101
|
Peptidomimetics: A Synthetic Tool for Inhibiting Protein–Protein Interactions in Cancer. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-019-09831-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
102
|
Choi M, Baek J, Han SB, Cho S. Facile analysis of protein-protein interactions in living cells by enriched visualization of the p-body. BMB Rep 2019. [PMID: 29898808 PMCID: PMC6235090 DOI: 10.5483/bmbrep.2018.51.10.051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Protein-Protein Interactions (PPIs) play essential roles in diverse biological processes and their misregulations are associated with a wide range of diseases. Especially, the growing attention to PPIs as a new class of therapeutic target is increasing the need for an efficient method of cell-based PPI analysis. Thus, we newly developed a robust PPI assay (SeePPI) based on the co-translocation of interacting proteins to the discrete subcellular compartment ‘processing body’ (p-body) inside living cells, enabling a facile analysis of PPI by the enriched fluorescent signal. The feasibility and strength of SeePPI (Signal enhancement exclusively on P-body for Protein-protein Interaction) assay was firmly demonstrated with FKBP12/FRB interaction induced by rapamycin within seconds in real-time analysis of living cells, indicating its recapitulation of physiological PPI dynamics. In addition, we applied p53/MDM2 interaction and its dissociation by Nutlin-3 to SeePPI assay and further confirmed that SeePPI was quantitative and well reflected the endogenous PPI. Our SeePPI assay will provide another useful tool to achieve an efficient analysis of PPIs and their modulators in cells.
Collapse
Affiliation(s)
- Miri Choi
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116; College of Pharmacy, Chungbuk National University, Cheongju 28644, South Korea
| | - Jiyeon Baek
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116; College of Pharmacy, Chungbuk National University, Cheongju 28644, South Korea
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, Cheongju 28644, South Korea
| | - Sungchan Cho
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, South Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, South Korea
| |
Collapse
|
103
|
Hassan M, Shahzadi S, Raza H, Abbasi MA, Alashwal H, Zaki N, Moustafa AA, Seo SY. Computational investigation of mechanistic insights of Aβ42 interactions against extracellular domain of nAChRα7 in Alzheimer's disease. Int J Neurosci 2019; 129:666-680. [PMID: 30422726 DOI: 10.1080/00207454.2018.1543670] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
AIM Amyloid beta (Aβ) 1-42, which is a basic constituent of amyloid plaques, binds with extracellular transmembrane receptor nicotine acetylcholine receptor α7 (nAChRα7) in Alzheimer's disease. MATERIALS AND METHODS In the current study, a computational approach was employed to explore the active binding sites of nAChRα7 through Aβ 1-42 interactions and their involvement in the activation of downstream signalling pathways. Sequential and structural analyses were performed on the extracellular part of nAChRα7 to identify its core active binding site. RESULTS Results showed that a conserved residual pattern and well superimposed structures were observed in all nAChRs proteins. Molecular docking servers were used to predict the common interactive residues in nAChRα7 and Aβ1-42 proteins. The docking profile results showed some common interactive residues such as Glu22, Ala42 and Trp171 may consider as the active key player in the activation of downstream signalling pathways. Moreover, the signal communication and receiving efficacy of best-docked complexes was checked through DynOmic online server. Furthermore, the results from molecular dynamic simulation experiment showed the stability of nAChRα7. The generated root mean square deviations and fluctuations (RMSD/F), solvent accessible surface area (SASA) and radius of gyration (Rg) graphs of nAChRα7 also showed its backbone stability and compactness, respectively. CONCLUSION Taken together, our predicted results intimated the structural insight on the molecular interactions of beta amyloid protein involved in the activation of nAChRα7 receptor. In future, a better understanding of nAChRα7 and their interconnected proteins signalling cascade may be consider as target to cure Alzheimer's disease.
Collapse
Affiliation(s)
- Mubashir Hassan
- a Department of Biological Sciences, College of Natural Sciences , Kongju National University , Gongju , Chungnam-do 32588 , Republic of Korea
| | - Saba Shahzadi
- b Institute of Molecular Science and Bioinformatics , Lahore , Pakistan.,c Department of Bioinformatics , Virtual University of Pakistan , Lahore , Pakistan
| | - Hussain Raza
- a Department of Biological Sciences, College of Natural Sciences , Kongju National University , Gongju , Chungnam-do 32588 , Republic of Korea
| | - Muhammad Athar Abbasi
- a Department of Biological Sciences, College of Natural Sciences , Kongju National University , Gongju , Chungnam-do 32588 , Republic of Korea.,d Department of Chemistry , Government College University , Lahore , Pakistan
| | - Hany Alashwal
- e Department of Computer Science and Software Engineering, College of Information Technology , United Arab Emirates University , Al-Ain , United Arab Emirates
| | - Nazar Zaki
- e Department of Computer Science and Software Engineering, College of Information Technology , United Arab Emirates University , Al-Ain , United Arab Emirates
| | - Ahmed A Moustafa
- f School of Social Sciences and Psychology.,g MARCS Institute for Brain and Behaviour , Western Sydney University , Sydney , New South Wales , Australia.,h Department of Social Sciences, College of Arts and Sciences , Qatar University , Doha , Qatar'
| | - Sung-Yum Seo
- a Department of Biological Sciences, College of Natural Sciences , Kongju National University , Gongju , Chungnam-do 32588 , Republic of Korea
| |
Collapse
|
104
|
Huang D, Wen W, Liu X, Li Y, Zhang JZH. Computational analysis of hot spots and binding mechanism in the PD-1/PD-L1 interaction. RSC Adv 2019; 9:14944-14956. [PMID: 35516311 PMCID: PMC9064197 DOI: 10.1039/c9ra01369e] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 05/05/2019] [Indexed: 12/14/2022] Open
Abstract
Programmed cell death protein-1 (PD-1) is an important immunological checkpoint and plays a vital role in maintaining the peripheral tolerance of the human body by interacting with its ligand PD-L1. The overexpression of PD-L1 in tumor cells induces local immune suppression and helps the tumor cells to evade the endogenous anti-tumor immunity. Developing monoclonal antibodies against the PD-1/PD-L1 protein–protein interaction to block the PD-1/PD-L1 signaling pathway has demonstrated superior anti-tumor efficacy in a variety of solid tumors and has made a profound impact on the field of cancer immunotherapy in recent years. Although the X-ray crystal structure of the PD-1/PD-L1 complex has been solved, the detailed binding mechanism of the PD-1/PD-L1 interaction is not fully understood from a theoretical point of view. In this study, we performed computational alanine scanning on the PD-1/PD-L1 complex to quantitatively identify the hot spots in the PD-1/PD-L1 interaction and characterize its binding mechanisms at the atomic level. To the best of our knowledge, this is the first time that theoretical calculations have been used to systematically and quantitatively predict the hot spots in the PD-1/PD-L1 interaction. We hope that the predicted hot spots and the energy profile of the PD-1/PD-L1 interaction presented in this work can provide guidance for the design of peptide and small molecule drugs targeting PD-1 or PD-L1. The hot spots quantitatively predicted by the recently developed MM/GBSA/IE method reveal a hydrophobic core in the PD-1/PD-L1 interaction.![]()
Collapse
Affiliation(s)
- Dading Huang
- State Key Laboratory for Precision Spectroscopy
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200062
| | - Wei Wen
- State Key Laboratory for Precision Spectroscopy
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200062
| | - Xiao Liu
- State Key Laboratory for Precision Spectroscopy
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200062
| | - Yang Li
- State Key Laboratory for Precision Spectroscopy
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200062
| | - John Z. H. Zhang
- State Key Laboratory for Precision Spectroscopy
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200062
| |
Collapse
|
105
|
Application of Support Vector Machines in Viral Biology. GLOBAL VIROLOGY III: VIROLOGY IN THE 21ST CENTURY 2019. [PMCID: PMC7114997 DOI: 10.1007/978-3-030-29022-1_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Novel experimental and sequencing techniques have led to an exponential explosion and spiraling of data in viral genomics. To analyse such data, rapidly gain information, and transform this information to knowledge, interdisciplinary approaches involving several different types of expertise are necessary. Machine learning has been in the forefront of providing models with increasing accuracy due to development of newer paradigms with strong fundamental bases. Support Vector Machines (SVM) is one such robust tool, based rigorously on statistical learning theory. SVM provides very high quality and robust solutions to classification and regression problems. Several studies in virology employ high performance tools including SVM for identification of potentially important gene and protein functions. This is mainly due to the highly beneficial aspects of SVM. In this chapter we briefly provide lucid and easy to understand details of SVM algorithms along with applications in virology.
Collapse
|
106
|
Ivanov SM, Huber RG, Alibay I, Warwicker J, Bond PJ. Energetic Fingerprinting of Ligand Binding to Paralogous Proteins: The Case of the Apoptotic Pathway. J Chem Inf Model 2018; 59:245-261. [DOI: 10.1021/acs.jcim.8b00765] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Stefan M. Ivanov
- Manchester Institute of Biotechnology, School of Chemistry, The University of Manchester, 131 Princess Street, Manchester M1 7DN, U.K
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Matrix 07-01, 30 Biopolis Street, Singapore 138671, Singapore
| | - Roland G. Huber
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Matrix 07-01, 30 Biopolis Street, Singapore 138671, Singapore
| | - Irfan Alibay
- Division of Pharmacy and Optometry, School of Health Sciences, The University of Manchester, Oxford Road, Manchester M13 9PT, U.K
| | - Jim Warwicker
- Manchester Institute of Biotechnology, School of Chemistry, The University of Manchester, 131 Princess Street, Manchester M1 7DN, U.K
| | - Peter J. Bond
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Matrix 07-01, 30 Biopolis Street, Singapore 138671, Singapore
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore
| |
Collapse
|
107
|
Huang D, Qi Y, Song J, Zhang JZH. Calculation of hot spots for protein–protein interaction in p53/PMI‐MDM2/MDMX complexes. J Comput Chem 2018; 40:1045-1056. [DOI: 10.1002/jcc.25592] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/04/2018] [Accepted: 08/23/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Dading Huang
- School of Physics and Material Science, Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular EngineeringEast China Normal University Shanghai 200062 China
| | - Yifei Qi
- School of Physics and Material Science, Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular EngineeringEast China Normal University Shanghai 200062 China
- NYU‐ECNU Center for Computational Chemistry at NYU Shanghai Shanghai 200062 China
| | - Jianing Song
- NYU‐ECNU Center for Computational Chemistry at NYU Shanghai Shanghai 200062 China
| | - John Z. H. Zhang
- School of Physics and Material Science, Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular EngineeringEast China Normal University Shanghai 200062 China
- NYU‐ECNU Center for Computational Chemistry at NYU Shanghai Shanghai 200062 China
- Department of ChemistryNew York University New York New York, 10003
- Collaborative Innovation Center of Extreme OpticsShanxi University Taiyuan Shanxi, 030006 China
| |
Collapse
|
108
|
Capriotti E, Ozturk K, Carter H. Integrating molecular networks with genetic variant interpretation for precision medicine. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2018; 11:e1443. [PMID: 30548534 PMCID: PMC6450710 DOI: 10.1002/wsbm.1443] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/23/2018] [Accepted: 10/30/2018] [Indexed: 02/01/2023]
Abstract
More reliable and cheaper sequencing technologies have revealed the vast mutational landscapes characteristic of many phenotypes. The analysis of such genetic variants has led to successful identification of altered proteins underlying many Mendelian disorders. Nevertheless the simple one‐variant one‐phenotype model valid for many monogenic diseases does not capture the complexity of polygenic traits and disorders. Although experimental and computational approaches have improved detection of functionally deleterious variants and important interactions between gene products, the development of comprehensive models relating genotype and phenotypes remains a challenge in the field of genomic medicine. In this context, a new view of the pathologic state as significant perturbation of the network of interactions between biomolecules is crucial for the identification of biochemical pathways associated with complex phenotypes. Seminal studies in systems biology combined the analysis of genetic variation with protein–protein interaction networks to demonstrate that even as biological systems evolve to be robust to genetic variation, their topologies create disease vulnerabilities. More recent analyses model the impact of genetic variants as changes to the “wiring” of the interactome to better capture heterogeneity in genotype–phenotype relationships. These studies lay the foundation for using networks to predict variant effects at scale using machine‐learning or algorithmic approaches. A wealth of databases and resources for the annotation of genotype–phenotype relationships have been developed to support developments in this area. This overview describes how study of the molecular interactome has generated insights linking the organization of biological systems to disease mechanism, and how this information can enable precision medicine. This article is categorized under:
Translational, Genomic, and Systems Medicine > Translational Medicine Biological Mechanisms > Cell Signaling Models of Systems Properties and Processes > Mechanistic Models Analytical and Computational Methods > Computational Methods
Collapse
Affiliation(s)
- Emidio Capriotti
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy
| | - Kivilcim Ozturk
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, California
| | - Hannah Carter
- Department of Medicine and Institute for Genomic Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
109
|
Dobson L, Mészáros B, Tusnády GE. Structural Principles Governing Disease-Causing Germline Mutations. J Mol Biol 2018; 430:4955-4970. [DOI: 10.1016/j.jmb.2018.10.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 10/11/2018] [Indexed: 01/03/2023]
|
110
|
Shi X, Liu Y, Zhao R, Li Z. Constructing Thioether/Vinyl Sulfide-tethered Helical Peptides Via Photo-induced Thiol-ene/yne Hydrothiolation. J Vis Exp 2018. [PMID: 30124641 DOI: 10.3791/57356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Here, we describe a detailed protocol for the preparation of thioether-tethered peptides using on-resin intramolecular/intermolecular thiol-ene hydrothiolation. In addition, this protocol describes the preparation of vinyl-sulfide-tethered peptides using in-solution intramolecular thiol-yne hydrothiolation between amino acids that possess alkene/alkyne side chains and cysteine residues at i, i+4 positions. Linear peptides were synthesized using a standard Fmoc-based solid-phase peptide synthesis (SPPS). Thiol-ene hydrothiolation is carried out using either an intramolecular thio-ene reaction or an intermolecular thio-ene reaction, depending on the peptide length. In this research, an intramolecular thio-ene reaction is carried out in the case of shorter peptides using on-resin deprotection of the trityl groups of cysteine residues following the complete synthesis of the linear peptide. The resin is then set to UV irradiation using photoinitiator 4-methoxyacetophenone (MAP) and 2-hydroxy-1-[4-(2-hydroxyethoxy)-phenyl]-2-methyl-1-propanone (MMP). The intermolecular thiol-ene reaction is carried out by dissolving Fmoc-Cys-OH in an N,N-dimethylformamide (DMF) solvent. This is then reacted with the peptide using the alkene-bearing residue on resin. After that, the macrolactamization is carried out using benzotriazole-1-yl-oxytripyrrolidinophosphonium hexafluorophosphate (PyBop), 1-hydroxybenzotriazole (HoBt), and 4-Methylmorpholine (NMM) as activation reagents on the resin. Following the macrolactamization, the peptide synthesis is continued using standard SPPS. In the case of the thio-yne hydrothiolation, the linear peptide is cleaved from the resin, dried, and subsequently dissolved in degassed DMF. This is then irradiated using UV light with photoinitiator 2,2-dimethoxy-2-phenylacetophenone (DMPA). Following the reaction, DMF is evaporated and the crude residue is precipitated and purified using high-performance liquid chromatography (HPLC). These methods could function to simplify the generation of thioether-tethered cyclic peptides due to the use of the thio-ene/yne click chemistry that possesses superior functional group tolerance and good yield. The introduction of thioether bonds into peptides takes advantage of the nucleophilic nature of cysteine residues and is redox-inert relative to disulfide bonds.
Collapse
Affiliation(s)
- Xiaodong Shi
- Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School
| | - Yinghuan Liu
- Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School
| | - Rongtong Zhao
- Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School
| | - Zigang Li
- Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School;
| |
Collapse
|
111
|
Banu H, Joseph MC, Nisar MN. In-silico approach to investigate death domains associated with nano-particle-mediated cellular responses. Comput Biol Chem 2018; 75:11-23. [DOI: 10.1016/j.compbiolchem.2018.04.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 04/01/2018] [Accepted: 04/21/2018] [Indexed: 11/29/2022]
|
112
|
Amiri Dash Atan N, Koushki M, Rezaei Tavirani M, Ahmadi NA. Protein-Protein Interaction Network Analysis of Salivary Proteomic Data in Oral Cancer Cases. Asian Pac J Cancer Prev 2018; 19:1639-1645. [PMID: 29937423 PMCID: PMC6103602 DOI: 10.22034/apjcp.2018.19.6.1639] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Background: Oral cancer is a frequently encountered neoplasm of the head and neck region, being the eight most common type of human malignancy worldwide. Despite improvement in its control, morbidity and mortality rates have improved little in the past decades. Therefore, prevention and/or early detection are a high priority. Proteomics with network analysis have emerged as a powerful tool to identify important proteins associated with cancer development and progression that can be potential targets for early diagnosis. In the present study, network- based protein- protein interactions (PPI) for oral cancer were identified and then analyzed for use as key proteins/potential biomarkers. Material and Methods: Gene expression data in articles which focused on saliva proteomics of oral cancer were collected and 74 candidate genes or proteins were extracted. Related protein networks of differentially expressed proteins were explored and visualized using cytoscape software. Further PPI analysis was performed by Molecular Complex Detection (MCODE) and BiNGO methods. Results: Network analysis of genes/proteins related to oral cancer identified kininogen-1, angiotensinogen, annexin A1, IL-8, IgG heavy variable and constant chains, CRP, collagen alpha-1 and fibronectin as 9 hub-bottleneck proteins. In addition, based on clustering with the MCODE tool, vitronectin, collagen alpha-2, IL-8 and integrin alpha-v were established as 5 distinct seed proteins. Conclusion: A hub-bottleneck protein panel may offer a potential /candidate biomarker pattern for diagnosis and treatment of oral cancer disease. Further investigation and validation of these proteins are warranted.
Collapse
Affiliation(s)
- Nasrin Amiri Dash Atan
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | | | | | |
Collapse
|
113
|
Exploring the mechanistic insights of Cas scaffolding protein family member 4 with protein tyrosine kinase 2 in Alzheimer's disease by evaluating protein interactions through molecular docking and dynamic simulations. Neurol Sci 2018; 39:1361-1374. [PMID: 29789968 DOI: 10.1007/s10072-018-3430-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 04/26/2018] [Indexed: 01/02/2023]
Abstract
Cas scaffolding protein family member 4 and protein tyrosine kinase 2 are signaling proteins, which are involved in neuritic plaques burden, neurofibrillary tangles, and disruption of synaptic connections in Alzheimer's disease. In the current study, a computational approach was employed to explore the active binding sites of Cas scaffolding protein family member 4 and protein tyrosine kinase 2 proteins and their significant role in the activation of downstream signaling pathways. Sequential and structural analyses were performed on Cas scaffolding protein family member 4 and protein tyrosine kinase 2 to identify their core active binding sites. Molecular docking servers were used to predict the common interacting residues in both Cas scaffolding protein family member 4 and protein tyrosine kinase 2 and their involvement in Alzheimer's disease-mediated pathways. Furthermore, the results from molecular dynamic simulation experiment show the stability of targeted proteins. In addition, the generated root mean square deviations and fluctuations, solvent-accessible surface area, and gyration graphs also depict their backbone stability and compactness, respectively. A better understanding of CAS and their interconnected protein signaling cascade may help provide a treatment for Alzheimer's disease. Further, Cas scaffolding protein family member 4 could be used as a novel target for the treatment of Alzheimer's disease by inhibiting the protein tyrosine kinase 2 pathway.
Collapse
|
114
|
Robertson NS, Spring DR. Using Peptidomimetics and Constrained Peptides as Valuable Tools for Inhibiting Protein⁻Protein Interactions. Molecules 2018; 23:molecules23040959. [PMID: 29671834 PMCID: PMC6017787 DOI: 10.3390/molecules23040959] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/16/2018] [Accepted: 04/18/2018] [Indexed: 02/07/2023] Open
Abstract
Protein–protein interactions (PPIs) are tremendously important for the function of many biological processes. However, because of the structure of many protein–protein interfaces (flat, featureless and relatively large), they have largely been overlooked as potential drug targets. In this review, we highlight the current tools used to study the molecular recognition of PPIs through the use of different peptidomimetics, from small molecules and scaffolds to peptides. Then, we focus on constrained peptides, and in particular, ways to constrain α-helices through stapling using both one- and two-component techniques.
Collapse
Affiliation(s)
- Naomi S Robertson
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| | - David R Spring
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| |
Collapse
|
115
|
Slough DP, McHugh SM, Cummings AE, Dai P, Pentelute BL, Kritzer JA, Lin YS. Designing Well-Structured Cyclic Pentapeptides Based on Sequence-Structure Relationships. J Phys Chem B 2018; 122:3908-3919. [PMID: 29589926 PMCID: PMC6071411 DOI: 10.1021/acs.jpcb.8b01747] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cyclic peptides are a promising class of molecules for unique applications. Unfortunately, cyclic peptide design is severely limited by the difficulty in predicting the conformations they will adopt in solution. In this work, we use explicit-solvent molecular dynamics simulations to design well-structured cyclic peptides by studying their sequence-structure relationships. Critical to our approach is an enhanced sampling method that exploits the essential transitional motions of cyclic peptides to efficiently sample their conformational space. We simulated a range of cyclic pentapeptides from all-glycine to a library of cyclo-(X1X2AAA) peptides to map their conformational space and determine cooperative effects of neighboring residues. By combining the results from all cyclo-(X1X2AAA) peptides, we developed a scoring function to predict the structural preferences for X1-X2 residues within cyclic pentapeptides. Using this scoring function, we designed a cyclic pentapeptide, cyclo-(GNSRV), predicted to be well structured in aqueous solution. Subsequent circular dichroism and NMR spectroscopy revealed that this cyclic pentapeptide is indeed well structured in water, with a nuclear Overhauser effect and J-coupling values consistent with the predicted structure.
Collapse
Affiliation(s)
- Diana P. Slough
- Department of Chemistry, Tufts University, Medford, Massachusetts 02155, USA
| | - Sean M. McHugh
- Department of Chemistry, Tufts University, Medford, Massachusetts 02155, USA
| | | | - Peng Dai
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Bradley L. Pentelute
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Joshua A. Kritzer
- Department of Chemistry, Tufts University, Medford, Massachusetts 02155, USA
| | - Yu -Shan Lin
- Department of Chemistry, Tufts University, Medford, Massachusetts 02155, USA
| |
Collapse
|
116
|
Mei S. In Silico Enhancing M. tuberculosis Protein Interaction Networks in STRING To Predict Drug-Resistance Pathways and Pharmacological Risks. J Proteome Res 2018; 17:1749-1760. [PMID: 29611419 DOI: 10.1021/acs.jproteome.7b00702] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Bacterial protein-protein interaction (PPI) networks are significant to reveal the machinery of signal transduction and drug resistance within bacterial cells. The database STRING has collected a large number of bacterial pathogen PPI networks, but most of the data are of low quality without being experimentally or computationally validated, thus restricting its further biomedical applications. We exploit the experimental data via four solutions to enhance the quality of M. tuberculosis H37Rv (MTB) PPI networks in STRING. Computational results show that the experimental data derived jointly by two-hybrid and copurification approaches are the most reliable to train an L2-regularized logistic regression model for MTB PPI network validation. On the basis of the validated MTB PPI networks, we further study the three problems via breadth-first graph search algorithm: (1) discovery of MTB drug-resistance pathways through searching for the paths between known drug-target genes and drug-resistance genes, (2) choosing potential cotarget genes via searching for the critical genes located on multiple pathways, and (3) choosing essential drug-target genes via analysis of network degree distribution. In addition, we further combine the validated MTB PPI networks with human PPI networks to analyze the potential pharmacological risks of known and candidate drug-target genes from the point of view of system pharmacology. The evidence from protein structure alignment demonstrates that the drugs that act on MTB target genes could also adversely act on human signaling pathways.
Collapse
Affiliation(s)
- Suyu Mei
- Software College , Shenyang Normal University , Shenyang 110034 , China
| |
Collapse
|
117
|
MultiBacMam Bimolecular Fluorescence Complementation (BiFC) tool-kit identifies new small-molecule inhibitors of the CDK5-p25 protein-protein interaction (PPI). Sci Rep 2018; 8:5083. [PMID: 29572554 PMCID: PMC5865166 DOI: 10.1038/s41598-018-23516-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 03/14/2018] [Indexed: 11/10/2022] Open
Abstract
Protein-protein interactions (PPIs) are at the core of virtually all biological processes in cells. Consequently, targeting PPIs is emerging at the forefront of drug discovery. Cellular assays which closely recapitulate native conditions in vivo are instrumental to understand how small molecule drugs can modulate such interactions. We have integrated MultiBacMam, a baculovirus-based mammalian gene delivery tool we developed, with bimolecular fluorescence complementation (BiFC), giving rise to a highly efficient system for assay development, identification and characterization of PPI modulators. We used our system to analyze compounds impacting on CDK5-p25 PPI, which is implicated in numerous diseases including Alzheimer’s. We evaluated our tool-kit with the known inhibitor p5T, and we established a mini-screen to identify compounds that modulate this PPI in dose-response experiments. Finally, we discovered several compounds disrupting CDK5-p25 PPI, which had not been identified by other screening or structure-based methods before.
Collapse
|
118
|
Jalili M, Gebhardt T, Wolkenhauer O, Salehzadeh-Yazdi A. Unveiling network-based functional features through integration of gene expression into protein networks. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2349-2359. [PMID: 29466699 DOI: 10.1016/j.bbadis.2018.02.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 01/31/2018] [Accepted: 02/13/2018] [Indexed: 02/02/2023]
Abstract
Decoding health and disease phenotypes is one of the fundamental objectives in biomedicine. Whereas high-throughput omics approaches are available, it is evident that any single omics approach might not be adequate to capture the complexity of phenotypes. Therefore, integrated multi-omics approaches have been used to unravel genotype-phenotype relationships such as global regulatory mechanisms and complex metabolic networks in different eukaryotic organisms. Some of the progress and challenges associated with integrated omics studies have been reviewed previously in comprehensive studies. In this work, we highlight and review the progress, challenges and advantages associated with emerging approaches, integrating gene expression and protein-protein interaction networks to unravel network-based functional features. This includes identifying disease related genes, gene prioritization, clustering protein interactions, developing the modules, extract active subnetworks and static protein complexes or dynamic/temporal protein complexes. We also discuss how these approaches contribute to our understanding of the biology of complex traits and diseases. This article is part of a Special Issue entitled: Cardiac adaptations to obesity, diabetes and insulin resistance, edited by Professors Jan F.C. Glatz, Jason R.B. Dyck and Christine Des Rosiers.
Collapse
Affiliation(s)
- Mahdi Jalili
- Hematology, Oncology and SCT Research Center, Tehran University of Medical Sciences, Tehran, Iran; Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Tom Gebhardt
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051 Rostock, Germany
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051 Rostock, Germany
| | - Ali Salehzadeh-Yazdi
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051 Rostock, Germany.
| |
Collapse
|
119
|
Díaz-Eufracio BI, Palomino-Hernández O, Houghten RA, Medina-Franco JL. Exploring the chemical space of peptides for drug discovery: a focus on linear and cyclic penta-peptides. Mol Divers 2018; 22:259-267. [PMID: 29446006 DOI: 10.1007/s11030-018-9812-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 01/23/2018] [Indexed: 10/18/2022]
Abstract
Peptide and peptide-like structures are regaining attention in drug discovery. Previous studies suggest that bioactive peptides have diverse structures and may have physicochemical properties attractive to become hit and lead compounds. However, chemoinformatic studies that characterize such diversity are limited. Herein, we report the physicochemical property profile and chemical space of four synthetic linear and cyclic combinatorial peptide libraries. As a case study, the analysis was focused on penta-peptides. The chemical space of the peptide and N-methylated peptides libraries was compared to compound data sets of pharmaceutical relevance. Results indicated that there is a major overlap in the chemical space of N-methylated cyclic peptides with inhibitors of protein-protein interactions and macrocyclic natural products available for screening. Also, there is an overlap between the chemical space of the synthetic peptides with peptides approved for clinical use (or in clinical trials), and to other approved drugs that are outside the traditional chemical space. Results further support that synthetic penta-peptides are suitable compounds to be used in drug discovery projects.
Collapse
Affiliation(s)
- Bárbara I Díaz-Eufracio
- School of Chemistry, Department of Pharmacy, Universidad Nacional Autónoma de México, Avenida Universidad 3000, 04510, Mexico City, Mexico
| | - Oscar Palomino-Hernández
- School of Chemistry, Department of Pharmacy, Universidad Nacional Autónoma de México, Avenida Universidad 3000, 04510, Mexico City, Mexico
| | - Richard A Houghten
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, FL, 34987, USA
| | - José L Medina-Franco
- School of Chemistry, Department of Pharmacy, Universidad Nacional Autónoma de México, Avenida Universidad 3000, 04510, Mexico City, Mexico.
| |
Collapse
|
120
|
Saberi Anvar M, Minuchehr Z, Shahlaei M, Kheitan S. Gastric cancer biomarkers; A systems biology approach. Biochem Biophys Rep 2018; 13:141-146. [PMID: 29556568 PMCID: PMC5857180 DOI: 10.1016/j.bbrep.2018.01.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/12/2017] [Accepted: 01/03/2018] [Indexed: 12/22/2022] Open
Abstract
Gastric cancer is one of the most fatal cancers in the world. Many efforts in recent years have attempted to find effective proteins in gastric cancer. By using a comprehensive list of proteins involved in gastric cancer, scientists were able to retrieve interaction information. The study of protein-protein interaction networks through systems biology based analysis provides appropriate strategies to discover candidate proteins and key biological pathways. In this study, we investigated dominant functional themes and centrality parameters including betweenness as well as the degree of each topological clusters and expressionally active sub-networks in the resulted network. The results of functional analysis on gene sets showed that neurotrophin signaling pathway, cell cycle and nucleotide excision possess the strongest enrichment signals. According to the computed centrality parameters, HNF4A, TAF1 and TP53 manifested as the most significant nodes in the interaction network of the engaged proteins in gastric cancer. This study also demonstrates pathways and proteins that are applicable as diagnostic markers and therapeutic targets for future attempts to overcome gastric cancer. A systematic study of protein-protein interaction networks through comprehensive extracted list of proteins involved in gastric cancer. Dominant functional theme and pathways of each topological clusters and expressionally active subnetworks were reported. The most effective proteins in gastric cancer formation were proposed according to the computed centrality parameters. HNF4A, TAF1and TP53 were mentioned as the key proteins in gastric cancer.
Collapse
Affiliation(s)
- Mohammad Saberi Anvar
- Department of Systems Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Zarrin Minuchehr
- Department of Systems Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mohsen Shahlaei
- Nano Drug Delivery Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Samira Kheitan
- Department of Systems Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
121
|
Bhojoo U, Biggar KK. Single-step purification of intrinsic protein complexes in Saccharomyces cerevisiae using regenerable calmodulin resin. MethodsX 2018; 5:613-619. [PMID: 29922592 PMCID: PMC6005797 DOI: 10.1016/j.mex.2018.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 06/10/2018] [Indexed: 12/03/2022] Open
Abstract
Characterization of protein- protein interactions is a vital aspect of molecular biology as multi-protein complexes are the functional units of cellular processes and defining their interactions provides valuable insights into their function based on a “guilt by association” concept. To identify the components in complexes, purification of the latter near homogeneity is required. The tandem affinity purification (i.e., TAP) method, coupled with mass spectrometry have been extensively used to define native protein complexes and transient protein-protein interactions under near physiological conditions (i.e., conditions approximate of the internal milieu) in Saccharomyces cerevisiae. Generally, TAP consists of two-stage protein enrichment using dual affinity tags, a calmodulin-binding peptide and a Staphylococcus aureus protein-A, separated by a tobacco etch virus protease site, which are fused to either the C- or N-terminal of the target protein. TAP-tagging has proved to be a powerful method for studying functional relationship between proteins and generating large-scale protein networks. The method described in this paper provides an inexpensive single-step purification alternative to the traditional two step affinity purification of TAP-tagged proteins using only the calmodulin-binding peptide affinity tag. Moreover, a novel protocol for the regeneration of the calmodulin-agarose resin is outlined and validated. This basic approach allows fast and cost-effective purification of proteins and their interacting partners from Saccharomyces cerevisiae.
Collapse
|
122
|
Vyas R, Bapat S, Goel P, Karthikeyan M, Tambe SS, Kulkarni BD. Application of Genetic Programming (GP) Formalism for Building Disease Predictive Models from Protein-Protein Interactions (PPI) Data. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2018; 15:27-37. [PMID: 28113781 DOI: 10.1109/tcbb.2016.2621042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Protein-protein interactions (PPIs) play a vital role in the biological processes involved in the cell functions and disease pathways. The experimental methods known to predict PPIs require tremendous efforts and the results are often hindered by the presence of a large number of false positives. Herein, we demonstrate the use of a new Genetic Programming (GP) based Symbolic Regression (SR) approach for predicting PPIs related to a disease. In a case study, a dataset consisting of one hundred and thirty five PPI complexes related to cancer was used to construct a generic PPI predicting model with good PPI prediction accuracy and generalization ability. A high correlation coefficient(CC) of 0.893, low root mean square error (RMSE) and mean absolute percentage error (MAPE) values of 478.221 and 0.239, respectively were achieved for both the training and test set outputs. To validate the discriminatory nature of the model, it was applied on a dataset of diabetes complexes where it yielded significantly low CC values. Thus, the GP model developed here serves a dual purpose: (a)a predictor of the binding energy of cancer related PPI complexes, and (b)a classifier for discriminating PPI complexes related to cancer from those of other diseases.
Collapse
|
123
|
Shin WH, Christoffer CW, Kihara D. In silico structure-based approaches to discover protein-protein interaction-targeting drugs. Methods 2017; 131:22-32. [PMID: 28802714 PMCID: PMC5683929 DOI: 10.1016/j.ymeth.2017.08.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/08/2017] [Accepted: 08/08/2017] [Indexed: 02/07/2023] Open
Abstract
A core concept behind modern drug discovery is finding a small molecule that modulates a function of a target protein. This concept has been successfully applied since the mid-1970s. However, the efficiency of drug discovery is decreasing because the druggable target space in the human proteome is limited. Recently, protein-protein interaction (PPI) has been identified asan emerging target space for drug discovery. PPI plays a pivotal role in biological pathways including diseases. Current human interactome research suggests that the number of PPIs is between 130,000 and 650,000, and only a small number of them have been targeted as drug targets. For traditional drug targets, in silico structure-based methods have been successful in many cases. However, their performance suffers on PPI interfaces because PPI interfaces are different in five major aspects: From a geometric standpoint, they have relatively large interface regions, flat geometry, and the interface surface shape tends to fluctuate upon binding. Also, their interactions are dominated by hydrophobic atoms, which is different from traditional binding-pocket-targeted drugs. Finally, PPI targets usually lack natural molecules that bind to the target PPI interface. Here, we first summarize characteristics of PPI interfaces and their known binders. Then, we will review existing in silico structure-based approaches for discovering small molecules that bind to PPI interfaces.
Collapse
Affiliation(s)
- Woong-Hee Shin
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | | | - Daisuke Kihara
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; Department of Computer Science, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
124
|
Miryala SK, Anbarasu A, Ramaiah S. Discerning molecular interactions: A comprehensive review on biomolecular interaction databases and network analysis tools. Gene 2017; 642:84-94. [PMID: 29129810 DOI: 10.1016/j.gene.2017.11.028] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/17/2017] [Accepted: 11/08/2017] [Indexed: 12/12/2022]
Abstract
Computational analysis of biomolecular interaction networks is now gaining a lot of importance to understand the functions of novel genes/proteins. Gene interaction (GI) network analysis and protein-protein interaction (PPI) network analysis play a major role in predicting the functionality of interacting genes or proteins and gives an insight into the functional relationships and evolutionary conservation of interactions among the genes. An interaction network is a graphical representation of gene/protein interactome, where each gene/protein is a node, and interaction between gene/protein is an edge. In this review, we discuss the popular open source databases that serve as data repositories to search and collect protein/gene interaction data, and also tools available for the generation of interaction network, visualization and network analysis. Also, various network analysis approaches like topological approach and clustering approach to study the network properties and functional enrichment server which illustrates the functions and pathway of the genes and proteins has been discussed. Hence the distinctive attribute mentioned in this review is not only to provide an overview of tools and web servers for gene and protein-protein interaction (PPI) network analysis but also to extract useful and meaningful information from the interaction networks.
Collapse
Affiliation(s)
- Sravan Kumar Miryala
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| | - Anand Anbarasu
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| | - Sudha Ramaiah
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India.
| |
Collapse
|
125
|
Smith JK, Jiang S, Pfaendtner J. Redefining the Protein-Protein Interface: Coarse Graining and Combinatorics for an Improved Understanding of Amino Acid Contributions to the Protein-Protein Binding Affinity. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017; 33:11511-11517. [PMID: 28850233 DOI: 10.1021/acs.langmuir.7b02438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The ability to intervene in biological pathways has for decades been limited by the lack of a quantitative description of protein-protein interactions (PPIs). Herein we generate and compare millions of simple PPI models for insight into the mechanisms of specific recognition and binding. We use a coarse-grained approach whereby amino acids are counted in the interface, and these counts are used as binding affinity predictors. We perform lasso regression, a modern regression technique aimed at interpretability, with every possible amino acid combination (over 106 unique feature sets) to select only those amino acid predictors that provide more information than noise. This approach circumvents arbitrary binning and assumptions about the binding environment that obscure other binding affinity models. Aggregated analysis of these models trained at various interfacial cutoff distances informs the roles of specific amino acids in different binding contexts. We find that a simple amino acid count model outperforms detailed intermolecular contact and binned residue type models. We identify the prevalence of serine, glycine, and tryptophan in the interface as particularly important for predicting binding affinity across a range of distance cutoffs. Although current sample size limitations prevent a robust consensus model for binding affinity prediction, our approach underscores the relevance of a residue-based description of the protein-protein interface to increase our understanding of specific interactions.
Collapse
Affiliation(s)
- Josh K Smith
- Department of Chemical Engineering, University of Washington , Seattle, Washington 98195, United States
| | - Shaoyi Jiang
- Department of Chemical Engineering, University of Washington , Seattle, Washington 98195, United States
| | - Jim Pfaendtner
- Department of Chemical Engineering, University of Washington , Seattle, Washington 98195, United States
| |
Collapse
|
126
|
Protein-Protein Interaction Modulators for Epigenetic Therapies. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2017; 110:65-84. [PMID: 29413000 DOI: 10.1016/bs.apcsb.2017.06.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Targeting protein-protein interactions (PPIs) is becoming an attractive approach for drug discovery. This is particularly true for difficult or emerging targets, such as epitargets that may be elusive to drugs that fall into the traditional chemical space. The chemical nature of the PPIs makes attractive the use of peptides or peptidomimetics to selectively modulate such interactions. Despite the fact peptide-based drug discovery has been challenging, the use of peptides as leads compounds for drug discovery is still a valid strategy. This chapter discusses the current status of PPIs in epigenetic drug discovery. A special emphasis is made on peptides and peptide-like compounds as potential drug candidates.
Collapse
|
127
|
Berlin R, Gruen R, Best J. Systems Medicine-Complexity Within, Simplicity Without. JOURNAL OF HEALTHCARE INFORMATICS RESEARCH 2017; 1:119-137. [PMID: 28713872 PMCID: PMC5491616 DOI: 10.1007/s41666-017-0002-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 04/12/2017] [Accepted: 04/25/2017] [Indexed: 12/14/2022]
Abstract
This paper presents a brief history of Systems Theory, progresses to Systems Biology, and its relation to the more traditional investigative method of reductionism. The emergence of Systems Medicine represents the application of Systems Biology to disease and clinical issues. The challenges faced by this transition from Systems Biology to Systems Medicine are explained; the requirements of physicians at the bedside, caring for patients, as well as the place of human-human interaction and the needs of the patients are addressed. An organ-focused transition to Systems Medicine, rather than a genomic-, molecular-, or cell-based effort is emphasized. Organ focus represents a middle-out approach to ease this transition and to maximize the benefits of scientific discovery and clinical application. This method manages the perceptions of time and space, the massive amounts of human- and patient-related data, and the ensuing complexity of information.
Collapse
Affiliation(s)
- Richard Berlin
- Department of Computer Science, University of Illinois, Urbana, IL USA
| | - Russell Gruen
- Nanyang Institute of Technology in Health and Medicine, Department of Surgery, Lee Kong Chian School of Medicine, Singapore, Singapore
| | - James Best
- Lee Kong Chian School of Medicine, Singapore, Singapore
| |
Collapse
|
128
|
Kathera C, Zhang J, Janardhan A, Sun H, Ali W, Zhou X, He L, Guo Z. Interacting partners of FEN1 and its role in the development of anticancer therapeutics. Oncotarget 2017; 8:27593-27602. [PMID: 28187440 PMCID: PMC5432360 DOI: 10.18632/oncotarget.15176] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 01/24/2017] [Indexed: 11/25/2022] Open
Abstract
Protein-protein interaction (PPI) plays a key role in cellular communication, Protein-protein interaction connected with each other with hubs and nods involved in signaling pathways. These interactions used to develop network based biomarkers for early diagnosis of cancer. FEN1(Flap endonuclease 1) is a central component in cellular metabolism, over expression and decrease of FEN1 levels may cause cancer, these regulation changes of Flap endonuclease 1reported in many cancer cells, to consider this data may needs to develop a network based biomarker. The current review focused on types of PPI, based on nature, detection methods and its role in cancer. Interacting partners of Flap endonuclease 1 role in DNA replication repair and development of anticancer therapeutics based on Protein-protein interaction data.
Collapse
Affiliation(s)
- Chandrasekhar Kathera
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jing Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Avilala Janardhan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Hongfang Sun
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Wajid Ali
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xiaolong Zhou
- The Laboratory of Animal Genetics, Breeding, and Reproduction, College of Animal Science and Technology, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Lingfeng He
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
129
|
Zarei O, Hamzeh-Mivehroud M, Benvenuti S, Ustun-Alkan F, Dastmalchi S. Characterizing the Hot Spots Involved in RON-MSPβ Complex Formation Using In Silico Alanine Scanning Mutagenesis and Molecular Dynamics Simulation. Adv Pharm Bull 2017; 7:141-150. [PMID: 28507948 PMCID: PMC5426727 DOI: 10.15171/apb.2017.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/18/2017] [Accepted: 03/20/2017] [Indexed: 12/30/2022] Open
Abstract
Purpose: Implication of protein-protein interactions (PPIs) in development of many diseases such as cancer makes them attractive for therapeutic intervention and rational drug design. RON (Recepteur d'Origine Nantais) tyrosine kinase receptor has gained considerable attention as promising target in cancer therapy. The activation of RON via its ligand, macrophage stimulation protein (MSP) is the most common mechanism of activation for this receptor. The aim of the current study was to perform in silico alanine scanning mutagenesis and to calculate binding energy for prediction of hot spots in protein-protein interface between RON and MSPβ chain (MSPβ). Methods: In this work the residues at the interface of RON-MSPβ complex were mutated to alanine and then molecular dynamics simulation was used to calculate binding free energy. Results: The results revealed that Gln193, Arg220, Glu287, Pro288, Glu289, and His424 residues from RON and Arg521, His528, Ser565, Glu658, and Arg683 from MSPβ may play important roles in protein-protein interaction between RON and MSP. Conclusion: Identification of these RON hot spots is important in designing anti-RON drugs when the aim is to disrupt RON-MSP interaction. In the same way, the acquired information regarding the critical amino acids of MSPβ can be used in the process of rational drug design for developing MSP antagonizing agents, the development of novel MSP mimicking peptides where inhibition of RON activation is required, and the design of experimental site directed mutagenesis studies.
Collapse
Affiliation(s)
- Omid Zarei
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Students Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Hamzeh-Mivehroud
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Silvia Benvenuti
- Molecular Therapeutics and Exploratory Research Laboratory, Candiolo Cancer Institute-FPO-IRCCS, Candiolo, Turin, Italy
| | - Fulya Ustun-Alkan
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Istanbul University, Istanbul, Turkey
| | - Siavoush Dastmalchi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
130
|
ProBiS tools (algorithm, database, and web servers) for predicting and modeling of biologically interesting proteins. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017; 128:24-32. [PMID: 28212856 DOI: 10.1016/j.pbiomolbio.2017.02.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 12/14/2016] [Accepted: 02/07/2017] [Indexed: 01/30/2023]
Abstract
ProBiS (Protein Binding Sites) Tools consist of algorithm, database, and web servers for prediction of binding sites and protein ligands based on the detection of structurally similar binding sites in the Protein Data Bank. In this article, we review the operations that ProBiS Tools perform, provide comments on the evolution of the tools, and give some implementation details. We review some of its applications to biologically interesting proteins. ProBiS Tools are freely available at http://probis.cmm.ki.si and http://probis.nih.gov.
Collapse
|
131
|
Vaseghi Maghvan P, Rezaei-Tavirani M, Zali H, Nikzamir A, Abdi S, Khodadoostan M, Asadzadeh-Aghdaei H. Network analysis of common genes related to esophageal, gastric, and colon cancers. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2017; 10:295-302. [PMID: 29379595 PMCID: PMC5758738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
AIM The aim of this study was to provide a biomarker panel for esophageal, gastric and colorectal cancers. It can help introducing some diagnostic biomarkers for these diseases. BACKGROUND Gastrointestinal cancers (GICs) including esophageal, gastric and colorectal cancers are the most common cancers in the world which are usually diagnosed in the final stages and due to heterogeneity of these diseases, the treatments usually are not successful. For this reason, many studies have been conducted to discover predictive biomarkers. METHODS In the present study, 507 genes related to esophageal, gastric and colon cancers were extracted.. The network was constructed by Cytoscape software (version 3.4.0). Then a main component of the network was analyzed considering centrality parameters including degree, betweenness, closeness and stress. Three clusters of the protein network accompanied with their seed nodes were determined by MCODE application in Cytoscape software. Furthermore, Gene Ontology (GO) analysis of the key genes in combination to the seed nodes was performed. RESULTS The network of 17 common differential expressed genes in three esophageal, gastric and colon adenocarcinomas including 1730 nodes and 9188 edges were constructed. Eight crucial genes were determined. Three Clusters of the network were analyzed by GO analysis. CONCLUSION The analyses of common genes of the three cancers showed that there are some common crucial genes including TP53, EGFR, MYC, AKT1, CDKN2A, CCND1 and HSP90AA1 which are tightly related to gastrointestinal cancers and can be predictive biomarkers for these cancers.
Collapse
Affiliation(s)
- Padina Vaseghi Maghvan
- Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hakimeh Zali
- Proteomics Research Center, Department of Tissue engineering and Applied Cell, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abdolrahim Nikzamir
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Abdi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Khodadoostan
- Department of Gastroenterology and Hepatology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Asadzadeh-Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
132
|
Chang NW, Dai HJ, Shih YY, Wu CY, Dela Rosa MAC, Obena RP, Chen YJ, Hsu WL, Oyang YJ. Biomarker identification of hepatocellular carcinoma using a methodical literature mining strategy. Database (Oxford) 2017; 2017:bax082. [PMID: 31725857 PMCID: PMC7243925 DOI: 10.1093/database/bax082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 10/11/2017] [Accepted: 10/11/2017] [Indexed: 12/31/2022]
Abstract
Hepatocellular carcinoma (HCC), one of the most common causes of cancer-related deaths, carries a 5-year survival rate of 18%, underscoring the need for robust biomarkers. In spite of the increased availability of HCC related literatures, many of the promising biomarkers reported have not been validated for clinical use. To narrow down the wide range of possible biomarkers for further clinical validation, bioinformaticians need to sort them out using information provided in published works. Biomedical text mining is an automated way to obtain information of interest within the massive collection of biomedical knowledge, thus enabling extraction of data for biomarkers associated with certain diseases. This method can significantly reduce both the time and effort spent on studying important maladies such as liver diseases. Herein, we report a text mining-aided curation pipeline to identify potential biomarkers for liver cancer. The curation pipeline integrates PubMed E-Utilities to collect abstracts from PubMed and recognize several types of named entities by machine learning-based and pattern-based methods. Genes/proteins from evidential sentences were classified as candidate biomarkers using a convolutional neural network. Lastly, extracted biomarkers were ranked depending on several criteria, such as the frequency of keywords and articles and the journal impact factor, and then integrated into a meaningful list for bioinformaticians. Based on the developed pipeline, we constructed MarkerHub, which contains 2128 candidate biomarkers extracted from PubMed publications from 2008 to 2017. Database URL: http://markerhub.iis.sinica.edu.tw.
Collapse
Affiliation(s)
- Nai-Wen Chang
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
- Institute of Information Science, Academia Sinica, Taipei, Taiwan
| | - Hong-Jie Dai
- Department of Computer Science and Information Engineering, National Taitung University, Taitung, Taiwan
- Interdisciplinary Program of Green and Information Technology, National Taitung University, Taitung, Taiwan
| | - Yung-Yu Shih
- Institute of Information Science, Academia Sinica, Taipei, Taiwan
| | - Chi-Yang Wu
- Institute of Information Science, Academia Sinica, Taipei, Taiwan
| | | | | | - Yu-Ju Chen
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Wen-Lian Hsu
- Institute of Information Science, Academia Sinica, Taipei, Taiwan
| | - Yen-Jen Oyang
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
133
|
Abstract
A tremendous asset to the analysis of protein-protein interactions is the yeast-2-hybrid (Y2H) method. The Y2H assay is a heterologous system that is expanding network biology knowledge via in vivo investigations of binary protein-protein interactions. Traditionally, the Y2H protocol entails the mating or co-transformation of yeast in solid agar media followed by visual analysis for diploid selection. Having played a key role in identifying protein-protein interactions for nearly three decades in a wide range of biological systems, the Y2H system assays the interaction between two proteins of interest which results in a reconstituted and/or activation of transcription factor allowing a reporter gene to be transcribed. Overall, the Y2H method takes advantage of two factors: (1) the auxotrophic yeast requires expression of the reporter gene to grow in media purposefully designed to lack one or more essential amino acids, and (2) the DNA-binding (DB) domain of transcription factor GAL4 is unable to initiate transcription unless it is physically associated with an activating domain (AD), which, together, DBs and ADs are fused to proteins of interest that must interact with each other to reconstitute the transcription factor and activate the reporter gene. The applications of Y2H are broad, entailing fields such as drug discovery, clinical trials for human disease including cancer and neurodegenerative disease, and extend even into synthetic biology applications and cellular engineering. This chapter begins with an introduction to the fundamental mechanics of Y2H utilizing a genetically engineered strain of yeast and proceeds with an in-depth look at the different types of Y2H and turn our focus particularly to the GAL4-based Y2H system to map protein-protein interactions. We will then provide a step-by-step protocol for the Y2H experimentation preceded by a materials listing while simultaneously including key notes throughout the entire experimental process of biological-mechanistic and historical understandings of the steps.
Collapse
|
134
|
Skrlj B, Kunej T. Computational identification of non-synonymous polymorphisms within regions corresponding to protein interaction sites. Comput Biol Med 2016; 79:30-35. [PMID: 27744178 DOI: 10.1016/j.compbiomed.2016.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 10/02/2016] [Accepted: 10/03/2016] [Indexed: 11/24/2022]
Abstract
BACKGROUND Protein-protein interactions (PPI) play an important role in function of all organisms and enable understanding of underlying metabolic processes. Computational predictions of PPIs are an important aspect in proteomics, as experimental methods may result in high degree of false positive results and are more expensive. Although there are many databases collecting predicted PPIs, exploration of genetics information underlying PPI interactions has not been investigated thoroughly. The aim of the present study was to identify genomic locations corresponding to regions involved in predicted PPIs and to collect non-synonymous polymorphisms (nsSNPs) located within those regions; which we termed PPI-SNPs. METHODS Predicted PPIs were obtained from PiSITE database (http://pisite.hgc.jp). Non-synonymous SNPs mapped on protein structural data (PDBs) were obtained from the UCSC server. Polymorphism locations on protein structures were mapped to predicted PPI regions. DAVID tool was used for pathway enrichment and gene cluster analysis (https://david.ncifcrf.gov/). RESULTS We collected 544 polymorphisms located within predicted PPI sites that map to 197 genes. We identified 9 SNPs, previously associated with diseases, but not yet associated with PPI sites. We also found examples in which polymorphisms located within predicted PPI regions are also occurring within previously experimentally validated PPIs and within experimentally determined functional domains. CONCLUSIONS Our study provides the first catalog of nsSNPs located within predicted PPIs. These prioritized SNPs present the basis for planning experimental validation of SNPs that cause gain or loss of PPIs. Our implementation is expandable, as datasets used are constantly updated.
Collapse
Affiliation(s)
- Blaz Skrlj
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Groblje 3, Slovenia.
| | - Tanja Kunej
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Groblje 3, Slovenia.
| |
Collapse
|
135
|
Multi-label ℓ 2-regularized logistic regression for predicting activation/inhibition relationships in human protein-protein interaction networks. Sci Rep 2016; 6:36453. [PMID: 27819359 PMCID: PMC5098220 DOI: 10.1038/srep36453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 10/17/2016] [Indexed: 11/30/2022] Open
Abstract
Protein-protein interaction (PPI) networks are naturally viewed as infrastructure to infer signalling pathways. The descriptors of signal events between two interacting proteins such as upstream/downstream signal flow, activation/inhibition relationship and protein modification are indispensable for inferring signalling pathways from PPI networks. However, such descriptors are not available in most cases as most PPI networks are seldom semantically annotated. In this work, we extend ℓ2-regularized logistic regression to the scenario of multi-label learning for predicting the activation/inhibition relationships in human PPI networks. The phenomenon that both activation and inhibition relationships exist between two interacting proteins is computationally modelled by multi-label learning framework. The problem of GO (gene ontology) sparsity is tackled by introducing the homolog knowledge as independent homolog instances. ℓ2-regularized logistic regression is accordingly adopted here to penalize the homolog noise and to reduce the computational complexity of the double-sized training data. Computational results show that the proposed method achieves satisfactory multi-label learning performance and outperforms the existing phenotype correlation method on the experimental data of Drosophila melanogaster. Several predictions have been validated against recent literature. The predicted activation/inhibition relationships in human PPI networks are provided in the supplementary file for further biomedical research.
Collapse
|
136
|
Folador EL, de Carvalho PVSD, Silva WM, Ferreira RS, Silva A, Gromiha M, Ghosh P, Barh D, Azevedo V, Röttger R. In silico identification of essential proteins in Corynebacterium pseudotuberculosis based on protein-protein interaction networks. BMC SYSTEMS BIOLOGY 2016; 10:103. [PMID: 27814699 PMCID: PMC5097352 DOI: 10.1186/s12918-016-0346-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 10/18/2016] [Indexed: 12/27/2022]
Abstract
Background Corynebacterium pseudotuberculosis (Cp) is a gram-positive bacterium that is classified into equi and ovis serovars. The serovar ovis is the etiological agent of caseous lymphadenitis, a chronic infection affecting sheep and goats, causing economic losses due to carcass condemnation and decreased production of meat, wool, and milk. Current diagnosis or treatment protocols are not fully effective and, thus, require further research of Cp pathogenesis. Results Here, we mapped known protein-protein interactions (PPI) from various species to nine Cp strains to reconstruct parts of the potential Cp interactome and to identify potentially essential proteins serving as putative drug targets. On average, we predict 16,669 interactions for each of the nine strains (with 15,495 interactions shared among all strains). An in silico sanity check suggests that the potential networks were not formed by spurious interactions but have a strong biological bias. With the inferred Cp networks we identify 181 essential proteins, among which 41 are non-host homologous. Conclusions The list of candidate interactions of the Cp strains lay the basis for developing novel hypotheses and designing according wet-lab studies. The non-host homologous essential proteins are attractive targets for therapeutic and diagnostic proposes. They allow for searching of small molecule inhibitors of binding interactions enabling modern drug discovery. Overall, the predicted Cp PPI networks form a valuable and versatile tool for researchers interested in Corynebacterium pseudotuberculosis. Electronic supplementary material The online version of this article (doi:10.1186/s12918-016-0346-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Edson Luiz Folador
- Department of General Biology, Instituto de Ciências Biológicas (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil.,Institute of Biological Sciences, Federal University of Para, Belém, PA, Brazil.,Biotechnology Center (CBiotec), Federal University of Paraiba (UFPB), João Pessoa, Brazil
| | - Paulo Vinícius Sanches Daltro de Carvalho
- Department of General Biology, Instituto de Ciências Biológicas (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil.,Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
| | - Wanderson Marques Silva
- Department of General Biology, Instituto de Ciências Biológicas (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Rafaela Salgado Ferreira
- Department of Biochemistry and Immunology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Artur Silva
- Institute of Biological Sciences, Federal University of Para, Belém, PA, Brazil
| | - Michael Gromiha
- Department of Biotechnology, Indian Institute of Technology (IIT) Madras, Tamilnadu, India
| | - Preetam Ghosh
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA, USA
| | - Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur, West Bengal, India
| | - Vasco Azevedo
- Department of General Biology, Instituto de Ciências Biológicas (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Richard Röttger
- Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
137
|
Hassan A, Naz A, Obaid A, Paracha RZ, Naz K, Awan FM, Muhmmad SA, Janjua HA, Ahmad J, Ali A. Pangenome and immuno-proteomics analysis of Acinetobacter baumannii strains revealed the core peptide vaccine targets. BMC Genomics 2016; 17:732. [PMID: 27634541 PMCID: PMC5025611 DOI: 10.1186/s12864-016-2951-4] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 07/19/2016] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Acinetobacter baumannii has emerged as a significant nosocomial pathogen during the last few years, exhibiting resistance to almost all major classes of antibiotics. Alternative treatment options such as vaccines tend to be most promising and cost effective approaches against this resistant pathogen. In the current study, we have explored the pan-genome of A. baumannii followed by immune-proteomics and reverse vaccinology approaches to identify potential core vaccine targets. RESULTS The pan-genome of all available A. baumannii strains (30 complete genomes) is estimated to contain 7,606 gene families and the core genome consists of 2,445 gene families (~32 % of the pan-genome). Phylogenetic tree, comparative genomic and proteomic analysis revealed both intra- and inter genomic similarities and evolutionary relationships. Among the conserved core genome, thirteen proteins, including P pilus assembly protein, pili assembly chaperone, AdeK, PonA, OmpA, general secretion pathway protein D, FhuE receptor, Type VI secretion system OmpA/MotB, TonB dependent siderophore receptor, general secretion pathway protein D, outer membrane protein, peptidoglycan associated lipoprotein and peptidyl-prolyl cis-trans isomerase are identified as highly antigenic. Epitope mapping of the target proteins revealed the presence of antigenic surface exposed 9-mer T-cell epitopes. Protein-protein interaction and functional annotation have shown their involvement in significant biological and molecular processes. The pipeline is validated by predicting already known immunogenic targets against Gram negative pathogen Helicobacter pylori as a positive control. CONCLUSION The study, based upon combinatorial approach of pan-genomics, core genomics, proteomics and reverse vaccinology led us to find out potential vaccine candidates against A. baumannii. The comprehensive analysis of all the completely sequenced genomes revealed thirteen putative antigens which could elicit substantial immune response. The integration of computational vaccinology strategies would facilitate in tackling the rapid dissemination of resistant A.baumannii strains. The scarcity of effective antibiotics and the global expansion of sequencing data making this approach desirable in the development of effective vaccines against A. baumannii and other bacterial pathogens.
Collapse
Affiliation(s)
- Afreenish Hassan
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Anam Naz
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Ayesha Obaid
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Rehan Zafar Paracha
- Research Center for Modeling and Simulation (RCMS), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Kanwal Naz
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Faryal Mehwish Awan
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Syed Aun Muhmmad
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan, Pakistan
| | - Hussnain Ahmed Janjua
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Jamil Ahmad
- Research Center for Modeling and Simulation (RCMS), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
- Department of Computer Science and Information Technology, Stratford University, Falls Church, VA 22043 USA
| | - Amjad Ali
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| |
Collapse
|
138
|
Wang P, Chen Y, Lü J, Wang Q, Yu X. Graphical Features of Functional Genes in Human Protein Interaction Network. IEEE TRANSACTIONS ON BIOMEDICAL CIRCUITS AND SYSTEMS 2016; 10:707-20. [PMID: 26841412 DOI: 10.1109/tbcas.2015.2487299] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
With the completion of the human genome project, it is feasible to investigate large-scale human protein interaction network (HPIN) with complex networks theory. Proteins are encoded by genes. Essential, viable, disease, conserved, housekeeping (HK) and tissue-enriched (TE) genes are functional genes, which are organized and functioned via interaction networks. Based on up-to-date data from various databases or literature, two large-scale HPINs and six subnetworks are constructed. We illustrate that the HPINs and most of the subnetworks are sparse, small-world, scale-free, disassortative and with hierarchical modularity. Among the six subnetworks, essential, disease and HK subnetworks are more densely connected than the others. Statistical analysis on the topological structures of the HPIN reveals that the lethal, the conserved, the HK and the TE genes are with hallmark graphical features. Receiver operating characteristic (ROC) curves indicate that the essential genes can be distinguished from the viable ones with accuracy as high as almost 70%. Closeness, semi-local and eigenvector centralities can distinguish the HK genes from the TE ones with accuracy around 82%. Furthermore, the Venn diagram, cluster dendgrams and classifications of disease genes reveal that some classes of disease genes are with hallmark graphical features, especially for cancer genes, HK disease genes and TE disease genes. The findings facilitate the identification of some functional genes via topological structures. The investigations shed some light on the characteristics of the compete interactome, which have potential implications in networked medicine and biological network control.
Collapse
|
139
|
McHugh SM, Rogers JR, Yu H, Lin YS. Insights into How Cyclic Peptides Switch Conformations. J Chem Theory Comput 2016; 12:2480-8. [DOI: 10.1021/acs.jctc.6b00193] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Sean M. McHugh
- Department
of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| | - Julia R. Rogers
- Department
of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| | - Hongtao Yu
- Department
of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| | - Yu-Shan Lin
- Department
of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
140
|
Iqbal U, Hsu CK, Nguyen PAA, Clinciu DL, Lu R, Syed-Abdul S, Yang HC, Wang YC, Huang CY, Huang CW, Chang YC, Hsu MH, Jian WS, Li YCJ. Cancer-disease associations: A visualization and animation through medical big data. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2016; 127:44-51. [PMID: 27000288 DOI: 10.1016/j.cmpb.2016.01.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 01/06/2016] [Accepted: 01/11/2016] [Indexed: 06/05/2023]
Abstract
OBJECTIVE Cancer is the primary disease responsible for death and disability worldwide. Currently, prevention and early detection represents the best hope for cure. Knowing the expected diseases that occur with a particular cancer in advance could lead to physicians being able to better tailor their treatment for cancer. The aim of this study was to build an animated visualization tool called as Cancer Associations Map Animation (CAMA), to chart the association of cancers with other disease over time. METHODS The study population was collected from the Taiwan National Health Insurance Database during the period January 2000 to December 2002, 782 million outpatient visits were used to compute the associations of nine major cancers with other diseases. A motion chart was used to quantify and visualize the associations between diseases and cancers. RESULTS The CAMA motion chart that was built successfully facilitated the observation of cancer-disease associations across ages and genders. The CAMA system can be accessed online at http://203.71.86.98/web/runq16.html. CONCLUSION The CAMA animation system is an animated medical data visualization tool which provides a dynamic, time-lapse, animated view of cancer-disease associations across different age groups and gender. Derived from a large, nationwide healthcare dataset, this exploratory data analysis tool can detect cancer comorbidities earlier than is possible by manual inspection. Taking into account the trajectory of cancer-specific comorbidity development may facilitate clinicians and healthcare researchers to more efficiently explore early stage hypotheses, develop new cancer treatment approaches, and identify potential effect modifiers or new risk factors associated with specific cancers.
Collapse
Affiliation(s)
- Usman Iqbal
- Graduate of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taiwan; International Center for Health Information Technology (ICHIT), Taipei Medical University, Taiwan
| | - Chun-Kung Hsu
- Graduate of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taiwan; International Center for Health Information Technology (ICHIT), Taipei Medical University, Taiwan
| | - Phung Anh Alex Nguyen
- Graduate of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taiwan; International Center for Health Information Technology (ICHIT), Taipei Medical University, Taiwan
| | - Daniel Livius Clinciu
- Graduate of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taiwan; International Center for Health Information Technology (ICHIT), Taipei Medical University, Taiwan
| | - Richard Lu
- Graduate of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taiwan; International Center for Health Information Technology (ICHIT), Taipei Medical University, Taiwan
| | - Shabbir Syed-Abdul
- Graduate of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taiwan; International Center for Health Information Technology (ICHIT), Taipei Medical University, Taiwan
| | - Hsuan-Chia Yang
- Graduate of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taiwan; International Center for Health Information Technology (ICHIT), Taipei Medical University, Taiwan; Institute of Biomedical Informatics, National Yang Ming University, Taiwan
| | - Yao-Chin Wang
- Graduate of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taiwan
| | - Chu-Ya Huang
- Graduate of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taiwan
| | - Chih-Wei Huang
- Graduate of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taiwan; International Center for Health Information Technology (ICHIT), Taipei Medical University, Taiwan
| | - Yo-Cheng Chang
- Graduate of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taiwan
| | - Min-Huei Hsu
- Graduate of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taiwan; International Center for Health Information Technology (ICHIT), Taipei Medical University, Taiwan; Bureau of International Cooperation, Ministry of Health and Welfare, Taipei, Taiwan
| | - Wen-Shan Jian
- Graduate of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taiwan; International Center for Health Information Technology (ICHIT), Taipei Medical University, Taiwan; School of Health Care Administration, Taipei Medical University, Taipei, Taiwan; Faculty of Health Sciences, Macau University of Science and Technology, Macau, China
| | - Yu-Chuan Jack Li
- Graduate of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taiwan; International Center for Health Information Technology (ICHIT), Taipei Medical University, Taiwan; Department of Dermatology, Taipei Medical University - Wan Fang Hospital, Taipei, Taiwan.
| |
Collapse
|
141
|
Mirzaei N, Poursina F, Moghim S, Ghaempanah AM, Safaei HG. The Bioinformatics Report of Mutation Outcome on NADPH Flavin Oxidoreductase Protein Sequence in Clinical Isolates of H. pylori. Curr Microbiol 2016; 72:596-605. [PMID: 26821239 DOI: 10.1007/s00284-016-0992-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 12/14/2015] [Indexed: 12/23/2022]
Abstract
frxA gene has been implicated in the metronidazole nitro reduction by H. pylori. Alternatively, frxA is expected to contribute to the protection of urease and to the in vivo survival of H. pylori. The aim of present study is to report the mutation effects on the frxA protein sequence in clinical isolates of H. pylori in our community. Metronidazole resistance was proven in 27 of 48 isolates. glmM and frxA genes were used for molecular confirmation of H. pylori isolates. The primer set for detection of whole sequence of frxA gene for the effect of mutation on protein sequence was used. DNA and protein sequence evaluation and analysis were done by blast, Clustal Omega, and T COFFEE programs. Then, FrxA protein sequences from six metronidazole-resistant clinical isolates were analyzed by web-based bioinformatics tools. The result of six metronidazole-resistant clinical isolates in comparison with strain 26695 showed ten missense mutations. The result with the STRING program revealed that no change was seen after alterations in these sequences. According to consensus data involving four methods, residue substitutions at 40, 13, and 141 increase the stability of protein sequence after mutation, while other alterations decrease. Residue substitutions at 40, 43, 141, 138, 169, and 179 are deleterious, while, V7I, Q10R, V34I, and V96I alterations are neutral. As FrxA contribute to survival of bacterium and in regard to the effect of mutations on protein function, it might affect the survival and bacterium phenotype and it need to be studied more. Also, none of the stability prediction tool is perfect; iStable is the best predictor method among all methods.
Collapse
Affiliation(s)
- Nasrin Mirzaei
- Department of Microbiology, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Farkhondeh Poursina
- Department of Microbiology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sharareh Moghim
- Department of Microbiology, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | | |
Collapse
|
142
|
He H, Cao S, Niu T, Zhou Y, Zhang L, Zeng Y, Zhu W, Wang YP, Deng HW. Network-Based Meta-Analyses of Associations of Multiple Gene Expression Profiles with Bone Mineral Density Variations in Women. PLoS One 2016; 11:e0147475. [PMID: 26808152 PMCID: PMC4726665 DOI: 10.1371/journal.pone.0147475] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 01/05/2016] [Indexed: 01/22/2023] Open
Abstract
Background Existing microarray studies of bone mineral density (BMD) have been critical for understanding the pathophysiology of osteoporosis, and have identified a number of candidate genes. However, these studies were limited by their relatively small sample sizes and were usually analyzed individually. Here, we propose a novel network-based meta-analysis approach that combines data across six microarray studies to identify functional modules from human protein-protein interaction (PPI) data, and highlight several differentially expressed genes (DEGs) and a functional module that may play an important role in BMD regulation in women. Methods Expression profiling studies were identified by searching PubMed, Gene Expression Omnibus (GEO) and ArrayExpress. Two meta-analysis methods were applied across different gene expression profiling studies. The first, a nonparametric Fisher’s method, combined p-values from individual experiments to identify genes with large effect sizes. The second method combined effect sizes from individual datasets into a meta-effect size to gain a higher precision of effect size estimation across all datasets. Genes with Q test’s p-values < 0.05 or I2 values > 50% were assessed by a random effects model and the remainder by a fixed effects model. Using Fisher’s combined p-values, functional modules were identified through an integrated analysis of microarray data in the context of large protein–protein interaction (PPI) networks. Two previously published meta-analysis studies of genome-wide association (GWA) datasets were used to determine whether these module genes were genetically associated with BMD. Pathway enrichment analysis was performed with a hypergeometric test. Results Six gene expression datasets were identified, which included a total of 249 (129 high BMD and 120 low BMD) female subjects. Using a network-based meta-analysis, a consensus module containing 58 genes (nodes) and 83 edges was detected. Pathway enrichment analysis of the 58 module genes revealed that these genes were enriched in several important KEGG pathways including Osteoclast differentiation, B cell receptor signaling pathway, MAPK signaling pathway, Chemokine signaling pathway and Insulin signaling pathway. The importance of module genes was replicated by demonstrating that most module genes were genetically associated with BMD in the GWAS data sets. Meta-analyses were performed at the individual gene level by combining p-values and effect sizes. Five candidate genes (ESR1, MAP3K3, PYGM, RAC1 and SYK) were identified based on gene expression meta-analysis, and their associations with BMD were also replicated by two BMD meta-analysis studies. Conclusions In summary, our network-based meta-analysis not only identified important differentially expressed genes but also discovered biologically meaningful functional modules for BMD determination. Our study may provide novel therapeutic targets for osteoporosis in women.
Collapse
Affiliation(s)
- Hao He
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Shaolong Cao
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana, United States of America
| | - Tianhua Niu
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Yu Zhou
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Lan Zhang
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Yong Zeng
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Wei Zhu
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Yu-ping Wang
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana, United States of America
| | - Hong-wen Deng
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
143
|
Hu G, Xiao F, Li Y, Li Y, Vongsangnak W. Protein-Protein Interface and Disease: Perspective from Biomolecular Networks. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2016; 160:57-74. [PMID: 27928579 DOI: 10.1007/10_2016_40] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Protein-protein interactions are involved in many important biological processes and molecular mechanisms of disease association. Structural studies of interfacial residues in protein complexes provide information on protein-protein interactions. Characterizing protein-protein interfaces, including binding sites and allosteric changes, thus pose an imminent challenge. With special focus on protein complexes, approaches based on network theory are proposed to meet this challenge. In this review we pay attention to protein-protein interfaces from the perspective of biomolecular networks and their roles in disease. We first describe the different roles of protein complexes in disease through several structural aspects of interfaces. We then discuss some recent advances in predicting hot spots and communication pathway analysis in terms of amino acid networks. Finally, we highlight possible future aspects of this area with respect to both methodology development and applications for disease treatment.
Collapse
Affiliation(s)
- Guang Hu
- Center for Systems Biology, School of Electronic and Information Engineering, Soochow University, Suzhou, 215006, China.
| | - Fei Xiao
- School of Basic Medicine and Biological Sciences, Medical College of Soochow University, Suzhou, 215123, China
| | - Yuqian Li
- School of Electronic Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Yuan Li
- Center for Systems Biology, School of Electronic and Information Engineering, Soochow University, Suzhou, 215006, China
| | - Wanwipa Vongsangnak
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand.
- Computational Biomodelling Laboratory for Agricultural Science and Technology (CBLAST), Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand.
| |
Collapse
|
144
|
Richardson TG, Shihab HA, Rivas MA, McCarthy MI, Campbell C, Timpson NJ, Gaunt TR. A Protein Domain and Family Based Approach to Rare Variant Association Analysis. PLoS One 2016; 11:e0153803. [PMID: 27128313 PMCID: PMC4851355 DOI: 10.1371/journal.pone.0153803] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 04/04/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND It has become common practice to analyse large scale sequencing data with statistical approaches based around the aggregation of rare variants within the same gene. We applied a novel approach to rare variant analysis by collapsing variants together using protein domain and family coordinates, regarded to be a more discrete definition of a biologically functional unit. METHODS Using Pfam definitions, we collapsed rare variants (Minor Allele Frequency ≤ 1%) together in three different ways 1) variants within single genomic regions which map to individual protein domains 2) variants within two individual protein domain regions which are predicted to be responsible for a protein-protein interaction 3) all variants within combined regions from multiple genes responsible for coding the same protein domain (i.e. protein families). A conventional collapsing analysis using gene coordinates was also undertaken for comparison. We used UK10K sequence data and investigated associations between regions of variants and lipid traits using the sequence kernel association test (SKAT). RESULTS We observed no strong evidence of association between regions of variants based on Pfam domain definitions and lipid traits. Quantile-Quantile plots illustrated that the overall distributions of p-values from the protein domain analyses were comparable to that of a conventional gene-based approach. Deviations from this distribution suggested that collapsing by either protein domain or gene definitions may be favourable depending on the trait analysed. CONCLUSION We have collapsed rare variants together using protein domain and family coordinates to present an alternative approach over collapsing across conventionally used gene-based regions. Although no strong evidence of association was detected in these analyses, future studies may still find value in adopting these approaches to detect previously unidentified association signals.
Collapse
Affiliation(s)
- Tom G. Richardson
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Hashem A. Shihab
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Manuel A. Rivas
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Mark I. McCarthy
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Colin Campbell
- Intelligent Systems Laboratory, University of Bristol, Bristol, United Kingdom
| | - Nicholas J. Timpson
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Tom R. Gaunt
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
- * E-mail:
| |
Collapse
|
145
|
Mei S, Zhu H. Multi-label multi-instance transfer learning for simultaneous reconstruction and cross-talk modeling of multiple human signaling pathways. BMC Bioinformatics 2015; 16:417. [PMID: 26718335 PMCID: PMC4697333 DOI: 10.1186/s12859-015-0841-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 07/13/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Signaling pathways play important roles in the life processes of cell growth, cell apoptosis and organism development. At present the signal transduction networks are far from complete. As an effective complement to experimental methods, computational modeling is suited to rapidly reconstruct the signaling pathways at low cost. To our knowledge, the existing computational methods seldom simultaneously exploit more than three signaling pathways into one predictive model for the discovery of novel signaling components and the cross-talk modeling between signaling pathways. RESULTS In this work, we propose a multi-label multi-instance transfer learning method to simultaneously reconstruct 27 human signaling pathways and model their cross-talks. Computational results show that the proposed method demonstrates satisfactory multi-label learning performance and rational proteome-wide predictions. Some predicted signaling components or pathway targeted proteins have been validated by recent literature. The predicted signaling components are further linked to pathways using the experimentally derived PPIs (protein-protein interactions) to reconstruct the human signaling pathways. Thus the map of the cross-talks via common signaling components and common signaling PPIs is conveniently inferred to provide valuable insights into the regulatory and cooperative relationships between signaling pathways. Lastly, gene ontology enrichment analysis is conducted to gain statistical knowledge about the reconstructed human signaling pathways. CONCLUSIONS Multi-label learning framework has been demonstrated effective in this work to model the phenomena that a signaling protein belongs to more than one signaling pathway. As results, novel signaling components and pathways targeted proteins are predicted to simultaneously reconstruct multiple human signaling pathways and the static map of their cross-talks for further biomedical research.
Collapse
Affiliation(s)
- Suyu Mei
- Software College, Shenyang Normal University, Shenyang, China. .,Bioinformatics Section, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Hao Zhu
- Bioinformatics Section, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
146
|
Mei S, Zhu H. A simple feature construction method for predicting upstream/downstream signal flow in human protein-protein interaction networks. Sci Rep 2015; 5:17983. [PMID: 26648121 PMCID: PMC4673612 DOI: 10.1038/srep17983] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 11/10/2015] [Indexed: 12/24/2022] Open
Abstract
Signaling pathways play important roles in understanding the underlying mechanism of cell growth, cell apoptosis, organismal development and pathways-aberrant diseases. Protein-protein interaction (PPI) networks are commonly-used infrastructure to infer signaling pathways. However, PPI networks generally carry no information of upstream/downstream relationship between interacting proteins, which retards our inferring the signal flow of signaling pathways. In this work, we propose a simple feature construction method to train a SVM (support vector machine) classifier to predict PPI upstream/downstream relations. The domain based asymmetric feature representation naturally embodies domain-domain upstream/downstream relations, providing an unconventional avenue to predict the directionality between two objects. Moreover, we propose a semantically interpretable decision function and a macro bag-level performance metric to satisfy the need of two-instance depiction of an interacting protein pair. Experimental results show that the proposed method achieves satisfactory cross validation performance and independent test performance. Lastly, we use the trained model to predict the PPIs in HPRD, Reactome and IntAct. Some predictions have been validated against recent literature.
Collapse
Affiliation(s)
- Suyu Mei
- Software College, Shenyang Normal University, Shenyang, China.,Bioinformatics Section, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hao Zhu
- Bioinformatics Section, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
147
|
Abstract
Enzymes are one of the most important groups of drug targets, and identifying possible ligand-enzyme interactions is of major importance in many drug discovery processes. Novel computational methods have been developed that can apply the information from the increasing number of resolved and available ligand-enzyme complexes to model new unknown interactions and therefore contribute to answer open questions in the field of drug discovery like the identification of unknown protein functions, off-target binding, ligand 3D homology modeling and induced-fit simulations.
Collapse
|
148
|
Regan K, Payne PRO. From Molecules to Patients: The Clinical Applications of Translational Bioinformatics. Yearb Med Inform 2015; 10:164-9. [PMID: 26293863 PMCID: PMC4587059 DOI: 10.15265/iy-2015-005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE In order to realize the promise of personalized medicine, Translational Bioinformatics (TBI) research will need to continue to address implementation issues across the clinical spectrum. In this review, we aim to evaluate the expanding field of TBI towards clinical applications, and define common themes and current gaps in order to motivate future research. METHODS Here we present the state-of-the-art of clinical implementation of TBI-based tools and resources. Our thematic analyses of a targeted literature search of recent TBI-related articles ranged across topics in genomics, data management, hypothesis generation, molecular epidemiology, diagnostics, therapeutics and personalized medicine. RESULTS Open areas of clinically-relevant TBI research identified in this review include developing data standards and best practices, publicly available resources, integrative systemslevel approaches, user-friendly tools for clinical support, cloud computing solutions, emerging technologies and means to address pressing legal, ethical and social issues. CONCLUSIONS There is a need for further research bridging the gap from foundational TBI-based theories and methodologies to clinical implementation. We have organized the topic themes presented in this review into four conceptual foci - domain analyses, knowledge engineering, computational architectures and computation methods alongside three stages of knowledge development in order to orient future TBI efforts to accelerate the goals of personalized medicine.
Collapse
Affiliation(s)
| | - P R O Payne
- Philip R.O. Payne, PhD, FACMI, The Ohio State University, Department of Biomedical Informatics, 250 Lincoln Tower, 1800 Cannon Drive, Columbus, OH 43210, USA, Tel: +1 614 292 4778, E-mail:
| |
Collapse
|
149
|
Ferreira LG, Dos Santos RN, Oliva G, Andricopulo AD. Molecular docking and structure-based drug design strategies. Molecules 2015; 20:13384-421. [PMID: 26205061 PMCID: PMC6332083 DOI: 10.3390/molecules200713384] [Citation(s) in RCA: 1043] [Impact Index Per Article: 104.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 07/14/2015] [Accepted: 07/20/2015] [Indexed: 02/07/2023] Open
Abstract
Pharmaceutical research has successfully incorporated a wealth of molecular modeling methods, within a variety of drug discovery programs, to study complex biological and chemical systems. The integration of computational and experimental strategies has been of great value in the identification and development of novel promising compounds. Broadly used in modern drug design, molecular docking methods explore the ligand conformations adopted within the binding sites of macromolecular targets. This approach also estimates the ligand-receptor binding free energy by evaluating critical phenomena involved in the intermolecular recognition process. Today, as a variety of docking algorithms are available, an understanding of the advantages and limitations of each method is of fundamental importance in the development of effective strategies and the generation of relevant results. The purpose of this review is to examine current molecular docking strategies used in drug discovery and medicinal chemistry, exploring the advances in the field and the role played by the integration of structure- and ligand-based methods.
Collapse
Affiliation(s)
- Leonardo G Ferreira
- Laboratório de Química Medicinal e Computacional, Centro de Pesquisa e Inovação em Biodiversidade e Fármacos, Instituto de Física de São Carlos, Universidade de São Paulo, Av. João Dagnone 1100, São Carlos-SP 13563-120, Brazil.
| | - Ricardo N Dos Santos
- Laboratório de Química Medicinal e Computacional, Centro de Pesquisa e Inovação em Biodiversidade e Fármacos, Instituto de Física de São Carlos, Universidade de São Paulo, Av. João Dagnone 1100, São Carlos-SP 13563-120, Brazil.
| | - Glaucius Oliva
- Laboratório de Química Medicinal e Computacional, Centro de Pesquisa e Inovação em Biodiversidade e Fármacos, Instituto de Física de São Carlos, Universidade de São Paulo, Av. João Dagnone 1100, São Carlos-SP 13563-120, Brazil.
| | - Adriano D Andricopulo
- Laboratório de Química Medicinal e Computacional, Centro de Pesquisa e Inovação em Biodiversidade e Fármacos, Instituto de Física de São Carlos, Universidade de São Paulo, Av. João Dagnone 1100, São Carlos-SP 13563-120, Brazil.
| |
Collapse
|
150
|
Gowthaman R, Lyskov S, Karanicolas J. DARC 2.0: Improved Docking and Virtual Screening at Protein Interaction Sites. PLoS One 2015; 10:e0131612. [PMID: 26181386 PMCID: PMC4504481 DOI: 10.1371/journal.pone.0131612] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 06/02/2015] [Indexed: 01/26/2023] Open
Abstract
Over the past decade, protein-protein interactions have emerged as attractive but challenging targets for therapeutic intervention using small molecules. Due to the relatively flat surfaces that typify protein interaction sites, modern virtual screening tools developed for optimal performance against “traditional” protein targets perform less well when applied instead at protein interaction sites. Previously, we described a docking method specifically catered to the shallow binding modes characteristic of small-molecule inhibitors of protein interaction sites. This method, called DARC (Docking Approach using Ray Casting), operates by comparing the topography of the protein surface when “viewed” from a vantage point inside the protein against the topography of a bound ligand when “viewed” from the same vantage point. Here, we present five key enhancements to DARC. First, we use multiple vantage points to more accurately determine protein-ligand surface complementarity. Second, we describe a new scheme for rapidly determining optimal weights in the DARC scoring function. Third, we incorporate sampling of ligand conformers “on-the-fly” during docking. Fourth, we move beyond simple shape complementarity and introduce a term in the scoring function to capture electrostatic complementarity. Finally, we adjust the control flow in our GPU implementation of DARC to achieve greater speedup of these calculations. At each step of this study, we evaluate the performance of DARC in a “pose recapitulation” experiment: predicting the binding mode of 25 inhibitors each solved in complex with its distinct target protein (a protein interaction site). Whereas the previous version of DARC docked only one of these inhibitors to within 2 Å RMSD of its position in the crystal structure, the newer version achieves this level of accuracy for 12 of the 25 complexes, corresponding to a statistically significant performance improvement (p < 0.001). Collectively then, we find that the five enhancements described here – which together make up DARC 2.0 – lead to dramatically improved speed and performance relative to the original DARC method.
Collapse
Affiliation(s)
- Ragul Gowthaman
- Center for Computational Biology, University of Kansas, 2030 Becker Dr., Lawrence, KS, 66045, United States of America
| | - Sergey Lyskov
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 North Charles St., Baltimore, MD, 21218, United States of America
| | - John Karanicolas
- Center for Computational Biology, University of Kansas, 2030 Becker Dr., Lawrence, KS, 66045, United States of America
- Department of Molecular Biosciences, University of Kansas, 2030 Becker Dr., Lawrence, KS, 66045, United States of America
- * E-mail:
| |
Collapse
|