore,et al.  mRNA structure regulates protein expression through changes in functional half-life , 2019, Proceedings of the National Academy of Sciences.

[13]  Robert D. Finn,et al.  HMMER web server: 2018 update , 2018, Nucleic Acids Res..

[14]  Arkady B. Khodursky,et al.  Global analysis of mRNA decay and abundance in Escherichia coli at single-gene resolution using two-color fluorescent DNA microarrays , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[15]  Robert D. Finn,et al.  Rfam 13.0: shifting to a genome-centric resource for non-coding RNA families , 2017, Nucleic Acids Res..

[16]  Eran Segal,et al.  Deciphering the rules by which 5′-UTR sequences affect protein expression in yeast , 2013, Proceedings of the National Academy of Sciences.

[17]  P. Gardner,et al.  Avoidance of stochastic RNA interactions can be harnessed to control protein expression levels in bacteria and archaea , 2015, bioRxiv.

[18]  B. Strukelj,et al.  Current state and recent advances in biopharmaceutical production in Escherichia coli, yeasts and mammalian cells , 2013, Journal of Industrial Microbiology & Biotechnology.

[19]  P. Gardner,et al.  Annotating RNA motifs in sequences and alignments , 2014, bioRxiv.

[20]  C. Pace,et al.  Toward a molecular understanding of protein solubility: increased negative surface charge correlates with increased solubility. , 2012, Biophysical journal.

[21]  L. Domingues,et al.  Fusion tags for protein solubility, purification and immunogenicity in Escherichia coli: the novel Fh8 system , 2014, Front. Microbiol..

[22]  Peter F. Stadler,et al.  ViennaRNA Package 2.0 , 2011, Algorithms for Molecular Biology.

[23]  Dominic Esposito,et al.  Enhancement of soluble protein expression through the use of fusion tags. , 2006, Current opinion in biotechnology.

[24]  G. W. Hatfield,et al.  Nonrandom utilization of codon pairs in Escherichia coli. , 1989, Proceedings of the National Academy of Sciences of the United States of America.

[25]  Amit Sharma,et al.  DNASU plasmid and PSI:Biology-Materials repositories: resources to accelerate biological research , 2013, Nucleic Acids Res..

[26]  John D. Westbrook,et al.  TargetDB: a target registration database for structural genomics projects , 2004, Bioinform..

[27]  T. Tuller,et al.  Multiple roles of the coding sequence 5′ end in gene expression regulation , 2014, Nucleic acids research.

[28]  Chris M. Brown,et al.  The exon–intron gene structure upstream of the initiation codon predicts translation efficiency , 2018, Nucleic acids research.

[29]  P. Sharp,et al.  The codon Adaptation Index--a measure of directional synonymous codon usage bias, and its potential applications. , 1987, Nucleic acids research.

[30]  J. van Duin,et al.  Secondary structure of the ribosome binding site determines translational efficiency: a quantitative analysis. , 1990, Proceedings of the National Academy of Sciences of the United States of America.

[31]  Chun-Nan Hsu,et al.  Learning to predict expression efficacy of vectors in recombinant protein production , 2010, BMC Bioinformatics.

[32]  Sean R. Eddy,et al.  Infernal 1.1: 100-fold faster RNA homology searches , 2013, Bioinform..

[33]  L. Wernisch,et al.  Solving the riddle of codon usage preferences: a test for translational selection. , 2004, Nucleic acids research.

[34]  E. Marcotte,et al.  Global signatures of protein and mRNA expression levelsw , 2009 .

[35]  Paul J. Choi,et al.  Quantifying E. coli Proteome and Transcriptome with Single-Molecule Sensitivity in Single Cells , 2010, Science.

[36]  E. Grayhack,et al.  Synonymous Codons: Choose Wisely for Expression. , 2017, Trends in genetics : TIG.