1
|
Atsavapranee B, Stark CD, Sunden F, Thompson S, Fordyce PM. Fundamentals to function: Quantitative and scalable approaches for measuring protein stability. Cell Syst 2021; 12:547-560. [PMID: 34139165 DOI: 10.1016/j.cels.2021.05.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/16/2021] [Accepted: 05/07/2021] [Indexed: 12/11/2022]
Abstract
Folding a linear chain of amino acids into a three-dimensional protein is a complex physical process that ultimately confers an impressive range of diverse functions. Although recent advances have driven significant progress in predicting three-dimensional protein structures from sequence, proteins are not static molecules. Rather, they exist as complex conformational ensembles defined by energy landscapes spanning the space of sequence and conditions. Quantitatively mapping the physical parameters that dictate these landscapes and protein stability is therefore critical to develop models that are capable of predicting how mutations alter function of proteins in disease and informing the design of proteins with desired functions. Here, we review the approaches that are used to quantify protein stability at a variety of scales, from returning multiple thermodynamic and kinetic measurements for a single protein sequence to yielding indirect insights into folding across a vast sequence space. The physical parameters derived from these approaches will provide a foundation for models that extend beyond the structural prediction to capture the complexity of conformational ensembles and, ultimately, their function.
Collapse
Affiliation(s)
| | - Catherine D Stark
- Department of Biochemistry, Stanford University, Stanford, CA 94305, USA; ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Fanny Sunden
- Department of Biochemistry, Stanford University, Stanford, CA 94305, USA
| | - Samuel Thompson
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.
| | - Polly M Fordyce
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; ChEM-H, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94110, USA.
| |
Collapse
|
2
|
Fang J. A critical review of five machine learning-based algorithms for predicting protein stability changes upon mutation. Brief Bioinform 2019; 21:1285-1292. [PMID: 31273374 DOI: 10.1093/bib/bbz071] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/14/2019] [Accepted: 05/16/2019] [Indexed: 01/02/2023] Open
Abstract
A number of machine learning (ML)-based algorithms have been proposed for predicting mutation-induced stability changes in proteins. In this critical review, we used hypothetical reverse mutations to evaluate the performance of five representative algorithms and found all of them suffer from the problem of overfitting. This approach is based on the fact that if a wild-type protein is more stable than a mutant protein, then the same mutant is less stable than the wild-type protein. We analyzed the underlying issues and suggest that the main causes of the overfitting problem include that the numbers of training cases were too small, and the features used in the models were not sufficiently informative for the task. We make recommendations on how to avoid overfitting in this important research area and improve the reliability and robustness of ML-based algorithms in general.
Collapse
Affiliation(s)
- Jianwen Fang
- Computational & Systems Biology Branch, Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Drive, Rockville, MD 20850, USA
| |
Collapse
|
3
|
Kent JA, Bommaraju TV, Barnicki SD, Kyung YS, Zhang GG. Industrial Production of Therapeutic Proteins: Cell Lines, Cell Culture, and Purification. HANDBOOK OF INDUSTRIAL CHEMISTRY AND BIOTECHNOLOGY 2017. [PMCID: PMC7121293 DOI: 10.1007/978-3-319-52287-6_29] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
A central pillar of the biotechnology and pharmaceutical industries continues to be the development of biological drug products manufactured from engineered mammalian cell lines. Since the hugely successful launch of human tissue plasminogen activator in 1987 and erythropoietin in 1988, the biopharmaceutical market has grown immensely. In 2014, biotherapeutics made up a significant portion of global drug sales as 7 of the top 10 and 21 of top 50 selling pharmaceuticals in the world were biologics with over US$100 billion in global sales (Table 1, [1]).
Collapse
|
4
|
Abstract
Using structure and sequence based analysis we can engineer proteins to increase their thermal stability.
Collapse
Affiliation(s)
- H. Pezeshgi Modarres
- Molecular Cell Biomechanics Laboratory
- Departments of Bioengineering and Mechanical Engineering
- University of California Berkeley
- Berkeley
- USA
| | - M. R. Mofrad
- Molecular Cell Biomechanics Laboratory
- Departments of Bioengineering and Mechanical Engineering
- University of California Berkeley
- Berkeley
- USA
| | - A. Sanati-Nezhad
- BioMEMS and Bioinspired Microfluidic Laboratory
- Department of Mechanical and Manufacturing Engineering
- University of Calgary
- Calgary
- Canada
| |
Collapse
|
5
|
Gregoire S, Zhang S, Costanzo J, Wilson K, Fernandez EJ, Kwon I. Cis-suppression to arrest protein aggregation in mammalian cells. Biotechnol Bioeng 2013; 111:462-74. [PMID: 24114411 DOI: 10.1002/bit.25119] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 08/18/2013] [Accepted: 09/09/2013] [Indexed: 12/20/2022]
Abstract
Protein misfolding and aggregation are implicated in numerous human diseases and significantly lower production yield of proteins expressed in mammalian cells. Despite the importance of understanding and suppressing protein aggregation in mammalian cells, a protein design and selection strategy to modulate protein misfolding/aggregation in mammalian cells has not yet been reported. In this work, we address the particular challenge presented by mutation-induced protein aggregation in mammalian cells. We hypothesize that an additional mutation(s) can be introduced in an aggregation-prone protein variant, spatially near the original mutation, to suppress misfolding and aggregation (cis-suppression). As a model protein, we chose human copper, zinc superoxide dismutase mutant (SOD1(A4V) ) containing an alanine to valine mutation at residue 4, associated with the familial form of amyotrophic lateral sclerosis. We used the program RosettaDesign to identify Phe20 in SOD1(A4V) as a key residue responsible for SOD1(A4V) conformational destabilization. This information was used to rationally develop a pool of candidate mutations at the Phe20 site. After two rounds of mammalian-cell based screening of the variants, three novel SOD1(A4V) variants with a significantly reduced aggregation propensity inside cells were selected. The enhanced stability and reduced aggregation propensity of the three novel SOD1(A4V) variants were verified using cell fractionation and in vitro stability assays.
Collapse
Affiliation(s)
- Simpson Gregoire
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, 22904-4741
| | | | | | | | | | | |
Collapse
|
6
|
Yennamalli RM, Rader AJ, Kenny AJ, Wolt JD, Sen TZ. Endoglucanases: insights into thermostability for biofuel applications. BIOTECHNOLOGY FOR BIOFUELS 2013; 6:136. [PMID: 24070146 PMCID: PMC3856469 DOI: 10.1186/1754-6834-6-136] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 09/24/2013] [Indexed: 05/03/2023]
Abstract
Obtaining bioethanol from cellulosic biomass involves numerous steps, among which the enzymatic conversion of the polymer to individual sugar units has been a main focus of the biotechnology industry. Among the cellulases that break down the polymeric cellulose are endoglucanases that act synergistically for subsequent hydrolytic reactions. The endoglucanases that have garnered relatively more attention are those that can withstand high temperatures, i.e., are thermostable. Although our understanding of thermostability in endoglucanases is incomplete, some molecular features that are responsible for increased thermostability have been recently identified. This review focuses on the investigations of endoglucanases and their implications for biofuel applications.
Collapse
Affiliation(s)
- Ragothaman M Yennamalli
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames 50011, IA, USA
- Present Address: Department of Biochemistry and Cell Biology, Rice University, Houston, TX 77005, USA
| | - Andrew J Rader
- Department of Physics, Indiana University-Purdue University Indianapolis, Indianapolis 46202, IN, USA
- Present Address: State Farm Insurance, Indianapolis 46240, IN, USA
| | - Adam J Kenny
- Biosafety Institute for Genetically Modified Agricultural Products and Department of Agronomy, Iowa State University, Ames 50011, IA, USA
- Present Address: Brownells, Inc, Montezuma, IA 50171, USA
| | - Jeffrey D Wolt
- Biosafety Institute for Genetically Modified Agricultural Products and Department of Agronomy, Iowa State University, Ames 50011, IA, USA
| | - Taner Z Sen
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames 50011, IA, USA
- Bioinformatics and Computational Biology Program, Iowa State University, Ames 50011, IA, USA
- 1025 Crop Genome Informatics Lab, Iowa State University, Ames 50011, IA, USA
| |
Collapse
|
7
|
Li Y, Fang J. PROTS-RF: a robust model for predicting mutation-induced protein stability changes. PLoS One 2012; 7:e47247. [PMID: 23077576 PMCID: PMC3471942 DOI: 10.1371/journal.pone.0047247] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 09/11/2012] [Indexed: 11/19/2022] Open
Abstract
The ability to improve protein thermostability via protein engineering is of great scientific interest and also has significant practical value. In this report we present PROTS-RF, a robust model based on the Random Forest algorithm capable of predicting thermostability changes induced by not only single-, but also double- or multiple-point mutations. The model is built using 41 features including evolutionary information, secondary structure, solvent accessibility and a set of fragment-based features. It achieves accuracies of 0.799,0.782, 0.787, and areas under receiver operating characteristic (ROC) curves of 0.873, 0.868 and 0.862 for single-, double- and multiple- point mutation datasets, respectively. Contrary to previous suggestions, our results clearly demonstrate that a robust predictive model trained for predicting single point mutation induced thermostability changes can be capable of predicting double and multiple point mutations. It also shows high levels of robustness in the tests using hypothetical reverse mutations. We demonstrate that testing datasets created based on physical principles can be highly useful for testing the robustness of predictive models.
Collapse
Affiliation(s)
- Yunqi Li
- Applied Bioinformatics Laboratory, The University of Kansas, Lawrence, Kansas, United States of America
| | - Jianwen Fang
- Applied Bioinformatics Laboratory, The University of Kansas, Lawrence, Kansas, United States of America
- * E-mail:
| |
Collapse
|
8
|
Bentley AA, Merkulov SM, Peng Y, Rozmarynowycz R, Qi X, Pusztai-Carey M, Merrick WC, Yee VC, McCrae KR, Komar AA. Chimeric glutathione S-transferases containing inserts of kininogen peptides: potential novel protein therapeutics. J Biol Chem 2012; 287:22142-50. [PMID: 22577144 DOI: 10.1074/jbc.m112.372854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The study of synthetic peptides corresponding to discrete regions of proteins has facilitated the understanding of protein structure-activity relationships. Short peptides can also be used as powerful therapeutic agents. However, in many instances, small peptides are prone to rapid degradation or aggregation and may lack the conformation required to mimic the functional motifs of the protein. For peptides to function as pharmacologically active agents, efficient production or expression, high solubility, and retention of biological activity through purification and storage steps are required. We report here the design, expression, and functional analysis of eight engineered GST proteins (denoted GSHKTs) in which peptides ranging in size from 8 to 16 amino acids and derived from human high molecular weight kininogen (HK) domain 5 were inserted into GST (between Gly-49 and Leu-50). Peptides derived from HK are known to inhibit cell proliferation, angiogenesis, and tumor metastasis, and the biological activity of the HK peptides was dramatically (>50-fold) enhanced following insertion into GST. GSHKTs are soluble and easily purified from Escherichia coli by affinity chromatography. Functionally, these hybrid proteins cause inhibition of endothelial cell proliferation. Crystallographic analysis of GSHKT10 and GSHKT13 (harboring 10- and 13-residue HK peptides, respectively) showed that the overall GST structure was not perturbed. These results suggest that the therapeutic efficacy of short peptides can be enhanced by insertion into larger proteins that are easily expressed and purified and that GST may potentially be used as such a carrier.
Collapse
Affiliation(s)
- Amber A Bentley
- Center for Gene Regulation in Health and Disease, Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, Ohio 44115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Li Y, Zhang J, Tai D, Middaugh CR, Zhang Y, Fang J. PROTS: a fragment based protein thermo-stability potential. Proteins 2011; 80:81-92. [PMID: 21976375 DOI: 10.1002/prot.23163] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 07/18/2011] [Accepted: 07/31/2011] [Indexed: 12/30/2022]
Abstract
Designing proteins with enhanced thermo-stability has been a main focus of protein engineering because of its theoretical and practical significance. Despite extensive studies in the past years, a general strategy for stabilizing proteins still remains elusive. Thus effective and robust computational algorithms for designing thermo-stable proteins are in critical demand. Here we report PROTS, a sequential and structural four-residue fragment based protein thermo-stability potential. PROTS is derived from a nonredundant representative collection of thousands of thermophilic and mesophilic protein structures and a large set of point mutations with experimentally determined changes of melting temperatures. To the best of our knowledge, PROTS is the first protein stability predictor based on integrated analysis and mining of these two types of data. Besides conventional cross validation and blind testing, we introduce hypothetical reverse mutations as a means of testing the robustness of protein thermo-stability predictors. In all tests, PROTS demonstrates the ability to reliably predict mutation induced thermo-stability changes as well as classify thermophilic and mesophilic proteins. In addition, this white-box predictor allows easy interpretation of the factors that influence mutation induced protein stability changes at the residue level.
Collapse
Affiliation(s)
- Yunqi Li
- Applied Bioinformatics Laboratory, the University of Kansas, Lawrence, Kansas 66047, USA
| | | | | | | | | | | |
Collapse
|
10
|
Nwankwo N, Seker H. A signal processing-based bioinformatics approach to assessing drug resistance: human immunodeficiency virus as a case study. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2011; 2010:1836-9. [PMID: 21096145 DOI: 10.1109/iembs.2010.5626439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Measuring drug resistance is one of the challenging and essential pharmaceutical activities. It is a laborious and costly laboratory-based experimentation. Various clinical and experimental analyses for measuring drug resistance have been carried out. Results have been obtained for different types of therapeutic agents as a consequence of changes in the amino acids compositions in the sequence (mutation) of the organisms involved. In the same manner, the positions of these amino acids alterations and the level of resistance (folds) have also been experimentally identified. For example, G36S and V38M mutation in the Human Immunodeficiency Virus (HIV) Transmembrane glycoprotein (gp41) has been found to cause 100-fold resistance. However, there does not seem to have bioinformatics method developed in which the amino acid information of the proteins involved in the studies were used to computationally assess the degree of drug resistance without involving laboratory-based experimental procedure. The post-genomic era has witnessed the relevance of Bioinformatics approaches in the analysis of huge biomedical data. One such approach is the analysis of protein residues using digital signal processing technique such as informational spectrum method (ISM). Therefore, we propose a new bioinformatics method that is capable of assessing drug resistance without the use of any laboratory-based experiments. This new method incorporates ISM, sequence information of the proteins and other relevant information. By using the ISM and EIIP amino acid scale, the technique was applied in three classes of anti-HIV/AIDS drugs as a case study. It is observed that the protein residues of the susceptible strains attained the maximal peak amplitude at the consensus frequency while the resistant strains maintained lower amplitudes. This result signifies lower contribution from the resistant strains due to the mutation. The findings are consistent with those of the experimental ones and therefore suggest that the approach taken can be used to help assess drug resistance without laboratory-based experimentation. It should also be noted that the method can be applied in other drug resistance studies where sequence information of proteins is available and help design a computer-aided drug resistance calculator.
Collapse
Affiliation(s)
- Norbert Nwankwo
- Bio-Health Informatics Research Group within the Centre for Computational Intelligence, Department of Informatics, Faculty of Technology, De Montfort University, Leicester, LE1 9BH, The United Kingdom.
| | | |
Collapse
|
11
|
Balaraman GS, Bhattacharya S, Vaidehi N. Structural insights into conformational stability of wild-type and mutant beta1-adrenergic receptor. Biophys J 2010; 99:568-77. [PMID: 20643076 DOI: 10.1016/j.bpj.2010.04.075] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Revised: 04/09/2010] [Accepted: 04/16/2010] [Indexed: 11/26/2022] Open
Abstract
Recent experiments to derive a thermally stable mutant of turkey beta-1-adrenergic receptor (beta1AR) have shown that a combination of six single point mutations resulted in a 20 degrees C increase in thermal stability in mutant beta1AR. Here we have used the all-atom force-field energy function to calculate a stability score to detect stabilizing point mutations in G-protein coupled receptors. The calculated stability score shows good correlation with the measured thermal stability for 76 single point mutations and 22 multiple mutants in beta1AR. We have demonstrated that conformational sampling of the receptor for various mutants improve the prediction of thermal stability by 50%. Point mutations Y227A5.58, V230A5.61, and F338M7.48 in the thermally stable mutant m23-beta1AR stabilizes key microdomains of the receptor in the inactive conformation. The Y227A5.58 and V230A5.61 mutations stabilize the ionic lock between R139(3.50) on transmembrane helix3 and E285(6.30) on transmembrane helix6. The mutation F338M7.48 on TM7 alters the interaction of the conserved motif NPxxY(x)5,6F with helix8 and hence modulates the interaction of TM2-TM7-helix8 microdomain. The D186-R317 salt bridge (in extracellular loops 2 and 3) is stabilized in the cyanopindolol-bound wild-type beta1AR, whereas the salt bridge between D184-R317 is preferred in the mutant m23. We propose that this could be the surrogate to a similar salt bridge found between the extracellular loop 2 and TM7 in beta2AR reported recently. We show that the binding energy difference between the inactive and active states is less in m23 compared to the wild-type, which explains the activation of m23 at higher norepinephrine concentration compared to the wild-type. Results from this work throw light into the mechanism behind stabilizing mutations. The computational scheme proposed in this work could be used to design stabilizing mutations for other G-protein coupled receptors.
Collapse
Affiliation(s)
- Gouthaman S Balaraman
- Division of Immunology, Beckman Research Institute of the City of Hope, Duarte, California, USA
| | | | | |
Collapse
|
12
|
Barakat NH, Barakat NH, Love JJ. Combined use of experimental and computational screens to characterize protein stability. Protein Eng Des Sel 2010; 23:799-807. [PMID: 20805093 DOI: 10.1093/protein/gzq052] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
One of the primary goals of protein design is to engineer proteins with improved stability. Protein stability is a key issue for chemical, biotechnology and pharmaceutical industries. The development of robust proteins/enzymes with the ability to withstand the potentially harsh conditions of industrial operations is of high importance. A number of strategies are currently being employed to achieve this goal. Two particular approaches, (i) directed evolution and (ii) computational protein design, are quite powerful yet have only recently been combined or applied and analyzed in parallel. In directed evolution, libraries of variants are searched experimentally for clones possessing the desired properties. With computational methods, protein design algorithms are utilized to perform in silico screening for stable protein sequences. Here, we used gene libraries of an unstable variant of streptococcal protein G (Gbeta1) and an in vivo screening method to identify stabilized variants. Many variants with notably increased thermal stabilities were isolated and characterized. Concomitantly, computational techniques and protein design algorithms were used to perform in silico screening of the same destabilized variant of Gbeta1. The combined use, and critical analysis, of these methods promises to advance the field of protein design.
Collapse
Affiliation(s)
- Nora H Barakat
- Department of Chemistry and Biochemistry, San Diego State University, 5500 Campanile Dr, San Diego, CA 92182-1030, USA
| | | | | |
Collapse
|
13
|
Distance-dependent statistical potentials for discriminating thermophilic and mesophilic proteins. Biochem Biophys Res Commun 2010; 396:736-41. [PMID: 20451495 DOI: 10.1016/j.bbrc.2010.05.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Accepted: 05/02/2010] [Indexed: 11/22/2022]
Abstract
Identification of the characteristic structural patterns responsible for protein thermostability is theoretically important and practically useful but largely remains an open problem. These patterns may be revealed through comparative study on thermophilic and mesophilic proteins that have distinct thermostability. In this study, we constructed several distance-dependant potentials from thermophilic and mesophilic proteins. These potentials were then used to evaluate the structural difference between thermophilic and mesophilic proteins. We found that using the subtraction or division of the potentials derived from thermophilic and mesophilic proteins can dramatically increase the discriminatory ability. This approach revealed that the ability to distinct the subtle structural features responsible for protein thermostability may be effectively enhanced through rationally designed comparative study.
Collapse
|
14
|
Solá RJ, Griebenow K. Glycosylation of therapeutic proteins: an effective strategy to optimize efficacy. BioDrugs 2010; 24:9-21. [PMID: 20055529 DOI: 10.2165/11530550-000000000-00000] [Citation(s) in RCA: 323] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
During their development and administration, protein-based drugs routinely display suboptimal therapeutic efficacies due to their poor physicochemical and pharmacological properties. These innate liabilities have driven the development of molecular strategies to improve the therapeutic behavior of protein drugs. Among the currently developed approaches, glycoengineering is one of the most promising, because it has been shown to simultaneously afford improvements in most of the parameters necessary for optimization of in vivo efficacy while allowing for targeting to the desired site of action. These include increased in vitro and in vivo molecular stability (due to reduced oxidation, cross-linking, pH-, chemical-, heating-, and freezing-induced unfolding/denaturation, precipitation, kinetic inactivation, and aggregation), as well as modulated pharmacodynamic responses (due to altered potencies from diminished in vitro enzymatic activities and altered receptor binding affinities) and improved pharmacokinetic profiles (due to altered absorption and distribution behaviors, longer circulation lifetimes, and decreased clearance rates). This article provides an account of the effects that glycosylation has on the therapeutic efficacy of protein drugs and describes the current understanding of the mechanisms by which glycosylation leads to such effects.
Collapse
Affiliation(s)
- Ricardo J Solá
- Laboratory for Applied Biochemistry and Biotechnology, Department of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, Puerto Rico 00931-3346, USA.
| | | |
Collapse
|
15
|
Fu H, Grimsley GR, Razvi A, Scholtz JM, Pace CN. Increasing protein stability by improving beta-turns. Proteins 2010; 77:491-8. [PMID: 19626709 DOI: 10.1002/prot.22509] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Our goal was to gain a better understanding of how protein stability can be increased by improving beta-turns. We studied 22 beta-turns in nine proteins with 66-370 residues by replacing other residues with proline and glycine and measuring the stability. These two residues are statistically preferred in some beta-turn positions. We studied: Cold shock protein B (CspB), Histidine-containing phosphocarrier protein, Ubiquitin, Ribonucleases Sa2, Sa3, T1, and HI, Tryptophan synthetase alpha-subunit, and Maltose binding protein. Of the 15 single proline mutations, 11 increased stability (Average = 0.8 +/- 0.3; Range = 0.3-1.5 kcal/mol), and the stabilizing effect of double proline mutants was additive. On the basis of this and our previous work, we conclude that proteins can generally be stabilized by replacing nonproline residues with proline residues at the i + 1 position of Type I and II beta-turns and at the i position in Type II beta-turns. Other turn positions can sometimes be used if the phi angle is near -60 degrees for the residue replaced. It is important that the side chain of the residue replaced is less than 50% buried. Identical substitutions in beta-turns in related proteins give similar results. Proline substitutions increase stability mainly by decreasing the entropy of the denatured state. In contrast, the large, diverse group of proteins considered here had almost no residues in beta-turns that could be replaced by Gly to increase protein stability. Improving beta-turns by substituting Pro residues is a generally useful way of increasing protein stability.
Collapse
Affiliation(s)
- Hailong Fu
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | | | | | | | | |
Collapse
|
16
|
Li Y, Middaugh CR, Fang J. A novel scoring function for discriminating hyperthermophilic and mesophilic proteins with application to predicting relative thermostability of protein mutants. BMC Bioinformatics 2010; 11:62. [PMID: 20109199 PMCID: PMC3098108 DOI: 10.1186/1471-2105-11-62] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Accepted: 01/28/2010] [Indexed: 11/10/2022] Open
Abstract
Background The ability to design thermostable proteins is theoretically important and practically useful. Robust and accurate algorithms, however, remain elusive. One critical problem is the lack of reliable methods to estimate the relative thermostability of possible mutants. Results We report a novel scoring function for discriminating hyperthermophilic and mesophilic proteins with application to predicting the relative thermostability of protein mutants. The scoring function was developed based on an elaborate analysis of a set of features calculated or predicted from 540 pairs of hyperthermophilic and mesophilic protein ortholog sequences. It was constructed by a linear combination of ten important features identified by a feature ranking procedure based on the random forest classification algorithm. The weights of these features in the scoring function were fitted by a hill-climbing algorithm. This scoring function has shown an excellent ability to discriminate hyperthermophilic from mesophilic sequences. The prediction accuracies reached 98.9% and 97.3% in discriminating orthologous pairs in training and the holdout testing datasets, respectively. Moreover, the scoring function can distinguish non-homologous sequences with an accuracy of 88.4%. Additional blind tests using two datasets of experimentally investigated mutations demonstrated that the scoring function can be used to predict the relative thermostability of proteins and their mutants at very high accuracies (92.9% and 94.4%). We also developed an amino acid substitution preference matrix between mesophilic and hyperthermophilic proteins, which may be useful in designing more thermostable proteins. Conclusions We have presented a novel scoring function which can distinguish not only HP/MP ortholog pairs, but also non-homologous pairs at high accuracies. Most importantly, it can be used to accurately predict the relative stability of proteins and their mutants, as demonstrated in two blind tests. In addition, the residue substitution preference matrix assembled in this study may reflect the thermal adaptation induced substitution biases. A web server implementing the scoring function and the dataset used in this study are freely available at http://www.abl.ku.edu/thermorank/.
Collapse
Affiliation(s)
- Yunqi Li
- Applied Bioinformatics Laboratory, the University of Kansas, Lawrence, KS 66047, USA
| | | | | |
Collapse
|
17
|
Arcangeli C, Cantale C, Galeffi P, Gianese G, Paparcone R, Rosato V. Understanding structural/functional properties of immunoconjugates for cancer therapy by computational approaches. J Biomol Struct Dyn 2008; 26:35-48. [PMID: 18533724 DOI: 10.1080/07391102.2008.10507221] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Monoclonal antibodies coupled to highly toxic molecules (immunoconjugates) are currently being developed for cancer therapy. We have used an in silico procedure for evaluating some physicochemical properties of two tumor-targeting anti-HER2 immunoconjugates: (a) the single-chain antibody scFv(FRP5) linked to a bacterial toxin, that has been recently progressed to phase I clinical trial in human cancer; (b) the putative molecule formed by the intrinsically stable scFv(800E6), which has been proposed as toxin carrier to cancer cells in human therapy, joined to the same toxin of (a). Structural models of the immunoconjugates have been built by homology modeling and assessed by molecular dynamics simulations. The trajectories have been analyzed to extract some biochemical properties and to assess the potential effects of the toxin on the structure and dynamics of the anti-HER2 antibodies. The results of the computational approach indicate that the antibodies maintain their correct folding even in presence of the toxin, whereas a certain stiffness in correspondence of some structural regions is observed. Furthermore, the toxin does not seem to affect the antibody solubility, whereas it enhances the structural stability. The proposed computational approach represent a promising tool for analyzing some physicochemical properties of immunoconjugates and for predicting the effects of the linked toxin on structure, dynamics, and functionality of the antibodies.
Collapse
Affiliation(s)
- C Arcangeli
- Computing and Modeling Unit, ENEA Casaccia Research Center, Via Anguillarese 301, 00123 S.Maria di Galeria, Italy.
| | | | | | | | | | | |
Collapse
|
18
|
Abstract
MOTIVATION The task of engineering a protein to perform a target biological function is known as protein design. A commonly used paradigm casts this functional design problem as a structural one, assuming a fixed backbone. In probabilistic protein design, positional amino acid probabilities are used to create a random library of sequences to be simultaneously screened for biological activity. Clearly, certain choices of probability distributions will be more successful in yielding functional sequences. However, since the number of sequences is exponential in protein length, computational optimization of the distribution is difficult. RESULTS In this paper, we develop a computational framework for probabilistic protein design following the structural paradigm. We formulate the distribution of sequences for a structure using the Boltzmann distribution over their free energies. The corresponding probabilistic graphical model is constructed, and we apply belief propagation (BP) to calculate marginal amino acid probabilities. We test this method on a large structural dataset and demonstrate the superiority of BP over previous methods. Nevertheless, since the results obtained by BP are far from optimal, we thoroughly assess the paradigm using high-quality experimental data. We demonstrate that, for small scale sub-problems, BP attains identical results to those produced by exact inference on the paradigmatic model. However, quantitative analysis shows that the distributions predicted significantly differ from the experimental data. These findings, along with the excellent performance we observed using BP on the smaller problems, suggest potential shortcomings of the paradigm. We conclude with a discussion of how it may be improved in the future.
Collapse
Affiliation(s)
- Menachem Fromer
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Israel.
| | | |
Collapse
|
19
|
Fung HK, Welsh WJ, Floudas CA. Computational De Novo Peptide and Protein Design: Rigid Templates versus Flexible Templates. Ind Eng Chem Res 2008. [DOI: 10.1021/ie071286k] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Ho Ki Fung
- Department of Chemical Engineering, Princeton University, Princeton, New Jersey 08544-5263, and Department of Pharmacology, University of Medicine & Dentistry of New Jersey (UMDNJ), Robert Wood Johnson Medical School, and the Informatics Institute of UMDNJ, Piscataway, New Jersey 08854
| | - William J. Welsh
- Department of Chemical Engineering, Princeton University, Princeton, New Jersey 08544-5263, and Department of Pharmacology, University of Medicine & Dentistry of New Jersey (UMDNJ), Robert Wood Johnson Medical School, and the Informatics Institute of UMDNJ, Piscataway, New Jersey 08854
| | - Christodoulos A. Floudas
- Department of Chemical Engineering, Princeton University, Princeton, New Jersey 08544-5263, and Department of Pharmacology, University of Medicine & Dentistry of New Jersey (UMDNJ), Robert Wood Johnson Medical School, and the Informatics Institute of UMDNJ, Piscataway, New Jersey 08854
| |
Collapse
|
20
|
Ytreberg FM, Zuckerman DM. Peptide conformational equilibria computed via a single-stage shifting protocol. J Phys Chem B 2007; 109:9096-103. [PMID: 16852082 DOI: 10.1021/jp0510692] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We study the conformational equilibria of two peptides using a novel statistical mechanics approach designed for calculating free energy differences between highly dissimilar conformational states. Our results elucidate the contrasting roles of entropy in implicitly solvated leucine dipeptide and decaglycine. The method extends earlier work by Voter and overcomes the notorious "overlap" problem in free energy computations by constructing a mathematically equivalent calculation with high conformational similarity. The approach requires only equilibrium simulations of the two states of interest, without the need for sampling transition states. We discuss possible extensions and optimizations of the approach.
Collapse
Affiliation(s)
- F Marty Ytreberg
- Department of Computational Biology and the Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, Pennsylvania 15261, USA.
| | | |
Collapse
|
21
|
Kampmann T, Mueller DS, Mark AE, Young PR, Kobe B. The Role of histidine residues in low-pH-mediated viral membrane fusion. Structure 2007; 14:1481-7. [PMID: 17027497 DOI: 10.1016/j.str.2006.07.011] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2006] [Revised: 07/18/2006] [Accepted: 07/23/2006] [Indexed: 11/20/2022]
Abstract
A central event in the invasion of a host cell by an enveloped virus is the fusion of viral and cell membranes. For many viruses, membrane fusion is driven by specific viral surface proteins that undergo large-scale conformational rearrangements, triggered by exposure to low pH in the endosome upon internalization. Here, we present evidence suggesting that in both class I (helical hairpin proteins) and class II (beta-structure-rich proteins) pH-dependent fusion proteins the protonation of specific histidine residues triggers fusion via an analogous molecular mechanism. These histidines are located in the vicinity of positively charged residues in the prefusion conformation, and they subsequently form salt bridges with negatively charged residues in the postfusion conformation. The molecular surfaces involved in the corresponding structural rearrangements leading to fusion are highly conserved and thus might provide a suitable common target for the design of antivirals, which could be active against a diverse range of pathogenic viruses.
Collapse
Affiliation(s)
- Thorsten Kampmann
- School of Molecular and Microbial Sciences, University of Queensland, Brisbane, Queensland 4072, Australia
| | | | | | | | | |
Collapse
|
22
|
Matheus S, Mahler HC, Friess W. A Critical Evaluation of Tm(FTIR) Measurements of High-Concentration IgG1 Antibody Formulations as a Formulation Development Tool. Pharm Res 2006; 23:1617-27. [PMID: 16783474 DOI: 10.1007/s11095-006-0283-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2005] [Accepted: 02/28/2006] [Indexed: 12/12/2022]
Abstract
PURPOSE Fourier-transform infrared (FTIR) spectroscopy was applied for the determination of protein melting temperature (Tm(FTIR)) and to assess the stability predictability of a 100-mg/mL liquid IgG1 antibody formulation. METHODS Tm(FTIR) values of various formulations (different pH, buffers, excipients) were compared to the results of a stability study under accelerated conditions (40 degrees C/75% relative humidity), using size-exclusion high-performance liquid chromatography (SE-HPLC) and sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) for the detection of soluble aggregates and covalent modifications. RESULTS The highest Tm(FTIR) was achieved at pH 5.5, and, similarly, SE-HPLC and SDS-PAGE results suggested a pH optimum between 5.5 and 6.0. Transition temperatures were comparable for all tested buffers. However, the decrease in the monomer fraction upon thermal storage was the lowest for citrate buffers. Whereas sugars and polyols resulted in an increase in Tm(FTIR) and enhanced monomer fraction after storage, amino acids showed a destabilization according to SE-HPLC analysis, albeit no change or even an increase in the melting temperature was observed. CONCLUSIONS All examples gave evidence that Tm(FTIR) values did not necessarily correspond to the storage stability at 40 degrees C analyzed by means of SE-HPLC and SDS-PAGE. Tm values, e.g., determined by FTIR, should only be employed as supportive information to the results from both real-time and accelerated stability studies.
Collapse
Affiliation(s)
- Susanne Matheus
- Merck KGaA, Global Pharmaceutical Development, Darmstadt, Germany
| | | | | |
Collapse
|
23
|
Grigoryan G, Zhou F, Lustig SR, Ceder G, Morgan D, Keating AE. Ultra-fast evaluation of protein energies directly from sequence. PLoS Comput Biol 2006; 2:e63. [PMID: 16789811 PMCID: PMC1479088 DOI: 10.1371/journal.pcbi.0020063] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2006] [Accepted: 04/24/2006] [Indexed: 11/22/2022] Open
Abstract
The structure, function, stability, and many other properties of a protein in a fixed environment are fully specified by its sequence, but in a manner that is difficult to discern. We present a general approach for rapidly mapping sequences directly to their energies on a pre-specified rigid backbone, an important sub-problem in computational protein design and in some methods for protein structure prediction. The cluster expansion (CE) method that we employ can, in principle, be extended to model any computable or measurable protein property directly as a function of sequence. Here we show how CE can be applied to the problem of computational protein design, and use it to derive excellent approximations of physical potentials. The approach provides several attractive advantages. First, following a one-time derivation of a CE expansion, the amount of time necessary to evaluate the energy of a sequence adopting a specified backbone conformation is reduced by a factor of 107 compared to standard full-atom methods for the same task. Second, the agreement between two full-atom methods that we tested and their CE sequence-based expressions is very high (root mean square deviation 1.1–4.7 kcal/mol, R2 = 0.7–1.0). Third, the functional form of the CE energy expression is such that individual terms of the expansion have clear physical interpretations. We derived expressions for the energies of three classic protein design targets—a coiled coil, a zinc finger, and a WW domain—as functions of sequence, and examined the most significant terms. Single-residue and residue-pair interactions are sufficient to accurately capture the energetics of the dimeric coiled coil, whereas higher-order contributions are important for the two more globular folds. For the task of designing novel zinc-finger sequences, a CE-derived energy function provides significantly better solutions than a standard design protocol, in comparable computation time. Given these advantages, CE is likely to find many uses in computational structural modeling. Many applications in computational structural biology involve evaluating the energy of a protein adopting a specific structure. A variety of functions are used for this purpose. Statistical potentials are fast to evaluate but do not have a clear biophysical basis, whereas physics-based functions consist of well-defined terms that can be costly to compute. This paper describes how the theory of cluster expansion, originally developed to describe the energies of alloys, can be applied to generate a physical potential for proteins that is extremely fast to evaluate. Cluster expansion is a way of representing a property of a system as a discrete function of its degrees of freedom. In this paper, it is used for the problem of protein design, where the energy is determined by the identities and conformations of amino acids at different sites on a fixed protein backbone. Application of cluster expansion to three small protein folds—the α-helical coiled coil, the zinc finger, and the WW domain—shows that protein sequence can be mapped directly to energy using a surprisingly simple function that maintains high accuracy. Promising results on these small systems suggest that the theory may have utility for macromolecular modeling more generally.
Collapse
Affiliation(s)
- Gevorg Grigoryan
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Fei Zhou
- Department of Physics, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Steve R Lustig
- DuPont Central Research and Development, Experimental Station, Wilmington, Delaware, United States of America
| | - Gerbrand Ceder
- Department of Material Science and Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Dane Morgan
- Department of Material Science and Engineering, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Amy E Keating
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
24
|
Song G, Lazar GA, Kortemme T, Shimaoka M, Desjarlais JR, Baker D, Springer TA. Rational design of intercellular adhesion molecule-1 (ICAM-1) variants for antagonizing integrin lymphocyte function-associated antigen-1-dependent adhesion. J Biol Chem 2005; 281:5042-9. [PMID: 16354667 PMCID: PMC1455478 DOI: 10.1074/jbc.m510454200] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The interaction between integrin lymphocyte function-associated antigen-1 (LFA-1) and its ligand intercellular adhesion molecule-1 (ICAM-1) is critical in immunological and inflammatory reactions but, like other adhesive interactions, is of low affinity. Here, multiple rational design methods were used to engineer ICAM-1 mutants with enhanced affinity for LFA-1. Five amino acid substitutions 1) enhance the hydrophobicity and packing of residues surrounding Glu-34 of ICAM-1, which coordinates to a Mg2+ in the LFA-1 I domain, and 2) alter associations at the edges of the binding interface. The affinity of the most improved ICAM-1 mutant for intermediate- and high-affinity LFA-1 I domains was increased by 19-fold and 22-fold, respectively, relative to wild type. Moreover, potency was similarly enhanced for inhibition of LFA-1-dependent ligand binding and cell adhesion. Thus, rational design can be used to engineer novel adhesion molecules with high monomeric affinity; furthermore, the ICAM-1 mutant holds promise for targeting LFA-1-ICAM-1 interaction for biological studies and therapeutic purposes.
Collapse
Affiliation(s)
- Gang Song
- From the CBR Institute for Biomedical Research, and
| | | | - Tanja Kortemme
- Howard Hughes Medical Institute and Department of Biochemistry, University of Washington, Seattle, Washington 98195
| | - Motomu Shimaoka
- Departments of Pathology and Anesthesia, Harvard Medical School, Boston, Massachusetts 02115
| | | | - David Baker
- Howard Hughes Medical Institute and Department of Biochemistry, University of Washington, Seattle, Washington 98195
| | - Timothy A. Springer
- From the CBR Institute for Biomedical Research, and
- To whom correspondence should be addressed: CBR Institute for Biochemical Research, 200 Longwood Ave., Boston, MA 02115. Tel.: 617-278-3225; Fax: 617-278-3232; E-mail:
| |
Collapse
|
25
|
Zakrzewska M, Krowarsch D, Wiedlocha A, Olsnes S, Otlewski J. Highly stable mutants of human fibroblast growth factor-1 exhibit prolonged biological action. J Mol Biol 2005; 352:860-75. [PMID: 16126225 DOI: 10.1016/j.jmb.2005.07.066] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2005] [Revised: 07/21/2005] [Accepted: 07/27/2005] [Indexed: 11/29/2022]
Abstract
Fibroblast growth factor 1 (FGF-1) shows strong angiogenic, osteogenic and tissue-injury repair properties that might be relevant to medical applications. Since FGF-1 is partially unfolded at physiological temperature we decided to increase significantly its conformational stability and test how such an improvement will affect its biological function. Using an homology approach and rational strategy we designed two new single FGF-1 mutations: Q40P and S47I that appeared to be the most strongly stabilizing substitutions among those reported so far, increasing the denaturation temperature by 7.8 deg. C and 9.0 deg. C, respectively. As our goal was to produce highly stable variants of the growth factor, we combined these two mutations with five previously described stabilizing substitutions. The multiple mutants showed denaturation temperatures up to 27 deg. C higher than the wild-type and exhibited full additivity of the mutational effects. All those mutants were biologically competent in several cell culture assays, maintaining typical FGF-1 activities, such as binding to specific cell surface receptors and activation of downstream signaling pathways. Thus, we demonstrate that the low denaturation temperature of wild-type FGF-1 is not related to its fundamental cellular functions, and that FGF-1 action is not affected by its stability. A more detailed analysis of the biological behavior of stable FGF-1 mutants revealed that, compared with the wild-type, their mitogenic properties, as probed by the DNA synthesis assay, were significantly increased in the absence of heparin, and that their half-lives were extensively prolonged. We found that the biological action of the mutants was dictated by their susceptibility to proteases, which strongly correlated with the stability. Mutants which were much more resistant to proteolytic degradation always displayed a significant improvement in the half-life and mitogenesis. Our results show that engineered stable growth factor variants exhibit enhanced and prolonged activity, which can be advantageous in terms of the potential therapeutic applications of FGF-1.
Collapse
Affiliation(s)
- Malgorzata Zakrzewska
- Protein Engineering Laboratory, Institute of Biochemistry and Molecular Biology, University of Wroclaw, Tamka 2, 50-137 Wroclaw, Poland
| | | | | | | | | |
Collapse
|
26
|
Wittrup KD. Directed evolution in chemical engineering. AIChE J 2005. [DOI: 10.1002/aic.10706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
27
|
Vielmetter J, Tishler J, Ary ML, Cheung P, Bishop R. Data management solutions for protein therapeutic research and development. Drug Discov Today 2005; 10:1065-71. [PMID: 16055023 DOI: 10.1016/s1359-6446(05)03495-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Protein therapeutics, including monoclonal antibodies, are a growing focus of drug discovery research organizations. High-throughput screening of large libraries of protein variants is therefore becoming increasingly important in R&D. As a result, there is a need to link large numbers of variant protein sequences with chemical and biological assay data. This integration will allow more efficient data mining and facilitate decision-making regarding hit identification, lead optimization and drug development. In this paper, we present an implementation in which a widely used small-molecule high-throughput screening data management system has been adapted to meet the unique needs of protein drug discovery and development.
Collapse
|
28
|
Rao BM, Lauffenburger DA, Wittrup KD. Integrating cell-level kinetic modeling into the design of engineered protein therapeutics. Nat Biotechnol 2005; 23:191-4. [PMID: 15696150 DOI: 10.1038/nbt1064] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Functional genomics and proteomics are identifying many potential drug targets for novel therapeutic proteins, and both rational and combinatorial protein engineering methods are available for creating drug candidates. A central challenge is the definition of the most appropriate design criteria, which will benefit critically from computational kinetic models that incorporate integration from the molecular level to the whole systems level. Interpretation of these processes will require mathematical models that are refined in combination with relevant data derived from quantitative assays, to correctly set biophysical objectives for protein design.
Collapse
Affiliation(s)
- Balaji M Rao
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | |
Collapse
|
29
|
Jaramillo A, Wodak SJ. Computational protein design is a challenge for implicit solvation models. Biophys J 2005; 88:156-71. [PMID: 15377512 PMCID: PMC1304995 DOI: 10.1529/biophysj.104.042044] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2004] [Accepted: 09/07/2004] [Indexed: 11/18/2022] Open
Abstract
Increasingly complex schemes for representing solvent effects in an implicit fashion are being used in computational analyses of biological macromolecules. These schemes speed up the calculations by orders of magnitude and are assumed to compromise little on essential features of the solvation phenomenon. In this work we examine this assumption. Five implicit solvation models, a surface area-based empirical model, two models that approximate the generalized Born treatment and a finite difference Poisson-Boltzmann method are challenged in situations differing from those where these models were calibrated. These situations are encountered in automatic protein design procedures, whose job is to select sequences, which stabilize a given protein 3D structure, from a large number of alternatives. To this end we evaluate the energetic cost of burying amino acids in thousands of environments with different solvent exposures belonging, respectively, to decoys built with random sequences and to native protein crystal structures. In addition we perform actual sequence design calculations. Except for the crudest surface area-based procedure, all the tested models tend to favor the burial of polar amino acids in the protein interior over nonpolar ones, a behavior that leads to poor performance in protein design calculations. We show, on the other hand, that three of the examined models are nonetheless capable of discriminating between the native fold and many nonnative alternatives, a test commonly used to validate force fields. It is concluded that protein design is a particularly challenging test for implicit solvation models because it requires accurate estimates of the solvation contribution of individual residues. This contrasts with native recognition, which depends less on solvation and more on other nonbonded contributions.
Collapse
Affiliation(s)
- Alfonso Jaramillo
- Service de Conformation de Macromolécules Biologiques et Bioinformatique, CP263 Université Libre de Bruxelles, Brussels, Belgium
| | | |
Collapse
|
30
|
Abstract
We consider highly specific protein-protein interactions in proteomes of simple model proteins. We are inspired by the work of Zarrinpar et al (2003 Nature 426 676). They took a binding domain in a signalling pathway in yeast and replaced it with domains of the same class but from different organisms. They found that the probability of a protein binding to a protein from the proteome of a different organism is rather high, around one half. We calculate the probability of a model protein from one proteome binding to the protein of a different proteome. These proteomes are obtained by sampling the space of functional proteomes uniformly. In agreement with Zarrinpar et al we find that the probability of a protein binding a protein from another proteome is rather high, of order one tenth. Our results, together with those of Zarrinpar et al, suggest that designing, say, a peptide to block or reconstitute a single signalling pathway, without affecting any other pathways, requires knowledge of all the partners of the class of binding domains the peptide is designed to mimic. This knowledge is required to use negative design to explicitly design out interactions of the peptide with proteins other than its target. We also found that patches that are required to bind with high specificity evolve more slowly than those that are required only to not bind to any other patch. This is consistent with some analysis of sequence data for proteins engaged in highly specific interactions.
Collapse
Affiliation(s)
- Richard P Sear
- The Isaac Newton Institute for Mathematical Sciences, University of Cambridge, 20 Clarkson Road, Cambridge CB3 0EH, UK.
| |
Collapse
|
31
|
Bunick CG, Nelson MR, Mangahas S, Hunter MJ, Sheehan JH, Mizoue LS, Bunick GJ, Chazin WJ. Designing sequence to control protein function in an EF-hand protein. J Am Chem Soc 2004; 126:5990-8. [PMID: 15137763 DOI: 10.1021/ja0397456] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The extent of conformational change that calcium binding induces in EF-hand proteins is a key biochemical property specifying Ca(2+) sensor versus signal modulator function. To understand how differences in amino acid sequence lead to differences in the response to Ca(2+) binding, comparative analyses of sequence and structures, combined with model building, were used to develop hypotheses about which amino acid residues control Ca(2+)-induced conformational changes. These results were used to generate a first design of calbindomodulin (CBM-1), a calbindin D(9k) re-engineered with 15 mutations to respond to Ca(2+) binding with a conformational change similar to that of calmodulin. The gene for CBM-1 was synthesized, and the protein was expressed and purified. Remarkably, this protein did not exhibit any non-native-like molten globule properties despite the large number of mutations and the nonconservative nature of some of them. Ca(2+)-induced changes in CD intensity and in the binding of the hydrophobic probe, ANS, implied that CBM-1 does undergo Ca(2+) sensorlike conformational changes. The X-ray crystal structure of Ca(2+)-CBM-1 determined at 1.44 A resolution reveals the anticipated increase in hydrophobic surface area relative to the wild-type protein. A nascent calmodulin-like hydrophobic docking surface was also found, though it is occluded by the inter-EF-hand loop. The results from this first calbindomodulin design are discussed in terms of progress toward understanding the relationships between amino acid sequence, protein structure, and protein function for EF-hand CaBPs, as well as the additional mutations for the next CBM design.
Collapse
Affiliation(s)
- Christopher G Bunick
- Department of Biochemistry, Center for Structural Biology, Vanderbilt University, 5140 BIOSCI/MRB III, Nashville, Tennessee 37232-8725, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Allert M, Rizk SS, Looger LL, Hellinga HW. Computational design of receptors for an organophosphate surrogate of the nerve agent soman. Proc Natl Acad Sci U S A 2004; 101:7907-12. [PMID: 15148405 PMCID: PMC419530 DOI: 10.1073/pnas.0401309101] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We report the computational design of soluble protein receptors for pinacolyl methyl phosphonic acid (PMPA), the predominant hydrolytic product of the nerve agent soman. Using recently developed computational protein design techniques, the ligand-binding pockets of two periplasmic binding proteins, glucose-binding protein and ribose-binding protein, were converted to bind PMPA instead of their cognate sugars. The designs introduce 9-12 mutations in the parent proteins. Twelve of 20 designs tested exhibited PMPA-dependent changes in emission intensity of a fluorescent reporter with affinities between 45 nM and 10 microM. The contributions to ligand binding by individual residues were determined in two designs by alanine-scanning mutagenesis, and are consistent with the molecular models. These results demonstrate that designed receptors with radically altered binding specificities and affinities that rival or exceed those of the parent proteins can be successfully predicted. The designs vary in parent scaffold, sequence diversity, and orientation of docked ligand, suggesting that the number of possible solutions to the design problem is large and degenerate. This observation has implications for the genesis of biological function by random processes. The designed receptors reported here may have utility in the development of fluorescent biosensors for monitoring nerve agents.
Collapse
Affiliation(s)
- Malin Allert
- Departments of Biochemistry and Pharmacology and Molecular Cancer Biology, Box 3711, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
33
|
Abstract
Globular proteins are characterized by the specific and tight packing of hydrophobic side-chains in the so-called "hydrophobic core." Formation of the core is key in folding, stabilization, and conformational specificity. The critical role of hydrophobic cores in maintaining the highly ordered structures present in natural proteins justifies the tremendous efforts devoted to their redesign. Both experimental and computational combinatorial-based approaches have been reported in the last years as powerful protein design tools. These manage to explore large regions of the sequence/conformational space, allowing the search for alternative protein core arrangements displaying native-like properties. The overall results obtained from core design projects have contributed significantly to our present knowledge of protein folding and function. In addition, core design has worked as a benchmark for the development of ambitious protein design projects that nowadays are allowing the de novo design of novel protein structures and functions.
Collapse
Affiliation(s)
- Salvador Ventura
- Institut de Biotecnologia i de Biomedicina and Departament de Bioquimica i Biologia Molecular, Universitat Autonoma de Barcelona, Barcelona, Spain.
| | | |
Collapse
|
34
|
Ytreberg FM, Zuckerman DM. Efficient use of nonequilibrium measurement to estimate free energy differences for molecular systems. J Comput Chem 2004; 25:1749-59. [PMID: 15362132 DOI: 10.1002/jcc.20103] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A promising method for calculating free energy differences DeltaF is to generate nonequilibrium data via "fast-growth" simulations or by experiments--and then use Jarzynski's equality. However, a difficulty with using Jarzynski's equality is that DeltaF estimates converge very slowly and unreliably due to the nonlinear nature of the calculation--thus requiring large, costly data sets. The purpose of the work presented here is to determine the best estimate for DeltaF given a (finite) set of work values previously generated by simulation or experiment. Exploiting statistical properties of Jarzynski's equality, we present two fully automated analyses of nonequilibrium data from a toy model, and various simulated molecular systems. Both schemes remove at least several k(B)T of bias from DeltaF estimates, compared to direct application of Jarzynski's equality, for modest sized data sets (100 work values), in all tested systems. Results from one of the new methods suggest that good estimates of DeltaF can be obtained using 5-40-fold less data than was previously possible. Extending previous work, the new results exploit the systematic behavior of bias due to finite sample size. A key innovation is better use of the more statistically reliable information available from the raw data.
Collapse
Affiliation(s)
- F Marty Ytreberg
- Center for Computational Biology and Bioinformatics, University of Pittsburgh, 200 Lothrop St., Pittsburgh, Pennsylvania 15261, USA.
| | | |
Collapse
|