153 results on '"computational structural biology"'
Search Results
2. Modeling and Analysis of HIV-1 Pol Polyprotein as a Case Study for Predicting Large Polyprotein Structures.
- Author
-
Hao, Ming, Imamichi, Tomozumi, and Chang, Weizhong
- Subjects
- *
REVERSE transcriptase , *AIDS , *RIBONUCLEASE H , *HIV , *LIFE cycles (Biology) , *TERTIARY structure , *PROTEIN structure - Abstract
Acquired immunodeficiency syndrome (AIDS) is caused by human immunodeficiency virus (HIV). HIV protease, reverse transcriptase, and integrase are targets of current drugs to treat the disease. However, anti-viral drug-resistant strains have emerged quickly due to the high mutation rate of the virus, leading to the demand for the development of new drugs. One attractive target is Gag-Pol polyprotein, which plays a key role in the life cycle of HIV. Recently, we found that a combination of M50I and V151I mutations in HIV-1 integrase can suppress virus release and inhibit the initiation of Gag-Pol autoprocessing and maturation without interfering with the dimerization of Gag-Pol. Additional mutations in integrase or RNase H domain in reverse transcriptase can compensate for the defect. However, the molecular mechanism is unknown. There is no tertiary structure of the full-length HIV-1 Pol protein available for further study. Therefore, we developed a workflow to predict the tertiary structure of HIV-1 NL4.3 Pol polyprotein. The modeled structure has comparable quality compared with the recently published partial HIV-1 Pol structure (PDB ID: 7SJX). Our HIV-1 NL4.3 Pol dimer model is the first full-length Pol tertiary structure. It can provide a structural platform for studying the autoprocessing mechanism of HIV-1 Pol and for developing new potent drugs. Moreover, the workflow can be used to predict other large protein structures that cannot be resolved via conventional experimental methods. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Hydrophobic gating and functional annotation of ion channels
- Author
-
Klesse, Gianni, Tucker, Stephen J., and Sansom, Mark S. P.
- Subjects
571.6 ,Computational Biochemistry ,Computational Structural Biology ,Computational Biophysics - Abstract
Ion channels are integral membrane proteins that facilitate the permeation of ions through lipid bilayers. By means of a molecular mechanism known as hydrophobic gating, these proteins can close their conduction pathway to ion flow without the need for steric occlusion. Hydrophobic gates pose a challenge in the analysis of novel ion channel structures, as they cannot be detected by measuring the physical dimensions of a channel pore. This thesis therefore presents the Channel Annotation Package (CHAP), an open-source software tool for the functional annotation of ion channel structures based on molecular dynamics (MD) simulation and capable of reliably indentifying hydrophobic gates. The cation-selective 5-HT
3 receptor and the anion-selective glycine receptor served as test cases to demonstrate the utility of CHAP. The functional status of several recent structures of these two channels was determined through MD simulation and hydrophobic gates were identified in both receptors. Furthermore, a high-throughput survey of ion channel structures enabled by CHAP revealed that hydrophobic gates occur in ~20% of the ion channel structural proteome. Next-generation polarisable force fields were used to explore the importance of electronic polarisation in MD simulations of ion channels. Water molecules passing a hydrophobic gate experienced a significant reduction of their dipole moment, mirroring the behaviour of water in a liquid-vapour phase transition. Polarisable ions interacted more strongly with uncharged protein residues than simulations employing traditional additive force fields suggest. Incorporating polarisation effects will thus be an important aspect of future computational studies of ion channel structure and function. The impact of a transmembrane potential on the hydrophobic gate in the 5-HT3 receptor was studied through atomistic MD simulations. Electrowetting occured only at supraphysiological voltages, while at lower field strengths pore de-wetting effectively prevented ion permeation. The hydration response of the channel could be quantitatively described by a thermodynamic model that includes the influence of the charged amino acid residues of the channel protein. Electric field effects are unlikely to play an important role for hydrophobic gating in biological ion channels, but may potentially be exploited in the design of gated artificial nanopores.- Published
- 2020
4. De novo protein design by inversion of the AlphaFold structure prediction network.
- Author
-
Goverde, Casper A., Wolf, Benedict, Khakzad, Hamed, Rosset, Stéphane, and Correia, Bruno E.
- Abstract
De novo protein design enhances our understanding of the principles that govern protein folding and interactions, and has the potential to revolutionize biotechnology through the engineering of novel protein functionalities. Despite recent progress in computational design strategies, de novo design of protein structures remains challenging, given the vast size of the sequence‐structure space. AlphaFold2 (AF2), a state‐of‐the‐art neural network architecture, achieved remarkable accuracy in predicting protein structures from amino acid sequences. This raises the question whether AF2 has learned the principles of protein folding sufficiently for de novo design. Here, we sought to answer this question by inverting the AF2 network, using the prediction weight set and a loss function to bias the generated sequences to adopt a target fold. Initial design trials resulted in de novo designs with an overrepresentation of hydrophobic residues on the protein surface compared to their natural protein family, requiring additional surface optimization. In silico validation of the designs showed protein structures with the correct fold, a hydrophilic surface and a densely packed hydrophobic core. In vitro validation showed that 7 out of 39 designs were folded and stable in solution with high melting temperatures. In summary, our design workflow solely based on AF2 does not seem to fully capture basic principles of de novo protein design, as observed in the protein surface's hydrophobic vs. hydrophilic patterning. However, with minimal post‐design intervention, these pipelines generated viable sequences as assessed experimental characterization. Thus, such pipelines show the potential to contribute to solving outstanding challenges in de novo protein design. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
5. Pharmacological Chaperones and Protein Conformational Diseases: Approaches of Computational Structural Biology.
- Author
-
Grasso, Daniela, Galderisi, Silvia, Santucci, Annalisa, and Bernini, Andrea
- Subjects
- *
COMPUTATIONAL biology , *SMALL molecules , *PROTEIN folding , *PROTEIN stability , *DRUG repositioning - Abstract
Whenever a protein fails to fold into its native structure, a profound detrimental effect is likely to occur, and a disease is often developed. Protein conformational disorders arise when proteins adopt abnormal conformations due to a pathological gene variant that turns into gain/loss of function or improper localization/degradation. Pharmacological chaperones are small molecules restoring the correct folding of a protein suitable for treating conformational diseases. Small molecules like these bind poorly folded proteins similarly to physiological chaperones, bridging non-covalent interactions (hydrogen bonds, electrostatic interactions, and van der Waals contacts) loosened or lost due to mutations. Pharmacological chaperone development involves, among other things, structural biology investigation of the target protein and its misfolding and refolding. Such research can take advantage of computational methods at many stages. Here, we present an up-to-date review of the computational structural biology tools and approaches regarding protein stability evaluation, binding pocket discovery and druggability, drug repurposing, and virtual ligand screening. The tools are presented as organized in an ideal workflow oriented at pharmacological chaperones' rational design, also with the treatment of rare diseases in mind. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. IRAA: A statistical tool for investigating a protein–protein interaction interface from multiple structures.
- Author
-
Belapure, Jaydeep, Sorokina, Marija, and Kastritis, Panagiotis L.
- Abstract
Understanding protein–protein interactions (PPIs) is fundamental to infer how different molecular systems work. A major component to model molecular recognition is the buried surface area (BSA), that is, the area that becomes inaccessible to solvent upon complex formation. To date, many attempts tried to connect BSA to molecular recognition principles, and in particular, to the underlying binding affinity. However, the most popular approach to calculate BSA is to use a single (or in some cases few) bound structures, consequently neglecting a wealth of structural information of the interacting proteins derived from ensembles corresponding to their unbound and bound states. Moreover, the most popular method inherently assumes the component proteins to bind as rigid entities. To address the above shortcomings, we developed a Monte Carlo method‐based Interface Residue Assessment Algorithm (IRAA), to calculate a combined distribution of BSA for a given complex. Further, we apply our algorithm to human ACE2 and SARS‐CoV‐2 Spike protein complex, a system of prime importance. Results show a much broader distribution of BSA compared to that obtained from only the bound structure or structures and extended residue members of the interface with implications to the underlying biomolecular recognition. We derive that specific interface residues of ACE2 and of S‐protein are consistently highly flexible, whereas other residues systematically show minor conformational variations. In effect, IRAA facilitates the use of all available structural data for any biomolecular complex of interest, extracting quantitative parameters with statistical significance, thereby providing a deeper biophysical understanding of the molecular system under investigation. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
7. Elucidating the Structural Impacts of Protein InDels.
- Author
-
Jilani, Muneeba, Turcan, Alistair, Haspel, Nurit, and Jagodzinski, Filip
- Subjects
- *
CYTOSKELETAL proteins , *AMINO acids , *STATISTICAL correlation , *COMPUTATIONAL biology - Abstract
The effects of amino acid insertions and deletions (InDels) remain a rather under-explored area of structural biology. These variations oftentimes are the cause of numerous disease phenotypes. In spite of this, research to study InDels and their structural significance remains limited, primarily due to a lack of experimental information and computational methods. In this work, we fill this gap by modeling InDels computationally; we investigate the rigidity differences between the wildtype and a mutant variant with one or more InDels. Further, we compare how structural effects due to InDels differ from the effects of amino acid substitutions, which are another type of amino acid mutation. We finish by performing a correlation analysis between our rigidity-based metrics and wet lab data for their ability to infer the effects of InDels on protein fitness. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
8. Overlaying a hypergraph with a graph with bounded maximum degree.
- Author
-
Havet, Frédéric, Mazauric, Dorian, Nguyen, Viet-Ha, and Watrigant, Rémi
- Subjects
- *
HYPERGRAPHS , *COMPUTATIONAL complexity , *POLYNOMIALS , *INTEGERS , *COMPUTATIONAL biology - Abstract
Let G and H be respectively a graph and a hypergraph defined on a same set of vertices, and let F be a fixed graph. We say that G F - overlays a hyperedge S of H if F is a spanning subgraph of the subgraph of G induced by S , and that G F - overlays H if it F -overlays every hyperedge of H. Motivated by structural biology, we study the computational complexity of two problems. The first problem, (Δ ≤ k) F - Overlay , consists in deciding whether there is a graph with maximum degree at most k that F -overlays a given hypergraph H. It is a particular case of the second problem Max (Δ ≤ k) F - Overlay , which takes a hypergraph H and an integer s as input, and consists in deciding whether there is a graph with maximum degree at most k that F -overlays at least s hyperedges of H. We give a complete polynomial/ NP -complete dichotomy for the Max (Δ ≤ k) − F - Overlay problems depending on the pairs (F , k) , and establish the complexity of (Δ ≤ k) F - Overlay for many pairs (F , k). [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
9. PANDORA: A Fast, Anchor-Restrained Modelling Protocol for Peptide: MHC Complexes.
- Author
-
Marzella, Dario F., Parizi, Farzaneh M., Tilborg, Derek van, Renaud, Nicolas, Sybrandi, Daan, Buzatu, Rafaella, Rademaker, Daniel T., 't Hoen, Peter A. C., and Xue, Li C.
- Subjects
IMMUNOTHERAPY ,T cell receptors ,PEPTIDES ,MAJOR histocompatibility complex ,PROTEOMICS ,MACHINE learning ,DEEP learning - Abstract
Deeper understanding of T-cell-mediated adaptive immune responses is important for the design of cancer immunotherapies and antiviral vaccines against pandemic outbreaks. T-cells are activated when they recognize foreign peptides that are presented on the cell surface by Major Histocompatibility Complexes (MHC), forming peptide:MHC (pMHC) complexes. 3D structures of pMHC complexes provide fundamental insight into T-cell recognition mechanism and aids immunotherapy design. High MHC and peptide diversities necessitate efficient computational modelling to enable whole proteome structural analysis. We developed PANDORA, a generic modelling pipeline for pMHC class I and II (pMHC-I and pMHC-II), and present its performance on pMHC-I here. Given a query, PANDORA searches for structural templates in its extensive database and then applies anchor restraints to the modelling process. This restrained energy minimization ensures one of the fastest pMHC modelling pipelines so far. On a set of 835 pMHC-I complexes over 78 MHC types, PANDORA generated models with a median RMSD of 0.70 Å and achieved a 93% success rate in top 10 models. PANDORA performs competitively with three pMHC-I modelling state-of-the-art approaches and outperforms AlphaFold2 in terms of accuracy while being superior to it in speed. PANDORA is a modularized and user-configurable python package with easy installation. We envision PANDORA to fuel deep learning algorithms with large-scale high-quality 3D models to tackle long-standing immunology challenges. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
10. PANDORA: A Fast, Anchor-Restrained Modelling Protocol for Peptide: MHC Complexes
- Author
-
Dario F. Marzella, Farzaneh M. Parizi, Derek van Tilborg, Nicolas Renaud, Daan Sybrandi, Rafaella Buzatu, Daniel T. Rademaker, Peter A. C. ‘t Hoen, and Li C. Xue
- Subjects
peptide:MHC ,integrative modelling ,computational structural biology ,large-scale 3D-modelling ,computational immunology ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Deeper understanding of T-cell-mediated adaptive immune responses is important for the design of cancer immunotherapies and antiviral vaccines against pandemic outbreaks. T-cells are activated when they recognize foreign peptides that are presented on the cell surface by Major Histocompatibility Complexes (MHC), forming peptide:MHC (pMHC) complexes. 3D structures of pMHC complexes provide fundamental insight into T-cell recognition mechanism and aids immunotherapy design. High MHC and peptide diversities necessitate efficient computational modelling to enable whole proteome structural analysis. We developed PANDORA, a generic modelling pipeline for pMHC class I and II (pMHC-I and pMHC-II), and present its performance on pMHC-I here. Given a query, PANDORA searches for structural templates in its extensive database and then applies anchor restraints to the modelling process. This restrained energy minimization ensures one of the fastest pMHC modelling pipelines so far. On a set of 835 pMHC-I complexes over 78 MHC types, PANDORA generated models with a median RMSD of 0.70 Å and achieved a 93% success rate in top 10 models. PANDORA performs competitively with three pMHC-I modelling state-of-the-art approaches and outperforms AlphaFold2 in terms of accuracy while being superior to it in speed. PANDORA is a modularized and user-configurable python package with easy installation. We envision PANDORA to fuel deep learning algorithms with large-scale high-quality 3D models to tackle long-standing immunology challenges.
- Published
- 2022
- Full Text
- View/download PDF
11. Structure and Function of the Nuclear Pore Complex Cytoplasmic mRNA Export Platform
- Author
-
Fernandez-Martinez, Javier, Kim, Seung Joong, Shi, Yi, Upla, Paula, Pellarin, Riccardo, Gagnon, Michael, Chemmama, Ilan E, Wang, Junjie, Nudelman, Ilona, Zhang, Wenzhu, Williams, Rosemary, Rice, William J, Stokes, David L, Zenklusen, Daniel, Chait, Brian T, Sali, Andrej, and Rout, Michael P
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,2.1 Biological and endogenous factors ,Generic health relevance ,Active Transport ,Cell Nucleus ,Fungal Proteins ,Nuclear Pore ,Nuclear Pore Complex Proteins ,RNA ,Messenger ,Saccharomyces cerevisiae ,Saccharomyces cerevisiae Proteins ,Yeasts ,Nup4 complex ,Nup82 complex ,computational structural biology ,cross-linking and mass spectrometry ,electron microscopy ,integrative structure determination ,mRNA export ,mRNP remodeling ,nuclear pore complex ,small-angle X-ray scattering ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
The last steps in mRNA export and remodeling are performed by the Nup82 complex, a large conserved assembly at the cytoplasmic face of the nuclear pore complex (NPC). By integrating diverse structural data, we have determined the molecular architecture of the native Nup82 complex at subnanometer precision. The complex consists of two compositionally identical multiprotein subunits that adopt different configurations. The Nup82 complex fits into the NPC through the outer ring Nup84 complex. Our map shows that this entire 14-MDa Nup82-Nup84 complex assembly positions the cytoplasmic mRNA export factor docking sites and messenger ribonucleoprotein (mRNP) remodeling machinery right over the NPC's central channel rather than on distal cytoplasmic filaments, as previously supposed. We suggest that this configuration efficiently captures and remodels exporting mRNP particles immediately upon reaching the cytoplasmic side of the NPC.
- Published
- 2016
12. Computationally grafting an IgE epitope onto a scaffold: Implications for a pan anti-allergy vaccine design
- Author
-
Sari S. Sabban
- Subjects
Protein design ,Epitope grafting ,Vaccine design ,Computational structural biology ,Allergy ,Type I hypersensitivity ,Biotechnology ,TP248.13-248.65 - Abstract
Allergy is becoming an intensifying disease among the world population, particularly in the developed world. Once allergy develops, sufferers are permanently trapped in a hyper-immune response that makes them sensitive to innocuous substances. The immune pathway concerned with developing allergy is the Th2 immune pathway where the IgE antibody binds to its Fc∊RI receptor on Mast and Basophil cells. This paper discusses a protocol that could disrupt the binding between the antibody and its receptor for a potential permanent treatment. Ten proteins were computationally designed to display a human IgE motif very close in proximity to the IgE antibody’s Fc∊RI receptor’s binding site in an effort for these proteins to be used as a vaccine against our own IgE antibody. The motif of interest was the FG loop motif and it was excised and grafted onto a Staphylococcus aureus protein (PDB ID 1YN3), then the motif + scaffold structure had its sequence re-designed around the motif to find an amino acid sequence that would fold to the designed structure correctly. These ten computationally designed proteins showed successful folding when simulated using Rosetta’s AbinitioRelax folding simulation and the IgE epitope was clearly displayed in its native three-dimensional structure in all of them. These designed proteins have the potential to be used as a pan anti-allergy vaccine. This work employedin silicobased methods for designing the proteins and did not include any experimental verifications.
- Published
- 2021
- Full Text
- View/download PDF
13. Fragment molecular orbital calculations for biomolecules.
- Author
-
Fukuzawa, Kaori and Tanaka, Shigenori
- Subjects
- *
MOLECULAR orbitals , *LIFE sciences , *MOLECULAR recognition , *BIOMOLECULES , *INTERMOLECULAR interactions , *BIOLOGICAL databases , *NUCLEIC acids - Abstract
Exploring biomolecule behavior, such as proteins and nucleic acids, using quantum mechanical theory can identify many life science phenomena from first principles. Fragment molecular orbital (FMO) calculations of whole single particles of biomolecules can determine the electronic state of the interior and surface of molecules and explore molecular recognition mechanisms based on intermolecular and intramolecular interactions. In this review, we summarized the current state of FMO calculations in drug discovery, virology, and structural biology, as well as recent developments from data science. • The fragment molecular orbital method allows quantitative analysis of intra- and intermolecular interactions of biomolecules. • It can be used to understand molecular recognition mechanism of complex systems such as proteins, nucleic acids and ligands. • Collaboration between structural biology and FMO database can build an information infrastructure for life sciences. • COVID-19 brought innovations in FMO research, including drug screening, multi-structure sampling and machine learning. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
14. Predicting Secondary Structure for Human Proteins Based on Chou-Fasman Method
- Author
-
Kounelis, Fotios, Kanavos, Andreas, Livieris, Ioannis E., Vonitsanos, Gerasimos, Pintelas, Panagiotis, Rannenberg, Kai, Editor-in-Chief, Sakarovitch, Jacques, Editorial Board Member, Goedicke, Michael, Editorial Board Member, Tatnall, Arthur, Editorial Board Member, Neuhold, Erich J., Editorial Board Member, Pras, Aiko, Editorial Board Member, Tröltzsch, Fredi, Editorial Board Member, Pries-Heje, Jan, Editorial Board Member, Kreps, David, Editorial Board Member, Reis, Ricardo, Editorial Board Member, Furnell, Steven, Editorial Board Member, Furbach, Ulrich, Editorial Board Member, Winckler, Marco, Editorial Board Member, Malaka, Rainer, Editorial Board Member, MacIntyre, John, editor, Maglogiannis, Ilias, editor, Iliadis, Lazaros, editor, and Pimenidis, Elias, editor
- Published
- 2019
- Full Text
- View/download PDF
15. Computational Structural Biology
- Author
-
Jaeger, Dieter, editor and Jung, Ranu, editor
- Published
- 2022
- Full Text
- View/download PDF
16. Pathogenicity of new BEST1 variants identified in Italian patients with best vitelliform macular dystrophy assessed by computational structural biology
- Author
-
Vladimir Frecer, Giancarlo Iarossi, Anna Paola Salvetti, Paolo Enrico Maltese, Giulia Delledonne, Marta Oldani, Giovanni Staurenghi, Benedetto Falsini, Angelo Maria Minnella, Lucia Ziccardi, Adriano Magli, Leonardo Colombo, Fabiana D’Esposito, Jan Miertus, Francesco Viola, Marcella Attanasio, Emilia Maggio, and Matteo Bertelli
- Subjects
Best vitelliform macular dystrophy ,Best disease ,Best-corrected visual acuity ,Computational structural biology ,Medicine - Abstract
Abstract Background Best vitelliform macular dystrophy (BVMD) is an autosomal dominant macular degeneration. The typical central yellowish yolk-like lesion usually appears in childhood and gradually worsens. Most cases are caused by variants in the BEST1 gene which encodes bestrophin-1, an integral membrane protein found primarily in the retinal pigment epithelium. Methods Here we describe the spectrum of BEST1 variants identified in a cohort of 57 Italian patients analyzed by Sanger sequencing. In 13 cases, the study also included segregation analysis in affected and unaffected relatives. We used molecular mechanics to calculate two quantitative parameters related to calcium-activated chloride channel (CaCC composed of 5 BEST1 subunits) stability and calcium-dependent activation and related them to the potential pathogenicity of individual missense variants detected in the probands. Results Thirty-six out of 57 probands (63% positivity) and 16 out of 18 relatives proved positive to genetic testing. Family study confirmed the variable penetrance and expressivity of the disease. Six of the 27 genetic variants discovered were novel: p.(Val9Gly), p.(Ser108Arg), p.(Asn179Asp), p.(Trp182Arg), p.(Glu292Gln) and p.(Asn296Lys). All BEST1 variants were assessed in silico for potential pathogenicity. Our computational structural biology approach based on 3D model structure of the CaCC showed that individual amino acid replacements may affect channel shape, stability, activation, gating, selectivity and throughput, and possibly also other features, depending on where the individual mutated amino acid residues are located in the tertiary structure of BEST1. Statistically significant correlations between mean logMAR best-corrected visual acuity (BCVA), age and modulus of computed BEST1 dimerization energies, which reflect variations in the in CaCC stability due to amino acid changes, permitted us to assess the pathogenicity of individual BEST1 variants. Conclusions Using this computational approach, we designed a method for estimating BCVA progression in patients with BEST1 variants.
- Published
- 2019
- Full Text
- View/download PDF
17. Computation-Aided Protein Engineering for Targeted Therapeutic Delivery
- Author
-
Ding, Xiaozhe
- Subjects
Computational Science and Engineering ,drug delivery ,protein-protein interactions ,protein engineering ,Bioengineering ,gene therapy ,computational structural biology - Abstract
My Ph.D. projects centered on using computational structural biology tools to develop protein engineering methods for targeted therapeutic delivery, emphasizing delivering molecules to the brain. In this thesis, I focus on three main projects. First, utilizing computational structural biology techniques, I investigate the molecular mechanism that enables engineered adeno-associated viral (AAV) capsids to cross the blood-brain barrier (BBB). I develop a pipeline to model the vast and dynamic complex between engineered AAV capsids and their BBB receptors. I also apply a tool, recently developed by myself and discussed in Chapter 3, to distinguish capsids that bind to different receptors. The findings of this study can lead to novel approaches for developing chemicals and biologicals that can penetrate the human brain (Chapter 2). Second, I describe the development of Automated Pairwise Peptide-Receptor AnalysIs for Screening Engineered proteins (APPRAISE). This computational pipeline predicts the receptor binding propensity of engineered proteins based on competitive modeling and physics-grounded analysis. I show that APPRAISE is capable of distinguishing between receptor-dependent and receptor-independent adeno-associated viral vectors and ranking various engineered proteins, such as miniproteins binding to the SARS-CoV-2 spike and nanobodies binding to a G-protein-coupled receptor. A top performer in an in silico screening using APPRAISE was validated experimentally (Chapter 3). Third, I show an example to engineer a genetically encoded transmitter indicator (GETI), which may eventually be a cargo delivered to the brain. The GETI has a novel scaffold based on bacterial repressors, a class of transcriptional regulators that are critical for bacteria to respond to environmental chemicals. I repurposed an antibiotic-sensing repressor protein to bind a neurotransmitter, melatonin, using machine-learning-guided directed evolution. A melatonin indicator was then created by integrating the repurposed receptor with a fluorescent protein. This engineering platform may be adapted to create bio-orthogonal GETIs for various neurotransmitters (Chapter 4).
- Published
- 2023
- Full Text
- View/download PDF
18. De novo protein design by inversion of the AlphaFold structure prediction network
- Author
-
Casper A. Goverde, Benedict Wolf, Hamed Khakzad, Stéphane Rosset, and Bruno E. Correia
- Subjects
machine learning ,AlphaFold2 ,Molecular Biology ,Biochemistry ,De novo protein design ,structure prediction network inversion ,computational structural biology - Abstract
De novoprotein design enhances our understanding of the principles that govern protein folding and interactions, and has the potential to revolutionize biotechnology through the engineering of novel protein functionalities. Despite recent progress in computational design strategies,de novodesign of protein structures remains challenging, given the vast size of the sequence-structure space. AlphaFold2 (AF2), a state-of-the-art neural network architecture, achieved remarkable accuracy in predicting protein structures from amino acid sequences. This raises the question whether AF2 has learned the principles of protein folding sufficiently for de novo design. Here, we sought to answer this question by inverting the AF2 network, using the prediction weight set and a loss function to bias the generated sequences to adopt a target fold. Initial design trials resulted in de novo designs with an overrepresentation of hydrophobic residues on the protein surface compared to their natural protein family, requiring additional surface optimization. In silico validation of the designs showed protein structures with the correct fold, a hydrophilic surface and a densely packed hydrophobic core. In vitro validation showed that several designs were folded and stable in solution with high melting temperatures. In summary, our design workflow solely based on AF2 does not seem to fully capture basic principles of de novo protein design, as observed in the protein surface’s hydrophobic vs. hydrophilic patterning. However, with minimal post-design intervention, these pipelines generated viable sequences as assessed experimental characterization. Thus such pipelines show the potential to contribute to solving outstanding challenges in de novo protein design.
- Published
- 2022
- Full Text
- View/download PDF
19. Roadmap of 3D-BioInfo-PT, the BioData.pt community of computational structural biology researchers
- Author
-
Melo, Manuel N., Lousa, Diana, Victor, Bruno L., Machuqueiro, Miguel, Costa, Paulo J., Martel, Paulo, Melo, Rita, Sousa, Sérgio F., and Moreira, Irina
- Subjects
computational biology ,3D-BioInfo ,computational structural biology - Abstract
Inception: the 3D-BioInfo-PT project is born, partially as a re-branding under Biodata.pt of the community involved in the EJIBCE meetings – Encontros de Jovens Investigadores em Biologia Computacional Estrutural. Materialization: Planned roadmap presented and debated at the Biodata.pt Technical Meeting. Teaching: the first 3D-BioInfo-PT workshop takes place at ITQB NOVA. It is an event with 35 participants (out of 62 applicants) hailing from 7 different institutions. Main support from the Oeiras Valley initiative and Wallfuture Lda. Continuity: maintain the 3D Biotalks seminar cycle and expand its reach. Networking: maintain the EJIBCE tradition of the annual meeting. Organize a satellite workshop. Collaboration: bring together the 3D-BioInfo-PT coordinating groups to write a perspective article on computational structural research in Portugal., https://3d-bioinfo-pt.github.io/
- Published
- 2022
- Full Text
- View/download PDF
20. Elucidating the Structural Impacts of Protein InDels
- Author
-
Muneeba Jilani, Alistair Turcan, Nurit Haspel, and Filip Jagodzinski
- Subjects
computational structural biology ,protein InDel mutations ,graph-theory ,rigidity ,INDEL Mutation ,Amino Acid Substitution ,Mutation ,Proteins ,Amino Acids ,Molecular Biology ,Biochemistry - Abstract
The effects of amino acid insertions and deletions (InDels) remain a rather under-explored area of structural biology. These variations oftentimes are the cause of numerous disease phenotypes. In spite of this, research to study InDels and their structural significance remains limited, primarily due to a lack of experimental information and computational methods. In this work, we fill this gap by modeling InDels computationally; we investigate the rigidity differences between the wildtype and a mutant variant with one or more InDels. Further, we compare how structural effects due to InDels differ from the effects of amino acid substitutions, which are another type of amino acid mutation. We finish by performing a correlation analysis between our rigidity-based metrics and wet lab data for their ability to infer the effects of InDels on protein fitness.
- Published
- 2022
21. Deep Learning for Validating and Estimating Resolution of Cryo-Electron Microscopy Density Maps †
- Author
-
Todor Kirilov Avramov, Dan Vyenielo, Josue Gomez-Blanco, Swathi Adinarayanan, Javier Vargas, and Dong Si
- Subjects
computational structural biology ,cryo-electron microscopy ,deep learning ,resolution validation ,Organic chemistry ,QD241-441 - Abstract
Cryo-electron microscopy (cryo-EM) is becoming the imaging method of choice for determining protein structures. Many atomic structures have been resolved based on an exponentially growing number of published three-dimensional (3D) high resolution cryo-EM density maps. However, the resolution value claimed for the reconstructed 3D density map has been the topic of scientific debate for many years. The Fourier Shell Correlation (FSC) is the currently accepted cryo-EM resolution measure, but it can be subjective, manipulated, and has its own limitations. In this study, we first propose supervised deep learning methods to extract representative 3D features at high, medium and low resolutions from simulated protein density maps and build classification models that objectively validate resolutions of experimental 3D cryo-EM maps. Specifically, we build classification models based on dense artificial neural network (DNN) and 3D convolutional neural network (3D CNN) architectures. The trained models can classify a given 3D cryo-EM density map into one of three resolution levels: high, medium, low. The preliminary DNN and 3D CNN models achieved 92.73% accuracy and 99.75% accuracy on simulated test maps, respectively. Applying the DNN and 3D CNN models to thirty experimental cryo-EM maps achieved an agreement of 60.0% and 56.7%, respectively, with the author published resolution value of the density maps. We further augment these previous techniques and present preliminary results of a 3D U-Net model for local resolution classification. The model was trained to perform voxel-wise classification of 3D cryo-EM density maps into one of ten resolution classes, instead of a single global resolution value. The U-Net model achieved 88.3% and 94.7% accuracy when evaluated on experimental maps with local resolutions determined by MonoRes and ResMap methods, respectively. Our results suggest deep learning can potentially improve the resolution evaluation process of experimental cryo-EM maps.
- Published
- 2019
- Full Text
- View/download PDF
22. Introduction
- Author
-
Levitt, Michael, Leontis, Neocles, editor, and Westhof, Eric, editor
- Published
- 2012
- Full Text
- View/download PDF
23. Computational Prediction of the Heterodimeric and Higher-Order Structure of gpE1/gpE2 Envelope Glycoproteins Encoded by Hepatitis C Virus.
- Author
-
Freedman, Holly, Logan, Michael R., Hockman, Darren, Leman, Julia Koehler, Law, John Lok Man, and Houghton, Michael
- Subjects
- *
HEPATITIS C treatment , *HEPATITIS C virus , *HETERODIMERS , *GLYCOPROTEINS , *ANTIVIRAL agents - Abstract
Despite the recent success of newly developed direct-acting antivirals against hepatitis C, the disease continues to be a global health threat due to the lack of diagnosis of most carriers and the high cost of treatment. The heterodimer formed by glycoproteins E1 and E2 within the hepatitis C virus (HCV) lipid envelope is a potential vaccine candidate and antiviral target. While the structure of E1/E2 has not yet been resolved, partial crystal structures of the E1 and E2 ectodomains have been determined. The unresolved parts of the structure are within the realm of what can be modeled with current computational modeling tools. Furthermore, a variety of additional experimental data is available to support computational predictions of E1/E2 structure, such as data from antibody binding studies, cryo-electron microscopy (cryo-EM), mutational analyses, peptide binding analysis, linker-scanning mutagenesis, and nuclear magnetic resonance (NMR) studies. In accordance with these rich experimental data, we have built an in silico model of the full-length E1/E2 heterodimer. Our model supports that E1/E2 assembles into a trimer, which was previously suggested from a study by Falson and coworkers (P. Falson, B. Bartosch, K. Alsaleh, B. A. Tews, A. Loquet, Y. Ciczora, L. Riva, C. Montigny, C. Montpellier, G. Duverlie, E. I. Pecheur, M. le Maire, F. L. Cosset, J. Dubuisson, and F. Penin, J. Virol. 89:10333-10346, 2015, https://doi.org/10.1128/JVI.00991-15). Size exclusion chromatography and Western blotting data obtained by using purified recombinant E1/E2 support our hypothesis. Our model suggests that during virus assembly, the trimer of E1/E2 may be further assembled into a pentamer, with 12 pentamers comprising a single HCV virion. We anticipate that this new model will provide a useful framework for HCV envelope structure and the development of antiviral strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
24. Correlation between the binding affinity and the conformational entropy of nanobodies targeting the SARSCoV- 2 spike protein
- Author
-
Mikolajek, Halina, Weckener, Miriam, Z. Faidon Brotzakis, Jiandong Huo, Dalietou, Evmorfia V, Bas, Audrey Le, Sormanni, Pietro, Harrison, Peter J, Ward, Philip N, Truong, Steven, Moynie, Lucile, Clare, Daniel, Dumoux, Maud, Dormon, Josh, Norman, Chelsea, Hussain, Naveed, Vogirala, Vinod, Owens, Raymond J, Vendruscolo, Michele, and Naismith, James H
- Subjects
SARS-Cov2 ,protein-protein interactions ,Computational structural biology ,Molecular-dynamics ,Camelid nano bodies ,Antibody design ,Affinity maturation ,Cryo-EM - Abstract
Camelid single-domain antibodies, also known as nanobodies, can be readily isolated from na.ve libraries for specific targets. However, they often bind too weakly to their targets to be immediately useful. Laboratory-based genetic engineering methods to enhance their affinity, a process known as maturation, can deliver useful reagents for different areas of biology and potentially medicine. Using the receptor binding domain (RBD) of the SARS-CoV-2 spike protein, we generated closely related nanobodies with micromolar to nanomolar binding affinities. By analysing the structure-activity relationship using X-ray crystallography, cryo-electron microscopy, and biophysical methods, we observed that higher conformational entropy losses in the formation of the spike protein-nanobody complex are associated with tighter binding. To investigate this, we generated structural ensembles of the different complexes from electron microscopy maps and correlated the conformational fluctuations with binding affinity. This insight guided the engineering of a nanobody with high binding affinity for the spike protein., These Cryo-EM based structural ensembles of spike-nanobody complexes generated by the EMMI method.
- Published
- 2022
- Full Text
- View/download PDF
25. De novo protein design by inversion of the AlphaFold structure prediction network.
- Author
-
Goverde CA, Wolf B, Khakzad H, Rosset S, and Correia BE
- Subjects
- Proteins chemistry, Amino Acid Sequence, Protein Folding, Furylfuramide, Protein Engineering methods
- Abstract
De novo protein design enhances our understanding of the principles that govern protein folding and interactions, and has the potential to revolutionize biotechnology through the engineering of novel protein functionalities. Despite recent progress in computational design strategies, de novo design of protein structures remains challenging, given the vast size of the sequence-structure space. AlphaFold2 (AF2), a state-of-the-art neural network architecture, achieved remarkable accuracy in predicting protein structures from amino acid sequences. This raises the question whether AF2 has learned the principles of protein folding sufficiently for de novo design. Here, we sought to answer this question by inverting the AF2 network, using the prediction weight set and a loss function to bias the generated sequences to adopt a target fold. Initial design trials resulted in de novo designs with an overrepresentation of hydrophobic residues on the protein surface compared to their natural protein family, requiring additional surface optimization. In silico validation of the designs showed protein structures with the correct fold, a hydrophilic surface and a densely packed hydrophobic core. In vitro validation showed that 7 out of 39 designs were folded and stable in solution with high melting temperatures. In summary, our design workflow solely based on AF2 does not seem to fully capture basic principles of de novo protein design, as observed in the protein surface's hydrophobic vs. hydrophilic patterning. However, with minimal post-design intervention, these pipelines generated viable sequences as assessed experimental characterization. Thus, such pipelines show the potential to contribute to solving outstanding challenges in de novo protein design., (© 2023 The Authors. Protein Science published by Wiley Periodicals LLC on behalf of The Protein Society.)
- Published
- 2023
- Full Text
- View/download PDF
26. ProDCoNN: Protein design using a convolutional neural network
- Author
-
Chun-Chao Lo, Chenran Wang, Xiuwen Liu, Jinfeng Zhang, Yang Chen, Wu Wei, and Yuan Zhang
- Subjects
Computer science ,Protein design ,Biophysics ,Datasets as Topic ,Protein Engineering ,Biochemistry ,Convolutional neural network ,Protein Structure, Secondary ,Article ,03 medical and health sciences ,Protein structure ,Structural Biology ,Computational structural biology ,Amino Acid Sequence ,Databases, Protein ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,business.industry ,030302 biochemistry & molecular biology ,Proteins ,Large numbers ,Pattern recognition ,Protein engineering ,Bond length ,Benchmarking ,Molecular geometry ,Artificial intelligence ,Neural Networks, Computer ,business ,Sequence Alignment ,Algorithm ,Software - Abstract
Designing protein sequences that fold to a given three-dimensional (3D) structure has long been a challenging problem in computational structural biology with significant theoretical and practical implications. In this study, we first formulated this problem as predicting the residue type given the 3D structural environment around the C(α) atom of a residue, which is repeated for each residue of a protein. We designed a nine-layer 3D deep convolutional neural network (CNN) that takes as input a gridded box with the atomic coordinates and types around a residue. Several CNN layers were designed to capture structure information at different scales, such as bond lengths, bond angles, torsion angles, and secondary structures. Trained on a very large number of protein structures, the method, called ProDCoNN (protein design with CNN), achieved state-of-the-art performance when tested on large numbers of test proteins and benchmark datasets.
- Published
- 2020
- Full Text
- View/download PDF
27. Computational Structural Biology for Drug Discovery
- Author
-
Andrey V. Ilatovskiy and Ruben Abagyan
- Subjects
Chemistry ,Drug discovery ,Transition state analog ,Protein–ligand complex ,Computational structural biology ,Computational biology ,Binding site - Published
- 2020
- Full Text
- View/download PDF
28. Computational approaches to macromolecular interactions in the cell
- Author
-
Ilya A. Vakser and Eric J. Deeds
- Subjects
Models, Molecular ,0303 health sciences ,Molecular Structure ,Macromolecular Substances ,Extramural ,Computer science ,Principal (computer security) ,Computational Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Structural Biology ,Strategic direction ,Computational structural biology ,Biochemical engineering ,Whole cell ,Molecular Biology ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Structural modeling of a cell is an evolving strategic direction in computational structural biology. It takes advantage of new powerful modeling techniques, deeper understanding of fundamental principles of molecular structure and assembly, and rapid growth of the amount of structural data generated by experimental techniques. Key modeling approaches to principal types of macromolecular assemblies in a cell already exist. The main challenge, along with the further development of these modeling approaches, is putting them together in a consistent, unified whole cell model. This opinion piece addresses the fundamental aspects of modeling macromolecular assemblies in a cell, and the state-of-the-art in modeling of the principal types of such assemblies.
- Published
- 2019
- Full Text
- View/download PDF
29. Computational Structural Biology
- Author
-
Jaeger, Dieter, editor and Jung, Ranu, editor
- Published
- 2015
- Full Text
- View/download PDF
30. Reduced B cell antigenicity of Omicron lowers host serologic response.
- Author
-
Tubiana, Jérôme, Xiang, Yufei, Fan, Li, Wolfson, Haim J., Chen, Kong, Schneidman-Duhovny, Dina, and Shi, Yi
- Abstract
The SARS-CoV-2 Omicron variant evades most neutralizing vaccine-induced antibodies and is associated with lower antibody titers upon breakthrough infections than previous variants. However, the mechanism remains unclear. Here, we find using a geometric deep-learning model that Omicron's extensively mutated receptor binding site (RBS) features reduced antigenicity compared with previous variants. Mice immunization experiments with different recombinant receptor binding domain (RBD) variants confirm that the serological response to Omicron is drastically attenuated and less potent. Analyses of serum cross-reactivity and competitive ELISA reveal a reduction in antibody response across both variable and conserved RBD epitopes. Computational modeling confirms that the RBS has a potential for further antigenicity reduction while retaining efficient receptor binding. Finally, we find a similar trend of antigenicity reduction over decades for hCoV229E, a common cold coronavirus. Thus, our study explains the reduced antibody titers associated with Omicron infection and reveals a possible trajectory of future viral evolution. [Display omitted] • Omicron breakthrough infection elicits lower antibody response than prior variants • Deep-learning model predicts reduced antigenicity of the Omicron RBD • Mice immunization experiments show reduced B cell immunogenicity of Omicron spike RBD • Additional mutations could reduce antigenicity while maintaining receptor binding SARS-CoV-2 Omicron variant evades most neutralizing vaccine-induced antibodies and is associated with lower antibody titers upon breakthrough infections than previous variants. Tubiana et al. investigate the underlying mechanism using geometric deep learning, mice immunization experiments, and biochemical assays. Mutations reduce antigenicity of the receptor binding site, leading to lower antibody response. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
31. Chapter 12. Machine Learning Algorithms for the Analysis of Molecular Dynamics Trajectories
- Author
-
Byu-Ri Sim, Abbas Khan, and Dong-qing Wei
- Subjects
Molecular dynamics ,business.industry ,Computational structural biology ,Artificial intelligence ,Molecular systems ,business ,Machine learning ,computer.software_genre ,Algorithm ,computer ,Field (computer science) - Abstract
Molecular dynamics (MD) simulations have been a pivotal tool to understand many processes, including drug binding, protein folding and many others in the field of materials science. In the past decades, machine learning (ML) has become a valuable tool in the field of MD simulations. The synergies between ML algorithms and MD simulations, including both classical and quantum mechanical simulations, can substantially transform the way we predict and solve computational structural biology, drug discovery and MD simulation problems. In this chapter, we describe how ML advances the understanding and interpretation of MD trajectories. This chapter also concentrates on the implementation of ML algorithms in MD simulations that can be programmed so that they can be used as input to train ML models for the quantitative comprehension of molecular systems.
- Published
- 2021
- Full Text
- View/download PDF
32. Algorithmique des graphes pour les réseaux et la biologie structurale computationnelle
- Author
-
Mazauric, Dorian, Mazauric, Dorian, Université Côte d'Azur (UCA), Algorithms, Biology, Structure (ABS), Inria Sophia Antipolis - Méditerranée (CRISAM), Institut National de Recherche en Informatique et en Automatique (Inria)-Institut National de Recherche en Informatique et en Automatique (Inria), Université Côte d'Azur, and Frédéric Cazals
- Subjects
[INFO.INFO-CC]Computer Science [cs]/Computational Complexity [cs.CC] ,graphs ,[INFO.INFO-DS]Computer Science [cs]/Data Structures and Algorithms [cs.DS] ,[INFO.INFO-DS] Computer Science [cs]/Data Structures and Algorithms [cs.DS] ,[INFO] Computer Science [cs] ,algorithms ,complexité ,biologie structurale computationnelle ,graphes ,[INFO]Computer Science [cs] ,[INFO.INFO-CC] Computer Science [cs]/Computational Complexity [cs.CC] ,[INFO.INFO-BI]Computer Science [cs]/Bioinformatics [q-bio.QM] ,complexity ,computational structural biology ,algorithmes ,[INFO.INFO-BI] Computer Science [cs]/Bioinformatics [q-bio.QM] - Published
- 2021
33. IRAA: A statistical tool for investigating a protein-protein interaction interface from multiple structures.
- Author
-
Belapure J, Sorokina M, and Kastritis PL
- Subjects
- Humans, Angiotensin-Converting Enzyme 2 metabolism, Binding Sites, Protein Binding, Proteins metabolism, SARS-CoV-2 metabolism, Spike Glycoprotein, Coronavirus chemistry, Algorithms, COVID-19
- Abstract
Understanding protein-protein interactions (PPIs) is fundamental to infer how different molecular systems work. A major component to model molecular recognition is the buried surface area (BSA), that is, the area that becomes inaccessible to solvent upon complex formation. To date, many attempts tried to connect BSA to molecular recognition principles, and in particular, to the underlying binding affinity. However, the most popular approach to calculate BSA is to use a single (or in some cases few) bound structures, consequently neglecting a wealth of structural information of the interacting proteins derived from ensembles corresponding to their unbound and bound states. Moreover, the most popular method inherently assumes the component proteins to bind as rigid entities. To address the above shortcomings, we developed a Monte Carlo method-based Interface Residue Assessment Algorithm (IRAA), to calculate a combined distribution of BSA for a given complex. Further, we apply our algorithm to human ACE2 and SARS-CoV-2 Spike protein complex, a system of prime importance. Results show a much broader distribution of BSA compared to that obtained from only the bound structure or structures and extended residue members of the interface with implications to the underlying biomolecular recognition. We derive that specific interface residues of ACE2 and of S-protein are consistently highly flexible, whereas other residues systematically show minor conformational variations. In effect, IRAA facilitates the use of all available structural data for any biomolecular complex of interest, extracting quantitative parameters with statistical significance, thereby providing a deeper biophysical understanding of the molecular system under investigation., (© 2022 The Authors. Protein Science published by Wiley Periodicals LLC on behalf of The Protein Society.)
- Published
- 2023
- Full Text
- View/download PDF
34. Next-Generation Computational Tools and Resources for Coronavirus Research: from Detection to Vaccine Discovery
- Author
-
Yumnam Silla, Rajiv Das Kangabam, Riya Roy, Namrata Misra, Mrutyunjay Suar, Arpan Ghosh, and Susrita Sahoo
- Subjects
0301 basic medicine ,Proteome ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Health Informatics ,Disease ,Computational biology ,Biology ,medicine.disease_cause ,Article ,03 medical and health sciences ,Databases ,0302 clinical medicine ,Pandemic ,medicine ,Humans ,Computer Simulation ,Computational structural biology ,Pandemics ,Coronavirus ,SARS-CoV-2 ,Computational Biology ,COVID-19 ,Computer Science Applications ,030104 developmental biology ,Computational Tools ,Novel virus ,Genomic ,030217 neurology & neurosurgery - Abstract
The COVID-19 pandemic has affected 215 countries and territories around the world with 60,187,347 coronavirus cases and 17,125,719 currently infected patients confirmed as of the 25th of November 2020. Currently, many countries are working on developing new vaccines and therapeutic drugs for this novel virus strain, and a few of them are in different phases of clinical trials. The advancement in high-throughput sequence technologies, along with the application of bioinformatics, offers invaluable knowledge on genomic characterization and molecular pathogenesis of coronaviruses. Recent multi-disciplinary studies using bioinformatics methods like sequence-similarity, phylogenomic, and computational structural biology have provided an in-depth understanding of the molecular and biochemical basis of infection, atomic-level recognition of the viral-host receptor interaction, functional annotation of important viral proteins, and evolutionary divergence across different strains. Additionally, various modern immunoinformatic approaches are also being used to target the most promiscuous antigenic epitopes from the SARS-CoV-2 proteome for accelerating the vaccine development process. In this review, we summarize various important computational tools and databases available for systematic sequence-structural study on coronaviruses. The features of these public resources have been comprehensively discussed, which may help experimental biologists with predictive insights useful for ongoing research efforts to find therapeutics against the infectious COVID-19 disease., Graphical abstract Image 1, Highlights • This review presents a comprehensive and up-to-date overview of newly developed computational resources including tools and web servers focused on coronaviruses. • We have categorized these popular tools and databases depending on the utility and application such as for coronavirus detection, comparative genomics analysis, vaccine and drug discovery, molecular docking, tools for SARS-CoV-2 detection, and other applications in coronaviruses study. • The features of each of the tools have been discussed along with their application in recent coronavirus research studies.
- Published
- 2020
35. Cotranscriptional kinetic folding of RNA secondary structures
- Author
-
Thanh H Vo and Pekka Orponen
- Subjects
chemistry.chemical_classification ,Folding (chemistry) ,chemistry ,Kinetic model ,RNA ,Nucleotide ,Computational structural biology ,Sequence (biology) ,Computational biology ,Rna folding ,Nucleic acid structure - Abstract
Computational prediction of RNA structures is an important problem in computational structural biology. Studies of RNA structure formation often assume that the process starts from a fully synthesized sequence. Experimental evidence, however, has shown that RNA folds concurrently with its elongation. We investigate RNA structure formation, taking into account also the cotranscriptional effects. We propose a single-nucleotide resolution kinetic model of the folding process of RNA molecules, where the polymerase-driven elongation of an RNA strand by a new nucleotide is included as a primitive operation, together with a stochastic simulation method that implements this folding concurrently with the transcriptional synthesis. Numerical case studies show that our cotranscriptional RNA folding model can predict the formation of metastable conformations that are favored in actual biological systems. Our new computational tool can thus provide quantitative predictions and offer useful insights into the kinetics of RNA folding.
- Published
- 2020
- Full Text
- View/download PDF
36. Cotranscriptional kinetic folding of RNA secondary structures including pseudoknots
- Author
-
Dani Korpela, Vo Hong Thanh, and Pekka Orponen
- Subjects
Models, Molecular ,RNA Folding ,Transcription, Genetic ,Sequence (biology) ,Computational biology ,Nucleic acid secondary structure ,Plant Viruses ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,RNA Viruses ,Nucleotide ,Computational structural biology ,Nucleic acid structure ,Molecular Biology ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Chemistry ,RNA ,Computational Biology ,Biomolecules (q-bio.BM) ,Folding (chemistry) ,Computational Mathematics ,Kinetics ,Computational Theory and Mathematics ,Quantitative Biology - Biomolecules ,030220 oncology & carcinogenesis ,Modeling and Simulation ,FOS: Biological sciences ,Nucleic Acid Conformation ,RNA, Viral ,Rna folding ,Signal Recognition Particle ,Algorithms - Abstract
Computational prediction of RNA structures is an important problem in computational structural biology. Studies of RNA structure formation often assume that the process starts from a fully synthesized sequence. Experimental evidence, however, has shown that RNA folds concurrently with its elongation. We investigate RNA secondary structure formation, including pseudoknots, that takes into account the cotranscriptional effects. We propose a single-nucleotide resolution kinetic model of the folding process of RNA molecules, where the polymerase-driven elongation of an RNA strand by a new nucleotide is included as a primitive operation, together with a stochastic simulation method that implements this folding concurrently with the transcriptional synthesis. Numerical case studies show that our cotranscriptional RNA folding model can predict the formation of conformations that are favored in actual biological systems. Our new computational tool can thus provide quantitative predictions and offer useful insights into the kinetics of RNA folding., 20 pages, 15 figures
- Published
- 2020
37. A simple strategy to enhance the speed of protein secondary structure prediction without sacrificing accuracy
- Author
-
Wei-Cheng Lo, Sheng Hung Juan, and Teng Ruei Chen
- Subjects
Computer science ,Entropy ,Information Theory ,Protein Structure Prediction ,Biochemistry ,Protein Structure, Secondary ,Reduction (complexity) ,Database and Informatics Methods ,Protein Structure Databases ,0302 clinical medicine ,Protein sequencing ,Protein structure ,Mathematical and Statistical Techniques ,Macromolecular Structure Analysis ,Databases, Protein ,Protein secondary structure ,0303 health sciences ,Multidisciplinary ,Physics ,Statistics ,Protein structure prediction ,Data Accuracy ,Physical Sciences ,Medicine ,Thermodynamics ,Information Technology ,Algorithm ,Information Entropy ,Sequence Analysis ,Algorithms ,Research Article ,Protein Structure ,Computer and Information Sciences ,Bioinformatics ,Science ,Sequence Databases ,Homology (mathematics) ,Research and Analysis Methods ,Universal Protein Resource ,03 medical and health sciences ,Entropy (information theory) ,Computational structural biology ,Statistical Methods ,Molecular Biology ,030304 developmental biology ,Sequence Homology, Amino Acid ,Biology and Life Sciences ,Proteins ,Computational Biology ,Data Reduction ,Biological Databases ,Sequence Alignment ,030217 neurology & neurosurgery ,Mathematics ,Forecasting - Abstract
The secondary structure prediction of proteins is a classic topic of computational structural biology with a variety of applications. During the past decade, the accuracy of prediction achieved by state-of-the-art algorithms has been >80%; meanwhile, the time cost of prediction increased rapidly because of the exponential growth of fundamental protein sequence data. Based on literature studies and preliminary observations on the relationships between the size/homology of the fundamental protein dataset and the speed/accuracy of predictions, we raised two hypotheses that might be helpful to determine the main influence factors of the efficiency of secondary structure prediction. Experimental results of size and homology reductions of the fundamental protein dataset supported those hypotheses. They revealed that shrinking the size of the dataset could substantially cut down the time cost of prediction with a slight decrease of accuracy, which could be increased on the contrary by homology reduction of the dataset. Moreover, the Shannon information entropy could be applied to explain how accuracy was influenced by the size and homology of the dataset. Based on these findings, we proposed that a proper combination of size and homology reductions of the protein dataset could speed up the secondary structure prediction while preserving the high accuracy of state-of-the-art algorithms. Testing the proposed strategy with the fundamental protein dataset of the year 2018 provided by the Universal Protein Resource, the speed of prediction was enhanced over 20 folds while all accuracy measures remained equivalently high. These findings are supposed helpful for improving the efficiency of researches and applications depending on the secondary structure prediction of proteins. To make future implementations of the proposed strategy easy, we have established a database of size and homology reduced protein datasets at http://10.life.nctu.edu.tw/UniRefNR.
- Published
- 2020
38. On the Conformational Dynamics of β-Amyloid Forming Peptides: A Computational Perspective
- Author
-
Konda Mani Saravanan, Yanjie Wei, Huiling Zhang, Wenhui Xi, and Haiping Zhang
- Subjects
0301 basic medicine ,Histology ,Amyloid ,lcsh:Biotechnology ,Biomedical Engineering ,Theoretical models ,Bioengineering ,Context (language use) ,Review ,02 engineering and technology ,Computational biology ,03 medical and health sciences ,Molecular level ,β amyloid ,lcsh:TP248.13-248.65 ,Computational structural biology ,Chemistry ,Aβ peptide ,Bioengineering and Biotechnology ,amyloid ,021001 nanoscience & nanotechnology ,molecular dynamics ,Folding (chemistry) ,030104 developmental biology ,machine learning ,conformation transition ,0210 nano-technology ,neural disorders ,Biotechnology - Abstract
Understanding the conformational dynamics of proteins and peptides involved in important functions is still a difficult task in computational structural biology. Because such conformational transitions in β-amyloid (Aβ) forming peptides play a crucial role in many neurological disorders, researchers from different scientific fields have been trying to address issues related to the folding of Aβ forming peptides together. Many theoretical models have been proposed in the recent years for studying Aβ peptides using mathematical, physicochemical, and molecular dynamics simulation, and machine learning approaches. In this article, we have comprehensively reviewed the developmental advances in the theoretical models for Aβ peptide folding and interactions, particularly in the context of neurological disorders. Furthermore, we have extensively reviewed the advances in molecular dynamics simulation as a tool used for studying the conversions between polymorphic amyloid forms and applications of using machine learning approaches in predicting Aβ peptides and aggregation-prone regions in proteins. We have also provided details on the theoretical advances in the study of Aβ peptides, which would enhance our understanding of these peptides at the molecular level and eventually lead to the development of targeted therapies for certain acute neurological disorders such as Alzheimer's disease in the future.
- Published
- 2020
- Full Text
- View/download PDF
39. Integrative modelling of biomolecular complexes
- Author
-
Koukos, P.I., Bonvin, A.M.J.J., Sub NMR Spectroscopy, NMR Spectroscopy, Sub NMR Spectroscopy, and NMR Spectroscopy
- Subjects
Models, Molecular ,Proteomics ,Magnetic Resonance Spectroscopy ,Macromolecular Substances ,Computer science ,Interactions ,Crystallography, X-Ray ,Biochemistry ,Mass Spectrometry ,Docking ,03 medical and health sciences ,0302 clinical medicine ,Membrane proteins ,Computational structural biology ,Molecular Biology ,Biology ,030304 developmental biology ,0303 health sciences ,Cryoelectron Microscopy ,Computational Biology ,Biochemical engineering ,Structural biology ,Molecular simulations ,030217 neurology & neurosurgery - Abstract
In recent years, the use of integrative, information-driven computational approaches for modeling the structure of biomolecules has been increasing in popularity. These are now recognized as a crucial complement to experimental structural biology techniques such as X-ray crystallography, nuclear magnetic resonance (NMR) spectroscopy and cryo-electron microscopy (cryo-EM). This trend can be credited to a few reasons such as the increased prominence of structures solved by cryo-EM, the improvements in proteomics approaches such as cross-linking mass spectrometry (XL-MS), the drive to study systems of higher complexity in their native state, and the maturation of many computational techniques combined with the widespread availability of information-driven integrative modeling platforms. In this review, we highlight recent works that exemplify how the use of integrative and/or information-driven approaches and platforms can produce highly accurate structural models. These examples include systems which present many challenges when studied with traditional structural biology techniques such as flexible and dynamic macromolecular assemblies and membrane-associated complexes. We also identify some key areas of interest for information-driven, integrative modeling and discuss how they relate to ongoing challenges in the fields of computational structural biology. These include the use of coarse-grained force fields for biomolecular simulations—allowing for simulations across longer (time-) and bigger (size-dimension) scales—the use of bioinformatics predictions to drive sampling and/or scoring in docking such as those derived from coevolution analysis and finally the study of membrane and membrane-associated protein complexes.
- Published
- 2020
40. Integrative Modelling of Biomolecular Complexes: From Small to Large
- Subjects
Integrative modelling ,Protein-protein interactions ,Membrane ,Computational structural biology ,HADDOCK ,Data-driven ,Information-driven ,Small molecule ,Docking - Abstract
Chapter 1 provided a detailed and comprehensive review on the types of data than can be used by Integrative Modelling software like HADDOCK, ROSETTA and IMP, with a particular emphasis on the experimental techniques which can be used to map interfaces, derive distance restraints or shape-based approaches. Another focal point of the chapter is how recent advancements have affected the field of membrane protein modelling. Chapters 2 and 3 also relate to membrane protein modelling with the former describing a recently available benchmark comprised of ready-to-dock membrane protein complexes and the baseline performance of HADDOCK for the entries of the benchmark, and the latter, ongoing work regarding development of a protocol for HADDOCK for the docking of transmembrane protein complexes. The remaining of the thesis focused on small molecule modelling with Chapters 4-6 detailing three separate protocols for the docking of small molecules and protein receptors, with every protocol and chapter reflecting methodological improvements over the previous one. In Chapter 4, I described the participation of HADDOCK in the 2016 iteration of the Grand Challenge, the blind docking experiment organised by the D3R consortium. While our performance in the pose prediction component was not impressive, we could identify the main factor limiting HADDOCK’s performance, namely the selection of appropriate templates for the receptor and came up with an improved way of selecting receptors. Chapter 5 described additional improvements in our protocol related to the way the compound conformers are selected prior to docking which led to our participation in the 2017 iteration of the Grand Challenge being evaluated as one of the best. Chapter 6 detailed the development of a new protocol for protein-small molecule docking, by combining the lessons and conclusions from Chapters 4-5 and formalising their approaches in a method that relies on HADDOCK’s main strength, its ability to incorporate information to guide the simulation. This new, shape-restrained docking protocol outperformed all our previous efforts while at the same time not relying on any external software. A common denominator between the membrane protein work and the small ligand docking discussed in this thesis is the use of shape information. Indeed, chapters 3 and 6 both describe applications of shape information represented as beads to drive the modelling process. In Chapter 3 one or more layers of beads are used to implicitly represent the membrane and in Chapter 6 ligand docking is restrained to a shape based on the structure of a homologous compound. Despite the commonalities between the two protocols, the outcome of the docking is very different between the two, with the small molecule protocol achieving high-quality results and improving upon our previous efforts in this area, whereas the membrane one achieves results which are only marginally better than defining centre-of-mass restraints between the transmembrane segments of the partners for the docking. A main limiting factor in the case of membrane protein complexes seems to be the size of the complex, which defines the number of restraints defined between shape and molecules and negatively impacts performance.
- Published
- 2020
41. Integrative Modelling of Biomolecular Complexes: From Small to Large
- Author
-
Koukos, Panagiotis, Sub NMR Spectroscopy, NMR Spectroscopy, and Bonvin, Alexandre
- Subjects
Integrative modelling ,Protein-protein interactions ,Membrane ,Computational structural biology ,HADDOCK ,Data-driven ,Information-driven ,Small molecule ,Docking - Abstract
Chapter 1 provided a detailed and comprehensive review on the types of data than can be used by Integrative Modelling software like HADDOCK, ROSETTA and IMP, with a particular emphasis on the experimental techniques which can be used to map interfaces, derive distance restraints or shape-based approaches. Another focal point of the chapter is how recent advancements have affected the field of membrane protein modelling. Chapters 2 and 3 also relate to membrane protein modelling with the former describing a recently available benchmark comprised of ready-to-dock membrane protein complexes and the baseline performance of HADDOCK for the entries of the benchmark, and the latter, ongoing work regarding development of a protocol for HADDOCK for the docking of transmembrane protein complexes. The remaining of the thesis focused on small molecule modelling with Chapters 4-6 detailing three separate protocols for the docking of small molecules and protein receptors, with every protocol and chapter reflecting methodological improvements over the previous one. In Chapter 4, I described the participation of HADDOCK in the 2016 iteration of the Grand Challenge, the blind docking experiment organised by the D3R consortium. While our performance in the pose prediction component was not impressive, we could identify the main factor limiting HADDOCK’s performance, namely the selection of appropriate templates for the receptor and came up with an improved way of selecting receptors. Chapter 5 described additional improvements in our protocol related to the way the compound conformers are selected prior to docking which led to our participation in the 2017 iteration of the Grand Challenge being evaluated as one of the best. Chapter 6 detailed the development of a new protocol for protein-small molecule docking, by combining the lessons and conclusions from Chapters 4-5 and formalising their approaches in a method that relies on HADDOCK’s main strength, its ability to incorporate information to guide the simulation. This new, shape-restrained docking protocol outperformed all our previous efforts while at the same time not relying on any external software. A common denominator between the membrane protein work and the small ligand docking discussed in this thesis is the use of shape information. Indeed, chapters 3 and 6 both describe applications of shape information represented as beads to drive the modelling process. In Chapter 3 one or more layers of beads are used to implicitly represent the membrane and in Chapter 6 ligand docking is restrained to a shape based on the structure of a homologous compound. Despite the commonalities between the two protocols, the outcome of the docking is very different between the two, with the small molecule protocol achieving high-quality results and improving upon our previous efforts in this area, whereas the membrane one achieves results which are only marginally better than defining centre-of-mass restraints between the transmembrane segments of the partners for the docking. A main limiting factor in the case of membrane protein complexes seems to be the size of the complex, which defines the number of restraints defined between shape and molecules and negatively impacts performance.
- Published
- 2020
42. A computational structural biology study to understand the impact of mutation on structure-function relationship of inward-rectifier potassium ion channel Kir6.2 in human
- Author
-
Ramakrishna Vadde and Manoj K. Gupta
- Subjects
endocrine system ,0303 health sciences ,Kcnj11 gene ,endocrine system diseases ,Chemistry ,Inward-rectifier potassium ion channel ,030303 biophysics ,Structure function ,nutritional and metabolic diseases ,General Medicine ,Kir6.2 ,Potassium channel ,Cell biology ,Molecular Docking Simulation ,03 medical and health sciences ,Structure-Activity Relationship ,Adenosine Triphosphate ,Diabetes Mellitus, Type 2 ,Structural Biology ,Mutation ,Humans ,Computational structural biology ,human activities ,Molecular Biology ,Biology - Abstract
Type 2 diabetes (T2D) is clinically characterized via hyperglycemia. Polymorphism rs5219 in the KCNJ11 gene is a risk factor for developing T2D in humans. KCNJ11 encodes the ‘inward-rectifier potas...
- Published
- 2020
43. Hydrophobic gating and functional annotation of ion channels
- Author
-
Klesse, G, Tucker, S, and Sansom, M
- Subjects
Computational Structural Biology ,Computational Biochemistry ,Computational Biophysics - Abstract
Ion channels are integral membrane proteins that facilitate the permeation of ions through lipid bilayers. By means of a molecular mechanism known as hydrophobic gating, these proteins can close their conduction pathway to ion flow without the need for steric occlusion. Hydrophobic gates pose a challenge in the analysis of novel ion channel structures, as they cannot be detected by measuring the physical dimensions of a channel pore. This thesis therefore presents the Channel Annotation Package (CHAP), an open-source software tool for the functional annotation of ion channel structures based on molecular dynamics (MD) simulation and capable of reliably indentifying hydrophobic gates. The cation-selective 5-HT3 receptor and the anion-selective glycine receptor served as test cases to demonstrate the utility of CHAP. The functional status of several recent structures of these two channels was determined through MD simulation and hydrophobic gates were identified in both receptors. Furthermore, a high-throughput survey of ion channel structures enabled by CHAP revealed that hydrophobic gates occur in ~20% of the ion channel structural proteome. Next-generation polarisable force fields were used to explore the importance of electronic polarisation in MD simulations of ion channels. Water molecules passing a hydrophobic gate experienced a significant reduction of their dipole moment, mirroring the behaviour of water in a liquid-vapour phase transition. Polarisable ions interacted more strongly with uncharged protein residues than simulations employing traditional additive force fields suggest. Incorporating polarisation effects will thus be an important aspect of future computational studies of ion channel structure and function. The impact of a transmembrane potential on the hydrophobic gate in the 5-HT3 receptor was studied through atomistic MD simulations. Electrowetting occured only at supraphysiological voltages, while at lower field strengths pore de-wetting effectively prevented ion permeation. The hydration response of the channel could be quantitatively described by a thermodynamic model that includes the influence of the charged amino acid residues of the channel protein. Electric field effects are unlikely to play an important role for hydrophobic gating in biological ion channels, but may potentially be exploited in the design of gated artificial nanopores.
- Published
- 2020
44. OpenStructure: an integrated software framework for computational structural biology.
- Author
-
Biasini, M., Schmidt, T., Bienert, S., Mariani, V., Studer, G., Haas, J., Johner, N., Schenk, A. D., Philippsen, A., and Schwede, T.
- Subjects
- *
COMPUTATIONAL biology , *INTEGRATED software , *MOLECULAR structure , *PROTEOMICS , *SEQUENCE alignment , *PYTHON programming language - Abstract
Research projects in structural biology increasingly rely on combinations of heterogeneous sources of information, e.g. evolutionary information from multiple sequence alignments, experimental evidence in the form of density maps and proximity constraints from proteomics experiments. The OpenStructure software framework, which allows the seamless integration of information of different origin, has previously been introduced. The software consists of C++ libraries which are fully accessible from the Python programming language. Additionally, the framework provides a sophisticated graphics module that interactively displays molecular structures and density maps in three dimensions. In this work, the latest developments in the OpenStructure framework are outlined. The extensive capabilities of the framework will be illustrated using short code examples that show how information from molecular-structure coordinates can be combined with sequence data and/or density maps. The framework has been released under the LGPL version 3 license and is available for download from http://www.openstructure.org. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
45. Advances in integrative modeling of biomolecular complexes.
- Author
-
Karaca, Ezgi and Bonvin, Alexandre M.J.J.
- Subjects
- *
BIOMOLECULES , *SIMULATED annealing , *OVERHAUSER effect (Nuclear physics) , *MOLECULAR dynamics , *ION mobility , *ATOMIC force microscopy - Abstract
Abstract: High-resolution structural information is needed in order to unveil the underlying mechanistic of biomolecular function. Due to the technical limitations or the nature of the underlying complexes, acquiring atomic resolution information is difficult for many challenging systems, while, often, low-resolution biochemical or biophysical data can still be obtained. To make best use of all the available information and shed light on these challenging systems, integrative computational tools are required that can judiciously combine and accurately translate sparse experimental data into structural information. In this review we discuss the current state of integrative approaches, the challenges they are confronting and the advances made regarding those challenges. Recent developments are underpinned by noteworthy application examples taken from the literature. Within this context, we also position our data-driven docking approach, HADDOCK that can integrate a variety of information sources to drive the modeling of biomolecular complexes. Only a synergistic combination of experiment and modeling will allow us to tackle the challenges of adding the structural dimension to interactomes, shed “atomic” light onto molecular processes and understand the underlying mechanistic of biomolecular function. The current state of integrative approaches indicates that they are poised to take those challenges. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
46. Modeling Macromolecular Assemblies
- Author
-
Kim, Michael F.
- Subjects
Biology, Bioinformatics ,Biophysics, General ,computational structural biology - Abstract
Macromolecular assemblies are fundamental to most biological processes and here we attempt to improve the structural characterization of assemblies in the hopes that with new and improved models will produce functional insights on assemblies.Modeling of macromolecular assemblies begins with an analysis of the computational and experimental data available on the entire assembly, subcomplexes, individual subunits and the interactions between subunits. Having collected the data on the assembly, the next challenge is to integrate the disparate data to produce a structural model. Hybrid approaches, which integrate multiple sources of data, provide a way to increase the coverage and accuracy of structure modeling for macromolecular complexes.Fitting in with the theme of hybrid methods, Chapters 2 and 3 describe methods for modeling macromolecular assemblies, combining overall shape information (e.g., from cryo-electron microscopy) with interaction data (e.g., tandem affinity purification assays); and protein structures (or models) with NMR spectroscopy, respectively.Chapter 4 proposes an assessment strategy for structure modeling methods that provides a way to measure how much improvement is left to be made, instead of the traditional approach of measuring how much improvement was already made. This assessment strategy provides more information on the specific limitations of the method and provides specific insight into how to best improve the method. The strategy also presents a more fair method of comparing competing methods that are assessed with different benchmark sets.Chapter 5 describes the representation of subunits and assemblies by systems of points and restraints, explores the assumptions that underlie using points and restraints to model macromolecular structures, describes the properties of binary and multiple docking, and models the structure modeling framework. The main contributions of this dissertation are two practical approaches for macromolecular assemblies; an assessment strategy that provides a more explicit description of the accuracy and limitation of assessed methods, improving the confidence with which the resulting models are used; and lastly, a deeper theoretical understanding of modeling macromolecular assemblies, including a path towards a more principled approach for integrating multiple sources of data.
- Published
- 2008
47. Genome structure determination via 3C-based data integration by the Integrative Modeling Platform
- Author
-
Baù, Davide and Marti-Renom, Marc A.
- Subjects
- *
GENOMES , *GENES , *MOLECULAR biology , *MICROSCOPY , *CHROMOSOMES , *CHROMATIN - Abstract
Abstract: The three-dimensional (3D) architecture of a genome determines the spatial localization of regulatory elements and the genes they regulate. Thus, elucidating the 3D structure of a genome may result in significant insights about how genes are regulated. The current state-of-the art in experimental methods, including light microscopy and cell/molecular biology, are now able to provide detailed information on the position of genes and their interacting partners. However, such methods by themselves are not able to determine the high-resolution 3D structure of genomes or genomic domains. Here we describe a computational module of the Integrative Modeling Platform (IMP, http://www.integrativemodeling.org) that uses chromosome conformation capture data to determine the 3D architecture of genomic domains and entire genomes at unprecedented resolutions. This approach, through the visualization of looping interactions between distal regulatory elements, allows characterizing global chromatin features and their relation to gene expression. We illustrate our work by outlining the determination of the 3D architecture of the α-globin domain in the human genome. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
48. Structure determination of genomic domains by satisfaction of spatial restraints.
- Author
-
Baù, Davide and Marti-Renom, Marc
- Abstract
The three-dimensional (3D) architecture of a genome is non-random and known to facilitate the spatial colocalization of regulatory elements with the genes they regulate. Determining the 3D structure of a genome may therefore probe an essential step in characterizing how genes are regulated. Currently, there are several experimental and theoretical approaches that aim at determining the 3D structure of genomes and genomic domains; however, approaches integrating experiments and computation to identify the most likely 3D folding of a genome at medium to high resolutions have not been widely explored. Here, we review existing methodologies and propose that the integrative modeling platform (), a computational package developed for structurally characterizing protein assemblies, could be used for integrating diverse experimental data towards the determination of the 3D architecture of genomic domains and entire genomes at unprecedented resolution. Our approach, through the visualization of looping interactions between distal regulatory elements, will allow for the characterization of global chromatin features and their relation to gene expression. We illustrate our work by outlining the recent determination of the 3D architecture of the α-globin domain in the human genome. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
49. Structural Similarity Between Native Proteins and Chimera Constructs Obtained by Inverting the Amino Acid Sequence.
- Author
-
Carugo, Oliviero
- Subjects
- *
PROTEIN structure , *CHIMERISM , *AMINO acid sequence , *BIOINFORMATICS , *SYMMETRY (Physics) - Abstract
The analysis of the symmetry of protein three-dimensional structures can be extremely useful in order to understand and classify the protein structural universe. The structures of proteins with back-traced amino acid sequence were modeled and compared to the structures of their native counterparts. Only in a very limited set of cases, the two objects showed a significant level of similarity. These extremely symmetric examples can be of any structural class and of any dimension. The lack of biunique "N to C" and "C to N" symmetry at the structural level mirrors that at the sequence level and we propose to design as a dlof symmetry the cases in which a protein structure is similar to its back-traced variant. [ABSTRACT FROM AUTHOR]
- Published
- 2010
50. The long coming of computational structural biology
- Author
-
Lupas, Andrei N.
- Subjects
- *
MOLECULAR biology , *COLLAGEN , *ANTISENSE nucleic acids , *PROTEIN structure , *PROTEIN folding , *MOLECULAR structure - Abstract
Abstract: Fifty years ago, the structures of the α-helix, the β-sheet, the α-helical coiled coil and the collagen triple helix raised the expectation that protein structure could be understood computationally, using a combination of geometric considerations, model-building and parametric equations. The first crystal structures dispelled this hope, revealing a disconcerting lack of regularity in the folding patterns of proteins. Gradually it became clear that the protein folding problem—namely deducing the structure of a protein from its amino acid sequence—was of exceptional difficulty. Its solution has remained outside our reach to this day and, arguably, it represents the most important unsolved problem in molecular biology. Nevertheless, our ability to understand and predict molecular structure by computational means has made steady progress, suggesting that we will eventually conquer the problem, not by a few heroic insights, but by steady advances in biophysical knowledge, biological databases, software applications and raw computer power. Computational structural biology, whose influence is already pervasive, will come to dominate structural approaches in the next decades. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.