51
|
Dilly S, Garnier M, Solé M, Bailly R, Taib N, Bestel I. In Silico Identification of a Key Residue for Substrate Recognition of the Riboflavin Membrane Transporter RFVT3. J Chem Inf Model 2020; 60:1368-1375. [DOI: 10.1021/acs.jcim.9b01020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Sébastien Dilly
- L’Institut de Chimie et Biologie des Membranes et des Nano-Objets (CBMN), Unité Mixte de Recherche (UMR) 5248, Centre National de la Recherche (CNRS), University of Bordeaux, Pessac 33600, France
| | - Mélanie Garnier
- L’Institut de Chimie et Biologie des Membranes et des Nano-Objets (CBMN), Unité Mixte de Recherche (UMR) 5248, Centre National de la Recherche (CNRS), University of Bordeaux, Pessac 33600, France
| | - Marion Solé
- L’Institut de Chimie et Biologie des Membranes et des Nano-Objets (CBMN), Unité Mixte de Recherche (UMR) 5248, Centre National de la Recherche (CNRS), University of Bordeaux, Pessac 33600, France
| | - Rémi Bailly
- L’Institut de Chimie et Biologie des Membranes et des Nano-Objets (CBMN), Unité Mixte de Recherche (UMR) 5248, Centre National de la Recherche (CNRS), University of Bordeaux, Pessac 33600, France
| | - Nada Taib
- L’Institut de Chimie et Biologie des Membranes et des Nano-Objets (CBMN), Unité Mixte de Recherche (UMR) 5248, Centre National de la Recherche (CNRS), University of Bordeaux, Pessac 33600, France
| | - Isabelle Bestel
- L’Institut de Chimie et Biologie des Membranes et des Nano-Objets (CBMN), Unité Mixte de Recherche (UMR) 5248, Centre National de la Recherche (CNRS), University of Bordeaux, Pessac 33600, France
| |
Collapse
|
52
|
In silico prediction of a new lead compound targeting enolase of trypanosomatids through structure-based virtual screening and molecular dynamic studies. J Mol Model 2020; 26:23. [PMID: 31912304 DOI: 10.1007/s00894-019-4284-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 12/20/2019] [Indexed: 10/25/2022]
Abstract
Enolase is one of the key glycolytic metalloenzyme in many organisms, and it is a potential therapeutic target including trypanosomatids. Sequence and structural analysis of enolase of Trypanosoma bruzi (TbENO), Trypanosoma cruzi (TcENO) and Leishmania donovani (LdENO) revealed conserved sequence pattern and structural features. Hence identification of an inhibitor against enolase of one trypanosomatid organism may have similar effects on enolase of homologous organisms belonging to same family. In the process to identify potent inhibitor compounds against TbENO by in silico methods, compounds containing the substructures of substrate, i.e. phosphoenolpyruvate (PEP) and the well-known inhibitors, fluoro-2-phosphono-acetohydroxamate (FPAH) and phosphono-acetohydroxamate (PAH), were collected. Virtual screening and induced fit docking studies were carried out to explore compounds that have better binding affinity than PEP and FPAH. PPPi was found to be the top hit exhibiting significant binding affinity towards enolase. Glide energy values of two other compounds represented by PubChem ID: 511392 and 101803456 was in good agreement with PEP and PAH. TbENO-PPPi complex was subjected to molecular orbital analysis and molecular dynamic studies by considering its remarkable binding affinity as it could be a potent inhibitor of enolase. Despite being an endogenous compound, based on the results of this study, we highlight PPPi to be a lead compound, and its structure can be treated as a model for further chemical modifications to obtain more potent antagonists.
Collapse
|
53
|
Bokhari H, Maryam A, Shahid R, Siddiqi AR. Oligosaccharyltransferase PglB of Campylobacter jejuni is a glycoprotein. World J Microbiol Biotechnol 2019; 36:9. [PMID: 31858269 DOI: 10.1007/s11274-019-2784-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 12/10/2019] [Indexed: 11/29/2022]
Abstract
Campylobacter jejuni is the one of the leading cause of bacterial food borne gastroenteritis. PglB, a glycosyltransferase, plays a crucial role of mediating glycosylation of numerous periplasmic proteins. It catalyzes N-glycosylation at the sequon D/E-X1-N-X2-S/T in its substrate proteins. Here we report that the PglB itself is a glycoprotein which self-glycosylates at N534 site in its DYNQS sequon by its own catalytic WWDYG motif. Site-directed mutagenesis, lectin Immunoblot, and mobility shift assays confirmed that the DYNQS is an N-glycosylation motif. PglB's N-glycosylation motif is structurally and functionally similar to its widely studied glycosylation substrate, the OMPH1. Its DYNQS motif forms a solvent-exposed crest. This motif is close to a cluster of polar and hydrophilic residues, which form a loop flanked by two α helices. This arrangement extremely apposite for auto-glycosylation at N534. This self-glycosylation ability of PglB could mediate C. jejuni's ability to colonize the intestinal epithelium. Further this capability may also bear significance for the development of novel conjugated vaccines and diagnostic tests.
Collapse
Affiliation(s)
- Habib Bokhari
- Department of Biosciences, COMSATS University, Islamabad, Chak Shazad Campus, Islamabad, Pakistan.
| | - Arooma Maryam
- Department of Biosciences, COMSATS University, Islamabad, Chak Shazad Campus, Islamabad, Pakistan
| | - Ramla Shahid
- Department of Biosciences, COMSATS University, Islamabad, Chak Shazad Campus, Islamabad, Pakistan
| | - Abdul Rauf Siddiqi
- Department of Biosciences, COMSATS University, Islamabad, Chak Shazad Campus, Islamabad, Pakistan
| |
Collapse
|
54
|
Manka SW, Bihan D, Farndale RW. Structural studies of the MMP-3 interaction with triple-helical collagen introduce new roles for the enzyme in tissue remodelling. Sci Rep 2019; 9:18785. [PMID: 31827179 PMCID: PMC6906530 DOI: 10.1038/s41598-019-55266-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 11/14/2019] [Indexed: 11/19/2022] Open
Abstract
Matrix metalloproteinase-3 (MMP-3) participates in normal extracellular matrix turnover during embryonic development, organ morphogenesis and wound healing, and in tissue-destruction associated with aneurysm, cancer, arthritis and heart failure. Despite its inability to cleave triple-helical collagens, MMP-3 can still bind to them, but the mechanism, location and role of binding are not known. We used the Collagen Toolkits, libraries of triple-helical peptides that embrace the entire helical domains of collagens II and III, to map MMP-3 interaction sites. The enzyme recognises five sites on collagen II and three sites on collagen III. They share a glycine-phenylalanine-hydroxyproline/alanine (GFO/A) motif that is recognised by the enzyme in a context-dependent manner. Neither MMP-3 zymogen (proMMP-3) nor the individual catalytic (Cat) and hemopexin (Hpx) domains of MMP-3 interact with the peptides, revealing cooperative binding of both domains to the triple helix. The Toolkit binding data combined with molecular modelling enabled us to deduce the putative collagen-binding mode of MMP-3, where all three collagen chains make contacts with the enzyme in the valley running across both Cat and Hpx domains. The observed binding pattern casts light on how MMP-3 could regulate collagen turnover and compete with various collagen-binding proteins regulating cell adhesion and proliferation.
Collapse
Affiliation(s)
- Szymon W Manka
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
- MRC Prion Unit at UCL, Institute of Prion Diseases, 33 Cleveland Street, London, W1W 7FF, UK.
| | - Dominique Bihan
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
55
|
Kao L, Azimov R, Shao XM, Abuladze N, Newman D, Zhekova H, Noskov S, Pushkin A, Kurtz I. SLC4A11 function: evidence for H +(OH -) and NH 3-H + transport. Am J Physiol Cell Physiol 2019; 318:C392-C405. [PMID: 31774702 PMCID: PMC7052617 DOI: 10.1152/ajpcell.00425.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Whether SLC4A11 transports ammonia and its potential mode of ammonia transport (NH4+, NH3, or NH3-2H+ transport have been proposed) are controversial. In the absence of ammonia, whether SLC4A11 mediates significant conductive H+(OH-) transport is also controversial. The present study was performed to determine the mechanism of human SLC4A11 ammonia transport and whether the transporter mediates conductive H+(OH-) transport in the absence of ammonia. We quantitated H+ flux by monitoring changes in intracellular pH (pHi) and measured whole cell currents in patch-clamp studies of HEK293 cells expressing the transporter in the absence and presence of NH4Cl. Our results demonstrate that SLC4A11 mediated conductive H+(OH-) transport that was stimulated by raising the extracellular pH (pHe). Ammonia-induced HEK293 whole cell currents were also stimulated by an increase in pHe. In studies using increasing NH4Cl concentrations with equal NH4+ extracellular and intracellular concentrations, the shift in the reversal potential (Erev) due to the addition of ammonia was compatible with NH3-H+ transport competing with H+(OH-) rather than NH3-nH+ (n ≥ 2) transport. The increase in equivalent H+(OH-) flux observed in the presence of a transcellular H+ gradient was also compatible with SLC4A11-mediated NH3-H+ flux. The NH3 versus Erev data fit a theoretical model suggesting that NH3-H+ and H+(OH-) competitively interact with the transporter. Studies of mutant SLC4A11 constructs in the putative SLC4A11 ion coordination site showed that both H+(OH-) transport and ammonia-induced whole cell currents were blocked suggesting that the H+(OH-) and NH3-H+ transport processes share common features involving the SLC4A11 transport mechanism.
Collapse
Affiliation(s)
- Liyo Kao
- Department of Medicine, Division of Nephrology, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Rustam Azimov
- Department of Medicine, Division of Nephrology, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Xuesi M Shao
- Department of Neurobiology, University of California, Los Angeles, California
| | - Natalia Abuladze
- Department of Medicine, Division of Nephrology, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Debra Newman
- Department of Medicine, Division of Nephrology, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Hristina Zhekova
- Department of Biological Sciences, Centre for Molecular Simulation, University of Calgary, Calgary, Alberta, Canada
| | - Sergei Noskov
- Department of Biological Sciences, Centre for Molecular Simulation, University of Calgary, Calgary, Alberta, Canada
| | - Alexander Pushkin
- Department of Medicine, Division of Nephrology, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Ira Kurtz
- Department of Medicine, Division of Nephrology, David Geffen School of Medicine, University of California, Los Angeles, California.,Brain Research Institute, University of California, Los Angeles, California
| |
Collapse
|
56
|
Teder T, Samel N, Lõhelaid H. Distinct characteristics of the substrate binding between highly homologous catalase-related allene oxide synthase and hydroperoxide lyase. Arch Biochem Biophys 2019; 676:108126. [PMID: 31589830 DOI: 10.1016/j.abb.2019.108126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/30/2019] [Accepted: 10/02/2019] [Indexed: 11/15/2022]
Abstract
A catalase-related allene oxide synthase (cAOS) or a hydroperoxide lyase (cHPL) fused together with an 8R-lipoxygenase is involved in the stress signaling of corals via an arachidonic acid pathway. cAOS gives rise to α-ketol and cyclopentenone, while cHPL catalyzes the cleavage of 8R-hydroperoxyeicosatetraenoic acid (8R-HpETE) to C8-oxo acid and C12 aldehyde. In silico analysis of the substrate entry sites of highly identical coral cAOS and cHPL indicated that two positively charged residues of cAOS, K60 and K107, and the corresponding residues of cHPL, E60 and K107, may be involved in the anchoring of the carboxy group of polyunsaturated fatty acid (PUFA) hydroperoxides. A mutational analysis of cAOS and cHPL revealed that K60 or E60 and K107 were not necessary in the tethering of 8R-HpETE, however, the E60 of cHPL was essential in the productive binding of PUFA hydroperoxides. The substrate preferences of cAOS and cHPL were determined with hydroperoxy derivatives of C18, C20, C22 PUFAs, anandamide (AEA), 1-arachidonoyl glycerol (1-AG) and selected methylated substrates. Although cAOS and cHPL were able to metabolize different free PUFA substrates and arachidonoyl derivatives, only cHPL catalyzed the reaction with methylated PUFA hydroperoxides. The differences in the substrate binding and preferences between cAOS and cHPL can be explained by the distinct properties of their substrate entry sites. The current study demonstrated that homologous PUFA metabolizing enzymes may contribute to the versatile usage of the substrate pool.
Collapse
Affiliation(s)
- Tarvi Teder
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Nigulas Samel
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Helike Lõhelaid
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia.
| |
Collapse
|
57
|
Skwark MJ, Torres PHM, Copoiu L, Bannerman B, Floto RA, Blundell TL. Mabellini: a genome-wide database for understanding the structural proteome and evaluating prospective antimicrobial targets of the emerging pathogen Mycobacterium abscessus. Database (Oxford) 2019; 2019:5611286. [PMID: 31681953 PMCID: PMC6853642 DOI: 10.1093/database/baz113] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/31/2019] [Accepted: 08/28/2019] [Indexed: 02/02/2023]
Abstract
Mycobacterium abscessus, a rapid growing, multidrug resistant, nontuberculous mycobacteria, can cause a wide range of opportunistic infections, particularly in immunocompromised individuals. M. abscessus has emerged as a growing threat to patients with cystic fibrosis, where it causes accelerated inflammatory lung damage, is difficult and sometimes impossible to treat and can prevent safe transplantation. There is therefore an urgent unmet need to develop new therapeutic strategies. The elucidation of the M. abscessus genome in 2009 opened a wide range of research possibilities in the field of drug discovery that can be more effectively exploited upon the characterization of the structural proteome. Where there are no experimental structures, we have used the available amino acid sequences to create 3D models of the majority of the remaining proteins that constitute the M. abscessus proteome (3394 proteins and over 13 000 models) using a range of up-to-date computational tools, many developed by our own group. The models are freely available for download in an on-line database, together with quality data and functional annotation. Furthermore, we have developed an intuitive and user-friendly web interface (http://www.mabellinidb.science) that enables easy browsing, querying and retrieval of the proteins of interest. We believe that this resource will be of use in evaluating the prospective targets for design of antimicrobial agents and will serve as a cornerstone to support the development of new molecules to treat M. abscessus infections.
Collapse
Affiliation(s)
- Marcin J Skwark
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| | - Pedro H M Torres
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| | - Liviu Copoiu
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| | - Bridget Bannerman
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| | - R Andres Floto
- Molecular Immunity Unit, Department of Medicine University of Cambridge, MRC-Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
and,Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge CB23 3RE, UK
| | - Tom L Blundell
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK,Corresponding author: Tel: +44 1223 333628; Fax: +44 1223 766002;
| |
Collapse
|
58
|
Balasubramanian PK, Lee Y, Kim Y. Identification of Ligand‐binding Hotspot Residues of CDK4 Using Molecular Docking and Molecular Dynamics Simulation. B KOREAN CHEM SOC 2019. [DOI: 10.1002/bkcs.11873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
| | - Yeongjoon Lee
- Department of Bioscience and BiotechnologyKonkuk University Seoul 05029 South Korea
| | - Yangmee Kim
- Department of Bioscience and BiotechnologyKonkuk University Seoul 05029 South Korea
| |
Collapse
|
59
|
Green CM, Li Z, Smith AD, Novikova O, Bacot-Davis VR, Gao F, Hu S, Banavali NK, Thiele DJ, Li H, Belfort M. Spliceosomal Prp8 intein at the crossroads of protein and RNA splicing. PLoS Biol 2019; 17:e3000104. [PMID: 31600193 PMCID: PMC6805012 DOI: 10.1371/journal.pbio.3000104] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 10/22/2019] [Accepted: 09/13/2019] [Indexed: 01/07/2023] Open
Abstract
The spliceosome is a large ribonucleoprotein complex that removes introns from pre-mRNAs. At its functional core lies the essential pre-mRNA processing factor 8 (Prp8) protein. Across diverse eukaryotes, this protein cofactor of RNA catalysis harbors a self-splicing element called an intein. Inteins in Prp8 are extremely pervasive and are found at 7 different sites in various species. Here, we focus on the Prp8 intein from Cryptococcus neoformans (Cne), a human fungal pathogen. We solved the crystal structure of this intein, revealing structural homology among protein splicing sequences in eukaryotes, including the Hedgehog C terminus. Working with the Cne Prp8 intein in a reporter assay, we find that the biologically relevant divalent metals copper and zinc inhibit intein splicing, albeit by 2 different mechanisms. Copper likely stimulates reversible modifications on a catalytically important cysteine, whereas zinc binds at the terminal asparagine and the same critical cysteine. Importantly, we also show that copper treatment inhibits Prp8 protein splicing in Cne. Lastly, an intein-containing Prp8 precursor model is presented, suggesting that metal-induced protein splicing inhibition would disturb function of both Prp8 and the spliceosome. These results indicate that Prp8 protein splicing can be modulated, with potential functional implications for the spliceosome.
Collapse
Affiliation(s)
- Cathleen M. Green
- Department of Biological Sciences and RNA Institute, University at Albany, Albany, New York, United States of America
| | - Zhong Li
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
| | - Aaron D. Smith
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Olga Novikova
- Department of Biological Sciences and RNA Institute, University at Albany, Albany, New York, United States of America
| | - Valjean R. Bacot-Davis
- Department of Biological Sciences and RNA Institute, University at Albany, Albany, New York, United States of America
| | - Fengshan Gao
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
| | - Saiyang Hu
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
| | - Nilesh K. Banavali
- Division of Translational Medicine, Wadsworth Center, New York State Department of Health, Albany, New York, United States of America,Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, New York, United States of America
| | - Dennis J. Thiele
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, United States of America,Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America,Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Hongmin Li
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, New York, United States of America,Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, New York, United States of America,* E-mail: (MB); (HL)
| | - Marlene Belfort
- Department of Biological Sciences and RNA Institute, University at Albany, Albany, New York, United States of America,Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, New York, United States of America,* E-mail: (MB); (HL)
| |
Collapse
|
60
|
Gae DD, Budamagunta MS, Hess JF, McCarrick RM, Lorigan GA, FitzGerald PG, Voss JC. Completion of the Vimentin Rod Domain Structure Using Experimental Restraints: A New Tool for Exploring Intermediate Filament Assembly and Mutations. Structure 2019; 27:1547-1560.e4. [PMID: 31402219 PMCID: PMC6774864 DOI: 10.1016/j.str.2019.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 05/23/2019] [Accepted: 07/22/2019] [Indexed: 11/28/2022]
Abstract
Electron paramagnetic resonance (EPR) spectroscopy of full-length vimentin and X-ray crystallography of vimentin peptides has provided concordant structural data for nearly the entire central rod domain of the protein. In this report, we use a combination of EPR spectroscopy and molecular modeling to determine the structure and dynamics of the missing region and unite the separate elements into a single structure. Validation of the linker 1-2 (L1-2) modeling approach is demonstrated by the close correlation between EPR and X-ray data in the previously solved regions. Importantly, molecular dynamic (MD) simulation of the constructed model agrees with spin label motion as determined by EPR. Furthermore, MD simulation shows L1-2 heterogeneity, with a concerted switching of states among the dimer chains. These data provide the first ever experimentally driven model of a complete intermediate filament rod domain, providing research tools for further modeling and assembly studies.
Collapse
Affiliation(s)
- David D Gae
- Department of Surgery, School of Medicine, University of California San Francisco, San Francisco, CA 94118, USA
| | - Madhu S Budamagunta
- Department of Biochemistry & Molecular Medicine, School of Medicine, University of California Davis, Davis, CA 95616, USA
| | - John F Hess
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California Davis, Davis, CA 95616, USA
| | - Robert M McCarrick
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH 45056, USA
| | - Gary A Lorigan
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH 45056, USA
| | - Paul G FitzGerald
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California Davis, Davis, CA 95616, USA.
| | - John C Voss
- Department of Biochemistry & Molecular Medicine, School of Medicine, University of California Davis, Davis, CA 95616, USA.
| |
Collapse
|
61
|
Sasidharan S, Saudagar P. Biochemical and structural characterization of tyrosine aminotransferase suggests broad substrate specificity and a two-state folding mechanism in Leishmania donovani. FEBS Open Bio 2019; 9:1769-1783. [PMID: 31393078 PMCID: PMC6768288 DOI: 10.1002/2211-5463.12715] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/20/2019] [Accepted: 08/07/2019] [Indexed: 11/09/2022] Open
Abstract
Tyrosine aminotransferase (TAT) is an aminotransferase with broad substrate specificity that catalyzes the transamination of aromatic amino acids in Leishmania donovani and plays a crucial role in the survival and pathogenicity of the parasite. In this study, we have biochemically characterized tyrosine aminotransferase from Leishmania donovani using in vitro and in silico techniques. Leishmania donovani tyrosine aminotransferase (LdTAT) was cloned into the pET28a(+) vector and expressed in the BL21 strain of Escherichia coli. The Ni-NTA-purified protein was then characterized biochemically, and its various kinetic parameters were investigated. The apparent Km value for the tyrosine-pyruvate pair was determined to be 3.5 ± 0.9 mm, and Vmax was analyzed to be at 11.7 ± 1.5 μm·min.μg-1 . LdTAT was found to exhibit maximum activity at 50 °C and at a pH of 8.0. Cofactor identification for LdTAT showed that pyridoxal-5-phosphate (PLP) binds with a Km value of 23.59 ± 3.99 μm and that the phosphate group is vital for the activity of the enzyme. Sequence analysis revealed that S151, Y256, K286, and P291 are conserved residues and form hydrogen bonds with PLP. Urea-based denaturation studies revealed a biphasic folding mechanism involving N→X→D states. Molecular dynamic simulations of modeled LdTAT at various conditions were performed to understand enzyme behavior and interactions at the molecular level. The biochemical and structural divergence between host and parasite TAT suggests the LdTAT has evolved to utilize pyruvate rather than α-ketoglutarate as co-substrate. Furthermore, our data suggest that LdTAT may be a potential drug target due to its divergence in structure and substrate specificity from the host.
Collapse
Affiliation(s)
- Santanu Sasidharan
- Department of BiotechnologyNational Institute of TechnologyWarangalIndia
| | - Prakash Saudagar
- Department of BiotechnologyNational Institute of TechnologyWarangalIndia
| |
Collapse
|
62
|
Abstract
Background: :
One of the best known to date GPCR class A (Rhodopsin) includes more
than 100 orphan receptors for which the endogenous ligand is not known or is unclear. One of them
is N-arachidonyl glycine receptor, named GPR18, a receptor that has been reported to be activated
by Δ9-THC, endogenous cannabinoid receptors agonist anandamide and other cannabinoid receptor
ligands suggesting it could be considered as third cannabinoid receptor. GPR18 activity, as well as
its distribution might suggest usage of GPR18 ligands in treatment of endometriosis, cancer, and
neurodegenerative disorders. Yet, so far only few GPR18 antagonists have been described, thus
only ligand-based design approaches appear to be most useful to identify new ligands for this orphan
receptor.
Methods: :
Main goal of this study, GPR18 inactive form homology model was built on the basis of
the evolutionary closest homologous template: Human P2Y1 Receptor crystal structure.
Results: :
Obtained model was further evaluated and showed active/nonactive ligands differentiating
properties with acceptable confidence. Moreover, it allowed for preliminary assessment of proteinligand
interactions for a set of previously described ligands.
Conclusion::
Thus collected data might serve as a starting point for a discovery of novel, active
GPR18 blocking ligands.
Collapse
Affiliation(s)
- Kamil J. Kuder
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Faculty of Pharmacy, Medyczna 9, 30-688 Kraków, Poland
| | - Tadeusz Karcz
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Faculty of Pharmacy, Medyczna 9, 30-688 Kraków, Poland
| | - Maria Kaleta
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Faculty of Pharmacy, Medyczna 9, 30-688 Kraków, Poland
| | - Katarzyna Kiec-Kononowicz
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Faculty of Pharmacy, Medyczna 9, 30-688 Kraków, Poland
| |
Collapse
|
63
|
De Las Rivas J, Bonavides-Martínez C, Campos-Laborie FJ. Bioinformatics in Latin America and SoIBio impact, a tale of spin-off and expansion around genomes and protein structures. Brief Bioinform 2019; 20:390-397. [PMID: 28981567 PMCID: PMC6433739 DOI: 10.1093/bib/bbx064] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/18/2017] [Indexed: 11/30/2022] Open
Abstract
Owing to the emerging impact of bioinformatics and computational biology, in this article, we present an overview of the history and current state of the research on this field in Latin America (LA). It will be difficult to cover without inequality all the efforts, initiatives and works that have happened for the past two decades in this vast region (that includes >19 million km2 and >600 million people). Despite the difficulty, we have done an analytical search looking for publications in the field made by researchers from 19 LA countries in the past 25 years. In this way, we find that research in bioinformatics in this region should develop twice to approach the average world scientific production in the field. We also found some of the pioneering scientists who initiated and led bioinformatics in the region and were promoters of this new scientific field. Our analysis also reveals that spin-off began around some specific areas within the biomolecular sciences: studies on genomes (anchored in the new generation of deep sequencing technologies, followed by developments in proteomics) and studies on protein structures (supported by three-dimensional structural determination technologies and their computational advancement). Finally, we show that the contribution to this endeavour of the Iberoamerican Society for Bioinformatics, founded in Mexico in 2009, has been significant, as it is a leading forum to join efforts of many scientists from LA interested in promoting research, training and education in bioinformatics.
Collapse
Affiliation(s)
- Javier De Las Rivas
- CSIC and Universidad de Salamanca, Bioinformatics and Functional Genomics Group, Cancer Research Center (IMBCC, CSIC/USAL/IBSAL), Salamanca, Spain
- Corresponding author. Javier De Las Rivas, Bioinformatics and Functional Genomics Group, Cancer Research Center (IMBCC, CSIC/USAL/IBSAL), Consejo Superior de Investigaciones Científicas (CSIC) and Universidad de Salamanca (USAL), Campus Miguel de Unamuno s/n, Salamanca 37007, Spain. Tel.: +34 923294819; Fax: +34923294743; E-mail:
| | - Cesar Bonavides-Martínez
- Universidad Nacional Autonoma de Mexico, Computational Genomics, Centro de Ciencias Genómicas, Cuernavaca, Morelos, Mexico
| | - Francisco Jose Campos-Laborie
- CSIC and Universidad de Salamanca, Bioinformatics and Functional Genomics Group, Cancer Research Center (IMBCC, CSIC/USAL/IBSAL), Salamanca, Spain
| |
Collapse
|
64
|
Almeida JM, Martini VP, Iulek J, Alnoch RC, Moure VR, Müller-Santos M, Souza EM, Mitchell DA, Krieger N. Biochemical characterization and application of a new lipase and its cognate foldase obtained from a metagenomic library derived from fat-contaminated soil. Int J Biol Macromol 2019; 137:442-454. [DOI: 10.1016/j.ijbiomac.2019.06.203] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 06/24/2019] [Accepted: 06/26/2019] [Indexed: 12/17/2022]
|
65
|
Veillet F, Chauvin L, Kermarrec MP, Sevestre F, Merrer M, Terret Z, Szydlowski N, Devaux P, Gallois JL, Chauvin JE. The Solanum tuberosum GBSSI gene: a target for assessing gene and base editing in tetraploid potato. PLANT CELL REPORTS 2019; 38:1065-1080. [PMID: 31101972 DOI: 10.1007/s00299-019-02426-w] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/08/2019] [Accepted: 05/08/2019] [Indexed: 05/20/2023]
Abstract
The StGBSSI gene was successfully and precisely edited in the tetraploid potato using gene and base-editing strategies, leading to plants with impaired amylose biosynthesis. Genome editing has recently become a method of choice for basic research and functional genomics, and holds great potential for molecular plant-breeding applications. The powerful CRISPR-Cas9 system that typically produces double-strand DNA breaks is mainly used to generate knockout mutants. Recently, the development of base editors has broadened the scope of genome editing, allowing precise and efficient nucleotide substitutions. In this study, we produced mutants in two cultivated elite cultivars of the tetraploid potato (Solanum tuberosum) using stable or transient expression of the CRISPR-Cas9 components to knock out the amylose-producing StGBSSI gene. We set up a rapid, highly sensitive and cost-effective screening strategy based on high-resolution melting analysis followed by direct Sanger sequencing and trace chromatogram analysis. Most mutations consisted of small indels, but unwanted insertions of plasmid DNA were also observed. We successfully created tetra-allelic mutants with impaired amylose biosynthesis, confirming the loss of function of the StGBSSI protein. The second main objective of this work was to demonstrate the proof of concept of CRISPR-Cas9 base editing in the tetraploid potato by targeting two loci encoding catalytic motifs of the StGBSSI enzyme. Using a cytidine base editor (CBE), we efficiently and precisely induced DNA substitutions in the KTGGL-encoding locus, leading to discrete variation in the amino acid sequence and generating a loss-of-function allele. The successful application of base editing in the tetraploid potato opens up new avenues for genome engineering in this species.
Collapse
Affiliation(s)
- Florian Veillet
- INRA, Agrocampus Ouest, Université Rennes 1, UMR 1349 IGEPP, Domaine de Kéraïber, 29260, Ploudaniel, France.
| | - Laura Chauvin
- INRA, Agrocampus Ouest, Université Rennes 1, UMR 1349 IGEPP, Domaine de Kéraïber, 29260, Ploudaniel, France
| | - Marie-Paule Kermarrec
- INRA, Agrocampus Ouest, Université Rennes 1, UMR 1349 IGEPP, Domaine de Kéraïber, 29260, Ploudaniel, France
| | - François Sevestre
- Unité de Glycobiologie Structurale et Fonctionnelle, Univ. Lille, CNRS, UMR8576, UGSF, Lille, France
- Univ. Lille, CNRS, USR 3290, MSAP, Miniaturisation pour la Synthèse, l'Analyse et la Protéomique, Lille, France
| | - Mathilde Merrer
- INRA, Agrocampus Ouest, Université Rennes 1, UMR 1349 IGEPP, Domaine de Kéraïber, 29260, Ploudaniel, France
| | - Zoé Terret
- GAFL, INRA, Montfavet, France
- SYNGENTA SEEDS SAS, 346 Route des Pasquiers, 84260, Sarrians, France
| | - Nicolas Szydlowski
- Unité de Glycobiologie Structurale et Fonctionnelle, Univ. Lille, CNRS, UMR8576, UGSF, Lille, France
- Univ. Lille, CNRS, USR 3290, MSAP, Miniaturisation pour la Synthèse, l'Analyse et la Protéomique, Lille, France
| | - Pierre Devaux
- Germicopa Breeding, Kerguivarc'h, 29520, Chateauneuf Du Faou, France
| | | | - Jean-Eric Chauvin
- INRA, Agrocampus Ouest, Université Rennes 1, UMR 1349 IGEPP, Domaine de Kéraïber, 29260, Ploudaniel, France
| |
Collapse
|
66
|
Raj S, Sasidharan S, Dubey VK, Saudagar P. Identification of lead molecules against potential drug target protein MAPK4 from L. donovani: An in-silico approach using docking, molecular dynamics and binding free energy calculation. PLoS One 2019; 14:e0221331. [PMID: 31425543 PMCID: PMC6699710 DOI: 10.1371/journal.pone.0221331] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 08/06/2019] [Indexed: 11/25/2022] Open
Abstract
Leishmaniasis caused by obligate intracellular parasites of genus Leishmania is one of the most neglected tropical diseases threatening 350 million people worldwide. Protein kinases have drawn much attention as potential drug targets due to their important role in various cellular processes. In Leishmania sp. mitogen-activated protein kinase 4 is essential for the parasite survival because of its involvement in various regulatory, apoptotic and developmental pathways. The current study reveals the identification of natural inhibitors of L. donovani mitogen-activated protein kinase-4 (LdMPK4). We have performed in silico docking of 110 natural inhibitors of Leishmania parasite that have been reported earlier and identified two compounds Genistein (GEN) and Chrysin (CHY). The homology model of LdMPK4 was developed, followed by binding affinity studies, and pharmacokinetic properties of the inhibitors were calculated by maintaining ATP as a standard molecule. The modelled structure was deposited in the protein model database with PMDB ID: PM0080988. Molecular dynamic simulation of the enzyme-inhibitor complex along with the free energy calculations over 50 ns showed that GEN and CHY are more stable in their binding. These two molecules, GEN and CHY, can be considered as lead molecules for targeting LdMPK4 enzyme and could emerge as potential LdMPK4 inhibitors.
Collapse
Affiliation(s)
- Shweta Raj
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal, (T.S.), India
| | - Santanu Sasidharan
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal, (T.S.), India
| | - Vikash Kumar Dubey
- School of Biochemical Engineering, Indian Institute of Technology-Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Prakash Saudagar
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal, (T.S.), India
| |
Collapse
|
67
|
Sadhasivam A, Nagarajan H, Umashankar V. Structure-based drug target prioritisation and rational drug design for targeting Chlamydia trachomatis eye infections. J Biomol Struct Dyn 2019; 38:3131-3143. [PMID: 31380730 DOI: 10.1080/07391102.2019.1652691] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Chlamydia trachomatis (C.t) is a major causative of infectious blindness in world. It is a real challenge to combat Chlamydial infection as it is an intracellular pathogen. Hence, it is essential to determine the most potential targets of C.t in order to inhibit or suppress its virulence during its infectious phase. Thus, in this study, the highly expressed-cum-most essential genes reported through our earlier study were reprioritized by structure-based comparative binding site analysis with host proteome. Therefore, computational approaches involving molecular modelling, large-scale binding site prediction and comparison, molecular dynamics simulation studies were performed to narrow down the most potential targets. Furthermore, high-throughput virtual screening and ADMETox were also performed to identify potential hits that shall efficiently inhibit the prioritised targets. Hence, by this study we report Pyruvoyl-dependent arginine decarboxylase (PvlArgDC), DNA-repair protein (RecO) and porin (outer membrane protein) as the most viable targets of C.t which can be potentially targeted by compounds, NSC_13086, MFCD00276409, MFCD05662003, respectively. AbbreviationsC.tChlamydia trachomatisSTDSexually transmitted diseaseHTVSHigh-throughput virtual screeningADMEToxAbsorption, Distribution, Metabolism, Excretion and ToxicityPMPocketMatchMDMolecular Dynamics simulationSPStandard precisionXPExtra precisionMMGBSAMolecular mechanics energies combined with generalised Born and surface area continuum solvationOMPOuter membrane proteinPvlArgDCPyruvoyl-dependent arginine decarboxylaseRecORecombination protein O.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Anupriya Sadhasivam
- Centre for Bioinformatics, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology, Vision Research Foundation, Chennai, India
| | - Hemavathy Nagarajan
- Centre for Bioinformatics, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology, Vision Research Foundation, Chennai, India
| | - Vetrivel Umashankar
- Centre for Bioinformatics, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology, Vision Research Foundation, Chennai, India
| |
Collapse
|
68
|
Solis AD. Reduced alphabet of prebiotic amino acids optimally encodes the conformational space of diverse extant protein folds. BMC Evol Biol 2019; 19:158. [PMID: 31362700 PMCID: PMC6668081 DOI: 10.1186/s12862-019-1464-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 06/19/2019] [Indexed: 11/10/2022] Open
Abstract
Background There is wide agreement that only a subset of the twenty standard amino acids existed prebiotically in sufficient concentrations to form functional polypeptides. We ask how this subset, postulated as {A,D,E,G,I,L,P,S,T,V}, could have formed structures stable enough to found metabolic pathways. Inspired by alphabet reduction experiments, we undertook a computational analysis to measure the structural coding behavior of sequences simplified by reduced alphabets. We sought to discern characteristics of the prebiotic set that would endow it with unique properties relevant to structure, stability, and folding. Results Drawing on a large dataset of single-domain proteins, we employed an information-theoretic measure to assess how well the prebiotic amino acid set preserves fold information against all other possible ten-amino acid sets. An extensive virtual mutagenesis procedure revealed that the prebiotic set excellently preserves sequence-dependent information regarding both backbone conformation and tertiary contact matrix of proteins. We observed that information retention is fold-class dependent: the prebiotic set sufficiently encodes the structure space of α/β and α + β folds, and to a lesser extent, of all-α and all-β folds. The prebiotic set appeared insufficient to encode the small proteins. Assessing how well the prebiotic set discriminates native vs. incorrect sequence-structure matches, we found that α/β and α + β folds exhibit more pronounced energy gaps with the prebiotic set than with nearly all alternatives. Conclusions The prebiotic set optimally encodes local backbone structures that appear in the folded environment and near-optimally encodes the tertiary contact matrix of extant proteins. The fold-class-specific patterns observed from our structural analysis confirm the postulated timeline of fold appearance in proteogenesis derived from proteomic sequence analyses. Polypeptides arising in a prebiotic environment will likely form α/β and α + β-like folds if any at all. We infer that the progressive expansion of the alphabet allowed the increased conformational stability and functional specificity of later folds, including all-α, all-β, and small proteins. Our results suggest that prebiotic sequences are amenable to mutations that significantly lower native conformational energies and increase discrimination amidst incorrect folds. This property may have assisted the genesis of functional proto-enzymes prior to the expansion of the full amino acid alphabet.
Collapse
Affiliation(s)
- Armando D Solis
- Biological Sciences Department, New York City College of Technology (City Tech), The City University of New York (CUNY), 285 Jay Street, Brooklyn, NY, 11201, USA.
| |
Collapse
|
69
|
Fierro F, Giorgetti A, Carloni P, Meyerhof W, Alfonso-Prieto M. Dual binding mode of "bitter sugars" to their human bitter taste receptor target. Sci Rep 2019; 9:8437. [PMID: 31186454 PMCID: PMC6560132 DOI: 10.1038/s41598-019-44805-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 05/22/2019] [Indexed: 12/21/2022] Open
Abstract
The 25 human bitter taste receptors (hTAS2Rs) are responsible for detecting bitter molecules present in food, and they also play several physiological and pathological roles in extraoral compartments. Therefore, understanding their ligand specificity is important both for food research and for pharmacological applications. Here we provide a molecular insight into the exquisite molecular recognition of bitter β-glycopyranosides by one of the members of this receptor subclass, hTAS2R16. Most of its agonists have in common the presence of a β-glycopyranose unit along with an extremely structurally diverse aglycon moiety. This poses the question of how hTAS2R16 can recognize such a large number of "bitter sugars". By means of hybrid molecular mechanics/coarse grained molecular dynamics simulations, here we show that the three hTAS2R16 agonists salicin, arbutin and phenyl-β-D-glucopyranoside interact with the receptor through a previously unrecognized dual binding mode. Such mechanism may offer a seamless way to fit different aglycons inside the binding cavity, while maintaining the sugar bound, similar to the strategy used by several carbohydrate-binding lectins. Our prediction is validated a posteriori by comparison with mutagenesis data and also rationalizes a wealth of structure-activity relationship data. Therefore, our findings not only provide a deeper molecular characterization of the binding determinants for the three ligands studied here, but also give insights applicable to other hTAS2R16 agonists. Together with our results for other hTAS2Rs, this study paves the way to improve our overall understanding of the structural determinants of ligand specificity in bitter taste receptors.
Collapse
Affiliation(s)
- Fabrizio Fierro
- Computational Biomedicine, Institute for Advanced Simulation IAS-5 and Institute of Neuroscience and Medicine INM-9, Forschungszentrum Jülich, Jülich, Germany
- Department of Biology, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Alejandro Giorgetti
- Computational Biomedicine, Institute for Advanced Simulation IAS-5 and Institute of Neuroscience and Medicine INM-9, Forschungszentrum Jülich, Jülich, Germany
- Department of Biotechnology, University of Verona, Verona, Italy
- JARA-HPC, IAS-5/INM-9 Computational Biomedicine, Forschungszentrum Jülich GmbH, Jülich, 52425, Germany
| | - Paolo Carloni
- Computational Biomedicine, Institute for Advanced Simulation IAS-5 and Institute of Neuroscience and Medicine INM-9, Forschungszentrum Jülich, Jülich, Germany
- JARA-HPC, IAS-5/INM-9 Computational Biomedicine, Forschungszentrum Jülich GmbH, Jülich, 52425, Germany
- Department of Physics, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
- VNU Key Laboratory "Multiscale Simulation of Complex Systems", VNU University of Science, Vietnam National University, Hanoi, Vietnam
| | - Wolfgang Meyerhof
- Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Mercedes Alfonso-Prieto
- Computational Biomedicine, Institute for Advanced Simulation IAS-5 and Institute of Neuroscience and Medicine INM-9, Forschungszentrum Jülich, Jülich, Germany.
- JARA-HPC, IAS-5/INM-9 Computational Biomedicine, Forschungszentrum Jülich GmbH, Jülich, 52425, Germany.
- Cécile and Oskar Vogt Institute for Brain Research, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
70
|
Mohammadyari H, Shirdel SA, Jafarian V, Khalifeh K. Designing and construction of novel variants of Chondroitinase ABC I to reduce aggregation rate. Arch Biochem Biophys 2019; 668:46-53. [PMID: 31103558 DOI: 10.1016/j.abb.2019.05.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/11/2019] [Accepted: 05/15/2019] [Indexed: 01/25/2023]
Abstract
Chondroitinase ABC I (cABC I) can degrade inhibitory molecules for axon regrowth at the site of damage after spinal cord injury (SCI). One of the main problems in the practical application is the possibility of structural changes that lead to the inactivation of the enzyme. In current work, three variants of cABC I was designed and constructed by manipulation of a short helix conformation (Gln678-Leu679-Ser680-Gln681); where Gln residues were converted to Glu. According to the enzyme kinetics studies, the catalytic efficiency of the Q681E and double mutant (Q678E/Q681E) increases in comparison with WT enzyme; while that of Q678E decreases. It was also shown that the rate of the inactivation of the enzyme variants over time is greater in WT and Q678E variants than that of the Q681E and double mutant. Negative values of entropy change of thermal inactivation measurements; demonstrate that inactivation of the WT and Q678E variants are mainly originated from aggregation. These observations can be explained by considering the repulsive electrostatic interaction between enzyme molecules that prevents protein aggregation over time. It is concluded that increasing the solubility of the Q681E and double mutant via favorable interactions of surface-exposed charged residues with dipole momentum of water molecules accompanied by the presence of intermolecular repulsive electrostatic interaction leads to decreasing the rate of aggregation in both long-term storage and heat-induced structural changes.
Collapse
Affiliation(s)
- Hamed Mohammadyari
- Department of Biology, Faculty of Sciences, University of Zanjan, Zanjan, Iran
| | - S Akram Shirdel
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Vahab Jafarian
- Department of Biology, Faculty of Sciences, University of Zanjan, Zanjan, Iran
| | - Khosrow Khalifeh
- Department of Biology, Faculty of Sciences, University of Zanjan, Zanjan, Iran.
| |
Collapse
|
71
|
Alfonso-Prieto M, Navarini L, Carloni P. Understanding Ligand Binding to G-Protein Coupled Receptors Using Multiscale Simulations. Front Mol Biosci 2019; 6:29. [PMID: 31131282 PMCID: PMC6510167 DOI: 10.3389/fmolb.2019.00029] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 04/11/2019] [Indexed: 12/18/2022] Open
Abstract
Human G-protein coupled receptors (GPCRs) convey a wide variety of extracellular signals inside the cell and they are one of the main targets for pharmaceutical intervention. Rational drug design requires structural information on these receptors; however, the number of experimental structures is scarce. This gap can be filled by computational models, based on homology modeling and docking techniques. Nonetheless, the low sequence identity across GPCRs and the chemical diversity of their ligands may limit the quality of these models and hence refinement using molecular dynamics simulations is recommended. This is the case for olfactory and bitter taste receptors, which constitute the first and third largest GPCR groups and show sequence identities with the available GPCR templates below 20%. We have developed a molecular dynamics approach, based on the combination of molecular mechanics and coarse grained (MM/CG), tailored to study ligand binding in GPCRs. This approach has been applied so far to bitter taste receptor complexes, showing significant predictive power. The protein/ligand interactions observed in the simulations were consistent with extensive mutagenesis and functional data. Moreover, the simulations predicted several binding residues not previously tested, which were subsequently verified by carrying out additional experiments. Comparison of the simulations of two bitter taste receptors with different ligand selectivity also provided some insights into the binding determinants of bitter taste receptors. Although the MM/CG approach has been applied so far to a limited number of GPCR/ligand complexes, the excellent agreement of the computational models with the mutagenesis and functional data supports the applicability of this method to other GPCRs for which experimental structures are missing. This is particularly important for the challenging case of GPCRs with low sequence identity with available templates, for which molecular docking shows limited predictive power.
Collapse
Affiliation(s)
- Mercedes Alfonso-Prieto
- Institute for Advanced Simulation IAS-5 and Institute of Neuroscience and Medicine INM-9, Computational Biomedicine, Forschungszentrum Jülich, Jülich, Germany.,Medical Faculty, Cécile and Oskar Vogt Institute for Brain Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | | | - Paolo Carloni
- Institute for Advanced Simulation IAS-5 and Institute of Neuroscience and Medicine INM-9, Computational Biomedicine, Forschungszentrum Jülich, Jülich, Germany.,Institute for Neuroscience and Medicine INM-11, Forschungszentrum Jülich, Jülich, Germany.,Department of Physics, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany.,VNU Key Laboratory "Multiscale Simulation of Complex Systems", VNU University of Science, Vietnam National University, Hanoi, Vietnam
| |
Collapse
|
72
|
Battisti A, Zamuner S, Sarti E, Laio A. Toward a unified scoring function for native state discrimination and drug-binding pocket recognition. Phys Chem Chem Phys 2019; 20:17148-17155. [PMID: 29900428 DOI: 10.1039/c7cp08170g] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Protein folding and receptor-ligand recognition are fundamental processes for any living organism. Although folding and ligand recognition are based on the same chemistry, the existing empirical scoring functions target just one problem: predicting the correct fold or the correct binding pose. We here introduce a statistical potential which considers moieties as fundamental units. The scoring function is able to deal with both folding and ligand pocket recognition problems with a performance comparable to the scoring functions specifically tailored for one of the two tasks. We foresee that the capability of the new scoring function to tackle both problems in a unified framework will be a key to deal with the induced fit phenomena, in which a target protein changes significantly its conformation upon binding. Moreover, the new scoring function might be useful in docking protocols towards intrinsically disordered proteins, whose flexibility cannot be handled with the available docking software.
Collapse
Affiliation(s)
- Anna Battisti
- International School for Advanced Studies (SISSA), Via Bonomea 265, I-34136 Trieste, Italy.
| | | | | | | |
Collapse
|
73
|
López-Blanco JR, Chacón P. KORP: knowledge-based 6D potential for fast protein and loop modeling. Bioinformatics 2019; 35:3013-3019. [DOI: 10.1093/bioinformatics/btz026] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 01/03/2019] [Accepted: 01/08/2019] [Indexed: 12/18/2022] Open
Abstract
Abstract
Motivation
Knowledge-based statistical potentials constitute a simpler and easier alternative to physics-based potentials in many applications, including folding, docking and protein modeling. Here, to improve the effectiveness of the current approximations, we attempt to capture the six-dimensional nature of residue–residue interactions from known protein structures using a simple backbone-based representation.
Results
We have developed KORP, a knowledge-based pairwise potential for proteins that depends on the relative position and orientation between residues. Using a minimalist representation of only three backbone atoms per residue, KORP utilizes a six-dimensional joint probability distribution to outperform state-of-the-art statistical potentials for native structure recognition and best model selection in recent critical assessment of protein structure prediction and loop-modeling benchmarks. Compared with the existing methods, our side-chain independent potential has a lower complexity and better efficiency. The superior accuracy and robustness of KORP represent a promising advance for protein modeling and refinement applications that require a fast but highly discriminative energy function.
Availability and implementation
http://chaconlab.org/modeling/korp.
Supplementary information
Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- José Ramón López-Blanco
- Department of Biological Chemical Physics, Rocasolano Institute of Physical Chemistry C.S.I.C, Madrid, Spain
| | - Pablo Chacón
- Department of Biological Chemical Physics, Rocasolano Institute of Physical Chemistry C.S.I.C, Madrid, Spain
| |
Collapse
|
74
|
Aliakbari N, Mirzaee Z, Jafarian V, Khalifeh K, Salehi M. Genetic and Biochemical Characterization of a Novel Thermostable Cyclomaltodextrinase From Anoxybacillus flavithermus. STARCH-STARKE 2019. [DOI: 10.1002/star.201800133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Neda Aliakbari
- Faculty of Sciences, Department of Biology, University of Zanjan; Zanjan I. R. Iran
| | - Ziba Mirzaee
- Faculty of Sciences, Department of Biology, University of Zanjan; Zanjan I. R. Iran
| | - Vahab Jafarian
- Faculty of Sciences, Department of Biology, University of Zanjan; Zanjan I. R. Iran
| | - Khosrow Khalifeh
- Faculty of Sciences, Department of Biology, University of Zanjan; Zanjan I. R. Iran
| | - Mehdi Salehi
- Faculty of Sciences, Department of Biology, University of Zanjan; Zanjan I. R. Iran
| |
Collapse
|
75
|
Zhuravlev AV, Vetrovoy OV, Savvateeva-Popova EV. Enzymatic and non-enzymatic pathways of kynurenines' dimerization: the molecular factors for oxidative stress development. PLoS Comput Biol 2018; 14:e1006672. [PMID: 30532237 PMCID: PMC6301705 DOI: 10.1371/journal.pcbi.1006672] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 12/20/2018] [Accepted: 11/26/2018] [Indexed: 11/19/2022] Open
Abstract
Kynurenines, the products of tryptophan oxidative degradation, are involved in multiple neuropathologies, such as Huntington's chorea, Parkinson's disease, senile dementia, etc. The major cause for hydroxykynurenines's neurotoxicity is the oxidative stress induced by the reactive oxygen species (ROS), the by-products of L-3-hydroxykynurenine (L-3HOK) and 3-hydroxyanthranilic acid (3HAA) oxidative self-dimerization. 2-aminophenol (2AP), a structural precursor of L-3HOK and 3HAA, undergoes the oxidative conjugation to form 2-aminophenoxazinone. There are several modes of 2AP dimerization, including both enzymatic and non-enzymatic stages. In this study, the free energies for 2AP, L-3HOK and 3HAA dimerization stages have been calculated at B3LYP/6-311G(d,p)//6-311+(O)+G(d) level, both in the gas phase and in heptane or water solution. For the intermediates, ionization potentials and electron affinities were calculated, as well as free energy and kinetics of molecular oxygen interaction with several non-enzymatically formed dimers. H-atom donating power of the intermediates increases upon the progress of the oxidation, making possible generation of hydroperoxyl radical or hydrogen peroxide from O2 at the last stages. Among the dimerization intermediates, 2-aminophenoxazinole derivatives have the lowest ionization potential and can reduce O2 to superoxide anion. The rate for O-H homolytic bond dissociation is significantly higher than that for C-H bond in non-enzymatic quinoneimine conjugate. However, the last reaction passes irreversibly, reducing O2 to hydroperoxyl radical. The inorganic ferrous iron and the heme group of Drosophila phenoxazinone synthase significantly reduce the energy cost of 2AP H-atom abstraction by O2. We have also shown experimentally that total antioxidant capacity decreases in Drosophila mutant cardinal with L-3HOK excess relative to the wild type Canton-S, and lipid peroxidation decreases in aged cardinal. Taken together, our data supports the conception of hydroxykynurenines' dual role in neurotoxicity: serving as antioxidants themselves, blocking lipid peroxidation by H-atom donation, they also can easily generate ROS upon dimerization, leading to the oxidative stress development.
Collapse
Affiliation(s)
- Aleksandr V. Zhuravlev
- Laboratory of Neurogenetics, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint-Petersburg, Russia
- * E-mail:
| | - Oleg V. Vetrovoy
- Laboratory of Regulation of the Brain Neuronal Functions, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint-Petersburg, Russia
- Department of Biochemistry, Faculty of Biology, Saint-Petersburg State University, Saint-Petersburg, Russia
| | - Elena V. Savvateeva-Popova
- Laboratory of Neurogenetics, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint-Petersburg, Russia
| |
Collapse
|
76
|
Sariyer E, Yakarsonmez S, Danis O, Turgut-Balik D. A study of Bos taurus muscle specific enolase; biochemical characterization, homology modelling and investigation of molecular interaction using molecular docking and dynamics simulations. Int J Biol Macromol 2018; 120:2346-2353. [DOI: 10.1016/j.ijbiomac.2018.08.184] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 08/28/2018] [Accepted: 08/30/2018] [Indexed: 01/09/2023]
|
77
|
Balasubramanian PK, Balupuri A, Bhujbal SP, Cho SJ. 3D-QSAR-aided design of potent c-Met inhibitors using molecular dynamics simulation and binding free energy calculation. J Biomol Struct Dyn 2018; 37:2165-2178. [PMID: 30044205 DOI: 10.1080/07391102.2018.1479309] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Mesenchymal-epithelial transition factor (c-Met) is a member of receptor tyrosine kinase. It involves in various cellular signaling pathways which includes proliferation, motility, migration, and invasion. Over-expression of c-Met has been reported in various cancers. Hence, it is an ideal therapeutic target for cancer. The main objective of the study is to identify crucial residues involved in the inhibition of c-Met kinase and to design a series of potent imidazo [4,5-b] pyrazine derivatives as c-Met inhibitors. Docking was used to identify important active site residues involved in the inhibition of c-Met kinase which was further validated by 100 ns of molecular dynamics simulation and free energy calculation using molecular mechanics generalized born surface area. Furthermore, binding energy decomposition identified that residues Tyr1230, Met1211, Asp1222, Tyr1159, Met1160, Val1092, Ala1108, and Leu1157 contributed favorably to the binding stability of compound 32. Receptor-guided Comparative Molecular Field Analysis (CoMFA) (q2 = 0.751, NOC = 6, r2 = 0.933) and Comparative Molecular Similarity Indices Analysis (COMSIA) (q2 = 0.744, NOC = 6, r2 = 0.950) models were generated based on the docked conformation of the most active compound 32. The robustness of these models was tested using various validation techniques and found to be predictive. The results of CoMFA and CoMSIA contour maps exposed the regions favorable to enhance the activity. Based on this information, 27 novel c-Met inhibitors were designed. These designed compounds exhibited potent activity than the most active compound of the existing dataset. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Pavithra K Balasubramanian
- a Department of Biomedical Sciences, College of Medicine , Chosun University , Gwangju 501-759 , Republic of Korea
| | - Anand Balupuri
- a Department of Biomedical Sciences, College of Medicine , Chosun University , Gwangju 501-759 , Republic of Korea
| | - Swapnil P Bhujbal
- a Department of Biomedical Sciences, College of Medicine , Chosun University , Gwangju 501-759 , Republic of Korea
| | - Seung Joo Cho
- a Department of Biomedical Sciences, College of Medicine , Chosun University , Gwangju 501-759 , Republic of Korea.,b Department of Cellular·Molecular Medicine, College of Medicine , Chosun University , Gwangju 501-759 , Republic of Korea
| |
Collapse
|
78
|
Jain S, Grandits M, Ecker GF. Interspecies comparison of putative ligand binding sites of human, rat and mouse P-glycoprotein. Eur J Pharm Sci 2018; 122:134-143. [PMID: 29936088 PMCID: PMC6422297 DOI: 10.1016/j.ejps.2018.06.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/18/2018] [Accepted: 06/19/2018] [Indexed: 01/16/2023]
Abstract
Prior to the clinical phases of testing, safety, efficacy and pharmacokinetic profiles of lead compounds are evaluated in animal studies. These tests are primarily performed in rodents, such as mouse and rats. In order to reduce the number of animal experiments, computational models that predict the outcome of these studies and thus aid in prioritization of preclinical candidates are heavily needed. However, although computational models for human off-target interactions with decent quality are available, they cannot easily be transferred to rodents due to lack of respective data. In this study, we assess the transferability of human P-glycoprotein activity data for development of in silico models to predict in vivo effects in rats and mouse using a structure-based approach. P-glycoprotein (P-gp) is an ATP-dependent efflux transporter that transports xenobiotic compounds such as toxins and drugs out of cells and has a broad substrate and inhibitor specificity. Being mostly expressed at barriers, it influences the bioavailability of drugs and thus contributes also to toxicity. Comparison of the binding site interaction profiles of human, rat and mouse P-gp derived from docking studies with a set of common inhibitors suggests that the inhibitors share potentially similar binding modes. These findings encourage the use of in vitro human P-gp data for predicting in vivo effects in rodents and thus contributes to the 3Rs (Replace, Reduce and Refine) of animal experiments.
Collapse
Affiliation(s)
- Sankalp Jain
- University of Vienna, Department of Pharmaceutical Chemistry, Althanstrasse 14, 1090 Vienna, Austria
| | - Melanie Grandits
- University of Vienna, Department of Pharmaceutical Chemistry, Althanstrasse 14, 1090 Vienna, Austria
| | - Gerhard F Ecker
- University of Vienna, Department of Pharmaceutical Chemistry, Althanstrasse 14, 1090 Vienna, Austria.
| |
Collapse
|
79
|
Alnoch RC, Stefanello AA, Paula Martini V, Richter JL, Mateo C, Souza EMD, Mitchell DA, Muller-Santos M, Krieger N. Co-expression, purification and characterization of the lipase and foldase of Burkholderia contaminans LTEB11. Int J Biol Macromol 2018; 116:1222-1231. [DOI: 10.1016/j.ijbiomac.2018.05.086] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 05/12/2018] [Accepted: 05/14/2018] [Indexed: 01/26/2023]
|
80
|
Comparative Studies of the Dynamics Effects of BAY60-2770 and BAY58-2667 Binding with Human and Bacterial H-NOX Domains. Molecules 2018; 23:molecules23092141. [PMID: 30149624 PMCID: PMC6225106 DOI: 10.3390/molecules23092141] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/11/2018] [Accepted: 08/22/2018] [Indexed: 11/16/2022] Open
Abstract
Soluble guanylate cyclase (sGC) is a key enzyme implicated in various physiological processes such as vasodilation, thrombosis and platelet aggregation. The enzyme’s Heme-Nitric oxide/Oxygen (H-NOX) binding domain is the only sensor of nitric oxide (NO) in humans, which on binding with NO activates sGC to produce the second messenger cGMP. H-NOX is thus a hot target for drug design programs. BAY60-2770 and BAY58-2667 are two widely studied activators of sGC. Here we present comparative molecular dynamics studies to understand the molecular details characterizing the binding of BAY60-2770 and BAY58-2667 with the human H-NOX (hH-NOX) and bacterial H-NOX (bH-NOX) domains. HartreeFock method was used for parametrization of both the activators. A 50 ns molecular dynamics (MD) simulation was run to identify the functionally critical regions of the H-NOX domains. The CPPTRAJ module was used for analysis. BAY60-2770 on binding with bH-NOX, triggered rotational movement in signaling helix F and significant dynamicity in loops α and β, but in hH-NOX domain the compound showed relatively lesser aforementioned structural fluctuations. Conversely, hH-NOX ligated BAY58-2667 experienced highest transitions in its helix F due to electrostatic interactions with D84, T85 and R88 residues which are not conserved in bH-NOX. These conformational transformations might be essential to communicate with downstream PAS, CC and cyclase domains of sGC. Comparative MD studies revealed that BAY bound bHNOX dynamics varied from that of hH-NOX, plausibly due to some key residues such as R40, F74 and Y112 which are not conserved in bacteria. These findings will help to the design of novel drug leads to cure diseases associated to human sGC.
Collapse
|
81
|
Anishchenko I, Kundrotas PJ, Vakser IA. Contact Potential for Structure Prediction of Proteins and Protein Complexes from Potts Model. Biophys J 2018; 115:809-821. [PMID: 30122295 DOI: 10.1016/j.bpj.2018.07.035] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 07/16/2018] [Accepted: 07/31/2018] [Indexed: 12/18/2022] Open
Abstract
The energy function is the key component of protein modeling methodology. This work presents a semianalytical approach to the development of contact potentials for protein structure modeling. Residue-residue and atom-atom contact energies were derived by maximizing the probability of observing native sequences in a nonredundant set of protein structures. The optimization task was formulated as an inverse statistical mechanics problem applied to the Potts model. Its solution by pseudolikelihood maximization provides consistent estimates of coupling constants at atomic and residue levels. The best performance was achieved when interacting atoms were grouped according to their physicochemical properties. For individual protein structures, the performance of the contact potentials in distinguishing near-native structures from the decoys is similar to the top-performing scoring functions. The potentials also yielded significant improvement in the protein docking success rates. The potentials recapitulated experimentally determined protein stability changes upon point mutations and protein-protein binding affinities. The approach offers a different perspective on knowledge-based potentials and may serve as the basis for their further development.
Collapse
Affiliation(s)
- Ivan Anishchenko
- Computational Biology Program and Department of Molecular Biosciences, The University of Kansas, Lawrence, Kansas
| | - Petras J Kundrotas
- Computational Biology Program and Department of Molecular Biosciences, The University of Kansas, Lawrence, Kansas.
| | - Ilya A Vakser
- Computational Biology Program and Department of Molecular Biosciences, The University of Kansas, Lawrence, Kansas.
| |
Collapse
|
82
|
Allosteric sodium binding cavity in GPR3: a novel player in modulation of Aβ production. Sci Rep 2018; 8:11102. [PMID: 30038319 PMCID: PMC6056553 DOI: 10.1038/s41598-018-29475-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 07/10/2018] [Indexed: 01/01/2023] Open
Abstract
The orphan G-protein coupled receptor 3 (GPR3) belongs to class A G-protein coupled receptors (GPCRs) and is highly expressed in central nervous system neurons. Among other functions, it is likely associated with neuron differentiation and maturation. Recently, GPR3 has also been linked to the production of Aβ peptides in neurons. Unfortunately, the lack of experimental structural information for this receptor hampers a deep characterization of its function. Here, using an in-silico and in-vitro combined approach, we describe, for the first time, structural characteristics of GPR3 receptor underlying its function: the agonist binding site and the allosteric sodium binding cavity. We identified and validated by alanine-scanning mutagenesis the role of three functionally relevant residues: Cys2676.55, Phe1203.36 and Asp2.50. The latter, when mutated into alanine, completely abolished the constitutive and agonist-stimulated adenylate cyclase activity of GPR3 receptor by disrupting its sodium binding cavity. Interestingly, this is correlated with a decrease in Aβ production in a model cell line. Taken together, these results suggest an important role of the allosteric sodium binding site for GPR3 activity and open a possible avenue for the modulation of Aβ production in the Alzheimer’s Disease.
Collapse
|
83
|
Rahman MZ, Tsujimori Y, Maeda M, Hossain MA, Ishimizu T, Kimura Y. Molecular characterization of second tomato α1,3/4-fucosidase (α-Fuc'ase Sl-2), a member of glycosyl hydrolase family 29 active toward the core α1,3-fucosyl residue in plant N-glycans. J Biochem 2018; 164:53-63. [PMID: 29444271 DOI: 10.1093/jb/mvy029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 02/04/2018] [Indexed: 01/07/2023] Open
Abstract
In a previous study, we molecular-characterized a tomato (Solanum lycopersicum) α1, 3/4-fucosidase (α-Fuc'ase Sl-1) encoded in a tomato gene (Solyc03g006980), indicating that α-Fuc'ase Sl-1 is involved in the turnover of Lea epitope-containing N-glycans. In this study, we have characterized another tomato gene (Solyc11g069010) encoding α1, 3/4-fucosidase (α-Fuc'ase Sl-2), which is also active toward the complex type N-glycans containing Lea epitope(s). The baculovirus-insect cell expression system was used to express that α-Fuc'ase Sl-2 with anti-FLAG tag, and the expression product (rFuc'ase Sl-2), was found as a 65 kDa protein using SDS-PAGE and has an optimum pH of around 5.0. Similarly to rFuc'ase Sl-1, rFuc'ase Sl-2 hydrolyzed the non-reducing terminal α1, 3-fucose residue on LNFP III and α1, 4-fucose residues of Lea epitopes on plant complex type N-glycans, but not the core α1, 3-fucose residue on Manβ1-4GlcNAcβ1-4(Fucα1-3)GlcNAc or Fucα1-3GlcNAc. However, we found that both α-Fuc'ases Sl-1 and Sl-2 were specifically active toward α1, 3-fucose residue on GlcNAcβ1-4(Fucα1-3)GlcNAc, indicating that the non-substituted β-GlcNAc linked to the proximal GlcNAc residue of the core tri-saccharide moiety of plant specific N-glycans must be a pre-requisite for α-Fuc'ase activity. A 3 D modelled structure of the catalytic sites of α-Fuc'ase Sl-2 suggested that Asp192 and Glu236 may be important for binding to the α1, 3/4 fucose residue.
Collapse
Affiliation(s)
- Md Ziaur Rahman
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan.,Institute of Food and Radiation Biology, Atomic Energy Research Establishment, Bangladesh Atomic Energy Commission, Ganakbari, Savar, Dhaka 1340, Bangladesh
| | - Yuta Tsujimori
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan
| | - Megumi Maeda
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan
| | - Md Anowar Hossain
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Takeshi Ishimizu
- College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan
| | - Yoshinobu Kimura
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan
| |
Collapse
|
84
|
Schönherz AA, Forsberg R, Guldbrandtsen B, Buitenhuis AJ, Einer-Jensen K. Introduction of Viral Hemorrhagic Septicemia Virus into Freshwater Cultured Rainbow Trout Is Followed by Bursts of Adaptive Evolution. J Virol 2018; 92:e00436-18. [PMID: 29643236 PMCID: PMC5974487 DOI: 10.1128/jvi.00436-18] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 03/20/2018] [Indexed: 12/25/2022] Open
Abstract
Viral hemorrhagic septicemia virus (VHSV), a rhabdovirus infecting teleost fish, has repeatedly crossed the boundary from marine fish species to freshwater cultured rainbow trout. These naturally replicated cross-species transmission events permit the study of general and repeatable evolutionary events occurring in connection with viral emergence in a novel host species. The purpose of the present study was to investigate the adaptive molecular evolution of the VHSV glycoprotein, one of the key virus proteins involved in viral emergence, following emergence from marine species into freshwater cultured rainbow trout. A comprehensive phylogenetic reconstruction of the complete coding region of the VHSV glycoprotein was conducted, and adaptive molecular evolution was investigated using a maximum likelihood approach to compare different codon substitution models allowing for heterogeneous substitution rate ratios among amino acid sites. Evidence of positive selection was detected at six amino acid sites of the VHSV glycoprotein, within the signal peptide, the confirmation-dependent major neutralizing epitope, and the intracellular tail. Evidence of positive selection was found exclusively in rainbow trout-adapted virus isolates, and amino acid combinations found at the six sites under positive selection pressure differentiated rainbow trout- from non-rainbow trout-adapted isolates. Furthermore, four adaptive sites revealed signs of recurring identical changes across phylogenetic groups of rainbow trout-adapted isolates, suggesting that repeated VHSV emergence in freshwater cultured rainbow trout was established through convergent routes of evolution that are associated with immune escape.IMPORTANCE This study is the first to demonstrate that VHSV emergence from marine species into freshwater cultured rainbow trout has been accompanied by bursts of adaptive evolution in the VHSV glycoprotein. Furthermore, repeated detection of the same adaptive amino acid sites across phylogenetic groups of rainbow trout-adapted isolates indicates that adaptation to rainbow trout was established through parallel evolution. In addition, signals of convergent evolution toward the maintenance of genetic variation were detected in the conformation-dependent neutralizing epitope or in close proximity to disulfide bonds involved in the structural conformation of the neutralizing epitope, indicating adaptation to immune response-related genetic variation across freshwater cultured rainbow trout.
Collapse
Affiliation(s)
- Anna A Schönherz
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | | - Bernt Guldbrandtsen
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Albert J Buitenhuis
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | |
Collapse
|
85
|
Dietrich R, Alberca L, Ruiz M, Palestro P, Carrillo C, Talevi A, Gavernet L. Identification of cisapride as new inhibitor of putrescine uptake in Trypanosoma cruzi by combined ligand- and structure-based virtual screening. Eur J Med Chem 2018; 149:22-29. [DOI: 10.1016/j.ejmech.2018.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/30/2018] [Accepted: 02/03/2018] [Indexed: 10/18/2022]
|
86
|
Clémençon B, Lüscher BP, Hediger MA. Establishment of a novel microscale thermophoresis ligand-binding assay for characterization of SLC solute carriers using oligopeptide transporter PepT1 (SLC15 family) as a model system. J Pharmacol Toxicol Methods 2018; 92:67-76. [PMID: 29580877 DOI: 10.1016/j.vascn.2018.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 03/05/2018] [Accepted: 03/14/2018] [Indexed: 10/17/2022]
Abstract
INTRODUCTION Membrane proteins represent roughly one third of the human proteome and many of them serve as targets of therapeutic drugs. An exception is the SLC solute carrier superfamily with only a handful of approved drugs targeting SLCs. Indeed, for many of the SLCs, the natural transport substrates are still unknown. A major limitation for SLCs has been the difficulty to thoroughly characterize these multimembrane spanning proteins. The intrinsic properties of membrane proteins with alternative hydrophobic and hydrophilic domains lead to instability, making the purification tasks even more challenging compared to soluble proteins. This issue also holds true for conventional ligand-binding assays (LBAs) which usually require high-quality, pure and concentrated protein samples. Herein, we report a novel binding assay strategy to overcome these issues, taking advantage of a unique combination of yeast expression and microscale thermophoresis (MST). Following yeast overexpression of SLC15A1/PepT1 ortholog from moss Physcomitrella patens, PepTPp, which exhibits remarkable similarity to human PepT1, the approach was validated using dipeptide glycylsarcosine (Gly-Sar) and antiviral prodrug valacyclovir as test substrates. METHOD The originality of our approach is based on the comparative analysis of solubilized total membrane preparations with or without expression of the SLC target of interest, using a yeast strain (S. cerevisiae), in which the corresponding endogenous SLC homolog is depleted. MST is a recently developed technique that takes advantage of the properties of biomolecules in solution to migrate along a temperature gradient. Importantly, this migration is affected by substrate binding. It is being monitored by fluorescence using labelled SLC molecules in the presence of different ligand concentrations. RESULTS We herein report a novel MST/yeast-based method to characterize binding of ligands to SLCs without the need for a prior SLC-purification step. For validation purposes, we used a close eukaryotic homolog of the human H+-coupled oligopeptide transporter PepT1 (SLC15A1) that mediates uptake of di-tripeptides and peptide-like drugs as a test model. This approach allowed the successful confirmation of the binding of Gly-Sar at the mM range and revealed for the first time the KD of the antiviral prodrug valacyclovir to the PepT1 homolog at around 50 μM. DISCUSSION This novel LBA approach is independent of protein purification. It is suitable for drug discovery as it is upscalable to high throughput compound screening. It works well for SLC transporters which are underrepresented targets due to their difficulties to study them. Moreover, this approach could make a significant contribution toward "deorphanization" of SLCs, revealing their transport substrates.
Collapse
Affiliation(s)
- Benjamin Clémençon
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland.
| | - Benjamin P Lüscher
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Matthias A Hediger
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland.
| |
Collapse
|
87
|
Vedithi SC, Malhotra S, Das M, Daniel S, Kishore N, George A, Arumugam S, Rajan L, Ebenezer M, Ascher DB, Arnold E, Blundell TL. Structural Implications of Mutations Conferring Rifampin Resistance in Mycobacterium leprae. Sci Rep 2018; 8:5016. [PMID: 29567948 PMCID: PMC5864748 DOI: 10.1038/s41598-018-23423-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/09/2018] [Indexed: 11/09/2022] Open
Abstract
The rpoB gene encodes the β subunit of RNA polymerase holoenzyme in Mycobacterium leprae (M. leprae). Missense mutations in the rpoB gene were identified as etiological factors for rifampin resistance in leprosy. In the present study, we identified mutations corresponding to rifampin resistance in relapsed leprosy cases from three hospitals in southern India which treat leprosy patients. DNA was extracted from skin biopsies of 35 relapse/multidrug therapy non-respondent leprosy cases, and PCR was performed to amplify the 276 bp rifampin resistance-determining region of the rpoB gene. PCR products were sequenced, and mutations were identified in four out of the 35 cases at codon positions D441Y, D441V, S437L and H476R. The structural and functional effects of these mutations were assessed in the context of three-dimensional comparative models of wild-type and mutant M. leprae RNA polymerase holoenzyme (RNAP), based on the recently solved crystal structures of RNAP of Mycobacterium tuberculosis, containing a synthetic nucleic acid scaffold and rifampin. The resistance mutations were observed to alter the hydrogen-bonding and hydrophobic interactions of rifampin and the 5' ribonucleotide of the growing RNA transcript. This study demonstrates that rifampin-resistant strains of M. leprae among leprosy patients in southern India are likely to arise from mutations that affect the drug-binding site and stability of RNAP.
Collapse
Affiliation(s)
- Sundeep Chaitanya Vedithi
- Department of Biochemistry, University of Cambridge, Tennis Court Rd., Cambridge, CB2 1GA, UK. .,Molecular Biology and Immunology Division, Schieffelin Institute of Health Research & Leprosy Center (SIH R & LC), Karigiri, Vellore, Tamil Nadu, 632106, India.
| | - Sony Malhotra
- Department of Biochemistry, University of Cambridge, Tennis Court Rd., Cambridge, CB2 1GA, UK
| | - Madhusmita Das
- Molecular Biology and Immunology Division, Schieffelin Institute of Health Research & Leprosy Center (SIH R & LC), Karigiri, Vellore, Tamil Nadu, 632106, India
| | - Sheela Daniel
- Molecular Biology and Immunology Division, Schieffelin Institute of Health Research & Leprosy Center (SIH R & LC), Karigiri, Vellore, Tamil Nadu, 632106, India
| | - Nanda Kishore
- Department of Dermatology, Father Muller Medical College & Hospital, Mangalore, Karnataka, 575 002, India
| | - Anuja George
- Department of Dermatology, Trivandrum Medical College, Trivandrum, Kerala, 695011, India
| | - Shantha Arumugam
- Molecular Biology and Immunology Division, Schieffelin Institute of Health Research & Leprosy Center (SIH R & LC), Karigiri, Vellore, Tamil Nadu, 632106, India
| | - Lakshmi Rajan
- Molecular Biology and Immunology Division, Schieffelin Institute of Health Research & Leprosy Center (SIH R & LC), Karigiri, Vellore, Tamil Nadu, 632106, India
| | - Mannam Ebenezer
- Molecular Biology and Immunology Division, Schieffelin Institute of Health Research & Leprosy Center (SIH R & LC), Karigiri, Vellore, Tamil Nadu, 632106, India
| | - David B Ascher
- Department of Biochemistry and Molecular Biology, Bio21 Institute, University of Melbourne, Parkville, VIC 3052, Australia
| | - Eddy Arnold
- Center for Advanced Biotechnology and Medicine (CABM), and Rutgers University Department of Chemistry and Chemical Biology, 679 Hoes Lane, Piscataway, NJ, 08854, USA
| | - Tom L Blundell
- Department of Biochemistry, University of Cambridge, Tennis Court Rd., Cambridge, CB2 1GA, UK.
| |
Collapse
|
88
|
Hassan SS, Jamal SB, Radusky LG, Tiwari S, Ullah A, Ali J, Behramand, de Carvalho PVSD, Shams R, Khan S, Figueiredo HCP, Barh D, Ghosh P, Silva A, Baumbach J, Röttger R, Turjanski AG, Azevedo VAC. The Druggable Pocketome of Corynebacterium diphtheriae: A New Approach for in silico Putative Druggable Targets. Front Genet 2018; 9:44. [PMID: 29487617 PMCID: PMC5816920 DOI: 10.3389/fgene.2018.00044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 01/30/2018] [Indexed: 01/20/2023] Open
Abstract
Diphtheria is an acute and highly infectious disease, previously regarded as endemic in nature but vaccine-preventable, is caused by Corynebacterium diphtheriae (Cd). In this work, we used an in silico approach along the 13 complete genome sequences of C. diphtheriae followed by a computational assessment of structural information of the binding sites to characterize the “pocketome druggability.” To this end, we first computed the “modelome” (3D structures of a complete genome) of a randomly selected reference strain Cd NCTC13129; that had 13,763 open reading frames (ORFs) and resulted in 1,253 (∼9%) structure models. The amino acid sequences of these modeled structures were compared with the remaining 12 genomes and consequently, 438 conserved protein sequences were obtained. The RCSB-PDB database was consulted to check the template structures for these conserved proteins and as a result, 401 adequate 3D models were obtained. We subsequently predicted the protein pockets for the obtained set of models and kept only the conserved pockets that had highly druggable (HD) values (137 across all strains). Later, an off-target host homology analyses was performed considering the human proteome using NCBI database. Furthermore, the gene essentiality analysis was carried out that gave a final set of 10-conserved targets possessing highly druggable protein pockets. To check the target identification robustness of the pipeline used in this work, we crosschecked the final target list with another in-house target identification approach for C. diphtheriae thereby obtaining three common targets, these were; hisE-phosphoribosyl-ATP pyrophosphatase, glpX-fructose 1,6-bisphosphatase II, and rpsH-30S ribosomal protein S8. Our predicted results suggest that the in silico approach used could potentially aid in experimental polypharmacological target determination in C. diphtheriae and other pathogens, thereby, might complement the existing and new drug-discovery pipelines.
Collapse
Affiliation(s)
- Syed S Hassan
- Department of Chemistry, Islamia College University Peshawar, Peshawar, Pakistan
| | - Syed B Jamal
- PG Program in Bioinformatics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Leandro G Radusky
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Sandeep Tiwari
- PG Program in Bioinformatics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Asad Ullah
- Department of Chemistry, Islamia College University Peshawar, Peshawar, Pakistan
| | - Javed Ali
- Department of Chemistry, Kohat University of Science and Technology, Kohat, Pakistan
| | - Behramand
- Department of Chemistry, Islamia College University Peshawar, Peshawar, Pakistan
| | - Paulo V S D de Carvalho
- PG Program in Bioinformatics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Rida Shams
- Department of Chemistry, Islamia College University Peshawar, Peshawar, Pakistan
| | - Sabir Khan
- Department of Analytical Chemistry, Institute of Chemistry, São Paulo State University, São Paulo, Brazil
| | - Henrique C P Figueiredo
- AQUACEN, National Reference Laboratory for Aquatic Animal Diseases, Ministry of Fisheries and Aquaculture, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Debmalya Barh
- PG Program in Bioinformatics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil.,Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology, Purba Medinipur, India
| | - Preetam Ghosh
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA, United States
| | - Artur Silva
- Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Jan Baumbach
- Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
| | - Richard Röttger
- Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
| | - Adrián G Turjanski
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.,INQUIMAE/UBA-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Vasco A C Azevedo
- PG Program in Bioinformatics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
89
|
Abstract
Inteins are intervening proteins that undergo an autocatalytic splicing reaction that ligates flanking host protein sequences termed exteins. Some intein-containing proteins have evolved to couple splicing to environmental signals; this represents a new form of posttranslational regulation. Of particular interest is RadA from the archaeon Pyrococcus horikoshii, for which long-range intein-extein interactions block splicing, requiring temperature and single-stranded DNA (ssDNA) substrate to splice rapidly and accurately. Here, we report that splicing of the intein-containing RadA from another archaeon, Thermococcus sibericus, is activated by significantly lower temperatures than is P. horikoshii RadA, consistent with differences in their growth environments. Investigation into variations between T. sibericus and P. horikoshii RadA inteins led to the discovery that a nonconserved region (NCR) of the intein, a flexible loop where a homing endonuclease previously resided, is critical to splicing. Deletion of the NCR leads to a substantial loss in the rate and accuracy of P. horikoshii RadA splicing only within native exteins. The influence of the NCR deletion can be largely overcome by ssDNA, demonstrating that the splicing-competent conformation can be achieved. We present a model whereby the NCR is a flexible hinge which acts as a switch by controlling distant intein-extein interactions that inhibit active site assembly. These results speak to the repurposing of the vestigial endonuclease loop to control an intein-extein partnership, which ultimately allows exquisite adaptation of protein splicing upon changes in the environment. Inteins are mobile genetic elements that interrupt coding sequences (exteins) and are removed by protein splicing. They are abundant elements in microbes, and recent work has demonstrated that protein splicing can be controlled by environmental cues, including the substrate of the intein-containing protein. Here, we describe an intein-extein collaboration that controls temperature-induced splicing of RadA from two archaea and how variation in this intein-extein partnership results in fine-tuning of splicing to closely match the environment. Specifically, we found that a small sequence difference between the two inteins, a flexible loop that likely once housed a homing endonuclease used for intein mobility, acts as a switch to control intein-extein interactions that block splicing. Our results argue strongly that some inteins have evolved away from a purely parasitic lifestyle to control the activity of host proteins, representing a new form of posttranslational regulation that is potentially widespread in the microbial world.
Collapse
|
90
|
Hirata T, Mishra SK, Nakamura S, Saito K, Motooka D, Takada Y, Kanzawa N, Murakami Y, Maeda Y, Fujita M, Yamaguchi Y, Kinoshita T. Identification of a Golgi GPI-N-acetylgalactosamine transferase with tandem transmembrane regions in the catalytic domain. Nat Commun 2018; 9:405. [PMID: 29374258 PMCID: PMC5785973 DOI: 10.1038/s41467-017-02799-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 12/28/2017] [Indexed: 12/31/2022] Open
Abstract
Many eukaryotic proteins are anchored to the cell surface via the glycolipid glycosylphosphatidylinositol (GPI). Mammalian GPIs have a conserved core but exhibit diverse N-acetylgalactosamine (GalNAc) modifications, which are added via a yet unresolved process. Here we identify the Golgi-resident GPI-GalNAc transferase PGAP4 and show by mass spectrometry that PGAP4 knockout cells lose GPI-GalNAc structures. Furthermore, we demonstrate that PGAP4, in contrast to known Golgi glycosyltransferases, is not a single-pass membrane protein but contains three transmembrane domains, including a tandem transmembrane domain insertion into its glycosyltransferase-A fold as indicated by comparative modeling. Mutational analysis reveals a catalytic site, a DXD-like motif for UDP-GalNAc donor binding, and several residues potentially involved in acceptor binding. We suggest that a juxtamembrane region of PGAP4 accommodates various GPI-anchored proteins, presenting their acceptor residue toward the catalytic center. In summary, we present insights into the structure of PGAP4 and elucidate the initial step of GPI-GalNAc biosynthesis.
Collapse
Affiliation(s)
- Tetsuya Hirata
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
- WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
- National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi, 444-8787, Japan
| | - Sushil K Mishra
- Structural Glycobiology Team, Systems Glycobiology Research Group, RIKEN Global Research Cluster, Wako, Saitama, 351-0198, Japan
| | - Shota Nakamura
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Kazunobu Saito
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Daisuke Motooka
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Yoko Takada
- WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Noriyuki Kanzawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
- WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Yoshiko Murakami
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
- WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Yusuke Maeda
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
- WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Morihisa Fujita
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Yoshiki Yamaguchi
- Structural Glycobiology Team, Systems Glycobiology Research Group, RIKEN Global Research Cluster, Wako, Saitama, 351-0198, Japan
| | - Taroh Kinoshita
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan.
- WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
91
|
Fierro F, Suku E, Alfonso-Prieto M, Giorgetti A, Cichon S, Carloni P. Agonist Binding to Chemosensory Receptors: A Systematic Bioinformatics Analysis. Front Mol Biosci 2017; 4:63. [PMID: 28932739 PMCID: PMC5592726 DOI: 10.3389/fmolb.2017.00063] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/22/2017] [Indexed: 12/17/2022] Open
Abstract
Human G-protein coupled receptors (hGPCRs) constitute a large and highly pharmaceutically relevant membrane receptor superfamily. About half of the hGPCRs' family members are chemosensory receptors, involved in bitter taste and olfaction, along with a variety of other physiological processes. Hence these receptors constitute promising targets for pharmaceutical intervention. Molecular modeling has been so far the most important tool to get insights on agonist binding and receptor activation. Here we investigate both aspects by bioinformatics-based predictions across all bitter taste and odorant receptors for which site-directed mutagenesis data are available. First, we observe that state-of-the-art homology modeling combined with previously used docking procedures turned out to reproduce only a limited fraction of ligand/receptor interactions inferred by experiments. This is most probably caused by the low sequence identity with available structural templates, which limits the accuracy of the protein model and in particular of the side-chains' orientations. Methods which transcend the limited sampling of the conformational space of docking may improve the predictions. As an example corroborating this, we review here multi-scale simulations from our lab and show that, for the three complexes studied so far, they significantly enhance the predictive power of the computational approach. Second, our bioinformatics analysis provides support to previous claims that several residues, including those at positions 1.50, 2.50, and 7.52, are involved in receptor activation.
Collapse
Affiliation(s)
- Fabrizio Fierro
- Computational Biomedicine, Institute for Advanced Simulation IAS-5 and Institute of Neuroscience and Medicine INM-9, Forschungszentrum JülichJülich, Germany
| | - Eda Suku
- Department of Biotechnology, University of VeronaVerona, Italy
| | - Mercedes Alfonso-Prieto
- Computational Biomedicine, Institute for Advanced Simulation IAS-5 and Institute of Neuroscience and Medicine INM-9, Forschungszentrum JülichJülich, Germany.,Cécile and Oskar Vogt Institute for Brain Research, Medical Faculty, Heinrich Heine University DüsseldorfDüsseldorf, Germany
| | - Alejandro Giorgetti
- Computational Biomedicine, Institute for Advanced Simulation IAS-5 and Institute of Neuroscience and Medicine INM-9, Forschungszentrum JülichJülich, Germany.,Department of Biotechnology, University of VeronaVerona, Italy
| | - Sven Cichon
- Institute of Neuroscience and Medicine INM-1, Forschungszentrum JülichJülich, Germany.,Institute for Human Genetics, Department of Genomics, Life&Brain Center, University of BonnBonn, Germany.,Division of Medical Genetics, Department of Biomedicine, University of BaselBasel, Switzerland
| | - Paolo Carloni
- Computational Biomedicine, Institute for Advanced Simulation IAS-5 and Institute of Neuroscience and Medicine INM-9, Forschungszentrum JülichJülich, Germany.,Department of Physics, Rheinisch-Westfälische Technische Hochschule AachenAachen, Germany.,VNU Key Laboratory "Multiscale Simulation of Complex Systems", VNU University of Science, Vietnam National UniversityHanoi, Vietnam
| |
Collapse
|
92
|
V M V, Dubey VK, Ponnuraj K. Identification of two natural compound inhibitors of Leishmania donovani Spermidine Synthase (SpdS) through molecular docking and dynamic studies. J Biomol Struct Dyn 2017; 36:2678-2693. [PMID: 28797195 DOI: 10.1080/07391102.2017.1366947] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Visceral leishmaniasis caused by the protozoan Leishmania donovani is the most severe form of leishmaniasis and it is potentially lethal if untreated. Despite the availability of drugs for treating the disease, the current drug regime suffers from drawbacks like antibiotic resistance and toxicity. New drugs have to be discovered in order to overcome these limitations. Our aim is to identify natural compounds from plant sources as putative inhibitors considering the occurrence of structural diversity in plant sources. Spermidine Synthase (SpdS) was chosen as the target enzyme as it plays a vital role in growth, survival, and due to its contribution in virulence. Our initial investigation started with a literature survey in identifying natural compounds that showed antileishmanial activity. Subsequently, we identified two monoterpenoid compounds, namely Geraniol and Linalool, that were structurally analogous to one of the substrates (putrescine) of SpdS. In the present study, homology model of L. donovani SpdS was generated and the binding affinity of the identified compounds was analyzed and also compared with the putrescine through molecular docking and dynamic studies. The pharmacokinetic properties of the identified compounds were validated and the binding efficiency of these ligands over the original substrate has been demonstrated. Based on these studies, Geraniol and Linalool can be considered as lead molecules for future investigations targeting SpdS. This study further emphasizes the choice of natural compounds as a good source of therapeutic agents.
Collapse
Affiliation(s)
- Vidhya V M
- a Centre of Advanced Study in Crystallography and Biophysics , University of Madras , Guindy Campus, Chennai - 600 025 , India
| | - Vikash Kumar Dubey
- b Department of Biosciences and Bioengineering , Indian Institute of Technology Guwahati , Guwahati - 781039 , India
| | - Karthe Ponnuraj
- a Centre of Advanced Study in Crystallography and Biophysics , University of Madras , Guindy Campus, Chennai - 600 025 , India
| |
Collapse
|
93
|
Lam SD, Das S, Sillitoe I, Orengo C. An overview of comparative modelling and resources dedicated to large-scale modelling of genome sequences. Acta Crystallogr D Struct Biol 2017; 73:628-640. [PMID: 28777078 PMCID: PMC5571743 DOI: 10.1107/s2059798317008920] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 06/14/2017] [Indexed: 12/02/2022] Open
Abstract
Computational modelling of proteins has been a major catalyst in structural biology. Bioinformatics groups have exploited the repositories of known structures to predict high-quality structural models with high efficiency at low cost. This article provides an overview of comparative modelling, reviews recent developments and describes resources dedicated to large-scale comparative modelling of genome sequences. The value of subclustering protein domain superfamilies to guide the template-selection process is investigated. Some recent cases in which structural modelling has aided experimental work to determine very large macromolecular complexes are also cited.
Collapse
Affiliation(s)
- Su Datt Lam
- Institute of Structural and Molecular Biology, UCL, Darwin Building, Gower Street, London WC1E 6BT, England
- School of Biosciences and Biotechnology, Faculty of Science and Technology, University Kebangsaan Malaysia, 43600 Bangi, Selangor, Malaysia
| | - Sayoni Das
- Institute of Structural and Molecular Biology, UCL, Darwin Building, Gower Street, London WC1E 6BT, England
| | - Ian Sillitoe
- Institute of Structural and Molecular Biology, UCL, Darwin Building, Gower Street, London WC1E 6BT, England
| | - Christine Orengo
- Institute of Structural and Molecular Biology, UCL, Darwin Building, Gower Street, London WC1E 6BT, England
| |
Collapse
|
94
|
Jain S, Grandits M, Richter L, Ecker GF. Structure based classification for bile salt export pump (BSEP) inhibitors using comparative structural modeling of human BSEP. J Comput Aided Mol Des 2017; 31:507-521. [PMID: 28527154 PMCID: PMC5487762 DOI: 10.1007/s10822-017-0021-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 04/30/2017] [Indexed: 01/13/2023]
Abstract
The bile salt export pump (BSEP) actively transports conjugated monovalent bile acids from the hepatocytes into the bile. This facilitates the formation of micelles and promotes digestion and absorption of dietary fat. Inhibition of BSEP leads to decreased bile flow and accumulation of cytotoxic bile salts in the liver. A number of compounds have been identified to interact with BSEP, which results in drug-induced cholestasis or liver injury. Therefore, in silico approaches for flagging compounds as potential BSEP inhibitors would be of high value in the early stage of the drug discovery pipeline. Up to now, due to the lack of a high-resolution X-ray structure of BSEP, in silico based identification of BSEP inhibitors focused on ligand-based approaches. In this study, we provide a homology model for BSEP, developed using the corrected mouse P-glycoprotein structure (PDB ID: 4M1M). Subsequently, the model was used for docking-based classification of a set of 1212 compounds (405 BSEP inhibitors, 807 non-inhibitors). Using the scoring function ChemScore, a prediction accuracy of 81% on the training set and 73% on two external test sets could be obtained. In addition, the applicability domain of the models was assessed based on Euclidean distance. Further, analysis of the protein-ligand interaction fingerprints revealed certain functional group-amino acid residue interactions that could play a key role for ligand binding. Though ligand-based models, due to their high speed and accuracy, remain the method of choice for classification of BSEP inhibitors, structure-assisted docking models demonstrate reasonably good prediction accuracies while additionally providing information about putative protein-ligand interactions.
Collapse
Affiliation(s)
- Sankalp Jain
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria
| | - Melanie Grandits
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria
| | - Lars Richter
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria
| | - Gerhard F Ecker
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria.
| |
Collapse
|
95
|
Johnson RM, Bai G, DeMott CM, Banavali NK, Montague CR, Moon C, Shekhtman A, VanderVen B, McDonough KA. Chemical activation of adenylyl cyclase Rv1625c inhibits growth of Mycobacterium tuberculosis on cholesterol and modulates intramacrophage signaling. Mol Microbiol 2017; 105:294-308. [PMID: 28464471 DOI: 10.1111/mmi.13701] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/24/2017] [Accepted: 04/25/2017] [Indexed: 12/31/2022]
Abstract
Mycobacterium tuberculosis (Mtb) uses a complex 3', 5'-cyclic AMP (cAMP) signaling network to sense and respond to changing environments encountered during infection, so perturbation of cAMP signaling might be leveraged to disrupt Mtb pathogenesis. However, understanding of cAMP signaling pathways is hindered by the presence of at least 15 distinct adenylyl cyclases (ACs). Recently, the small molecule V-58 was shown to inhibit Mtb replication within macrophages and stimulate cAMP production in Mtb. Here we determined that V-58 rapidly and directly activates Mtb AC Rv1625c to produce high levels of cAMP regardless of the bacterial environment or growth medium. Metabolic inhibition by V-58 was carbon source dependent in Mtb and did not occur in Mycobacterium smegmatis, suggesting that V-58-mediated growth inhibition is due to interference with specific Mtb metabolic pathways rather than a generalized cAMP toxicity. Chemical stimulation of cAMP production by Mtb within macrophages also caused down regulation of TNF-α production by the macrophages, indicating a complex role for cAMP in Mtb pathogenesis. Together these studies describe a novel approach for targeted stimulation of cAMP production in Mtb, and provide new insights into the myriad roles of cAMP signaling in Mtb, particularly during Mtb's interactions with macrophages.
Collapse
Affiliation(s)
- Richard M Johnson
- Department of Biomedical Sciences, School of Public Health, University at Albany, SUNY, Albany, NY, USA
| | - Guangchun Bai
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | | | - Nilesh K Banavali
- Department of Biomedical Sciences, School of Public Health, University at Albany, SUNY, Albany, NY, USA.,New York State Department of Health, Wadsworth Center, Albany, NY, USA
| | | | - Caroline Moon
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | | | - Brian VanderVen
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Kathleen A McDonough
- Department of Biomedical Sciences, School of Public Health, University at Albany, SUNY, Albany, NY, USA.,New York State Department of Health, Wadsworth Center, Albany, NY, USA
| |
Collapse
|
96
|
Balasubramanian PK, Balupuri A, Kang HY, Cho SJ. Receptor-guided 3D-QSAR studies, molecular dynamics simulation and free energy calculations of Btk kinase inhibitors. BMC SYSTEMS BIOLOGY 2017; 11:6. [PMID: 28361711 PMCID: PMC5374705 DOI: 10.1186/s12918-017-0385-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
BACKGROUND Bruton tyrosine kinase (Btk) plays an important role in B-cell development, differentiation, and signaling. It is also found be in involved in male immunodeficiency disease such as X-linked agammaglobulinemia (XLA). Btk is considered as a potential therapeutic target for treating autoimmune diseases and hematological malignancies. RESULTS In this work, a combined molecular modeling study was performed on a series of thieno [3,2-c] pyridine-4-amine derivatives as Btk inhibitors. Receptor-guided COMFA (q 2 = 0.574, NOC = 3, r 2 = 0.924) and COMSIA (q 2 = 0.646, NOC = 6, r 2 = 0.971) models were generated based on the docked conformation of the most active compound 26. All the developed models were tested for robustness using various validation techniques. Furthermore, a 5-ns molecular dynamics (MD) simulation and binding free energy calculations were carried out to determine the binding modes of the inhibitors and to identify crucial interacting residues. The rationality and stability of molecular docking and 3D-QSAR results were validated by MD simulation. The binding free energies calculated by the MM/PBSA method showed the importance of the van der Waals interaction. CONCLUSIONS A good correlation between the MD results, docking studies, and the contour map analysis were observed. The study has identified the key amino acid residues in Btk binding pocket. The results from this study can provide some insights into the development of potent, novel Btk inhibitors.
Collapse
Affiliation(s)
- Pavithra K Balasubramanian
- Department of Biomedical Sciences, College of Medicine, Chosun University, 375 Seosuk-dong, Dong-gu, Gwangju, 61452, Republic of Korea
| | - Anand Balupuri
- Department of Biomedical Sciences, College of Medicine, Chosun University, 375 Seosuk-dong, Dong-gu, Gwangju, 61452, Republic of Korea
| | - Hee-Young Kang
- Department of Nursing, Chosun University, Gwangju, 61452, Republic of Korea
| | - Seung Joo Cho
- Department of Biomedical Sciences, College of Medicine, Chosun University, 375 Seosuk-dong, Dong-gu, Gwangju, 61452, Republic of Korea.
- Department of Cellular Molecular Medicine, College of Medicine, Chosun University, Gwangju, 61452, Republic of Korea.
| |
Collapse
|
97
|
Gangopadhyay A, Chakraborty HJ, Datta A. Targeting the dengue β-OG with serotype-specific alkaloid virtual leads. J Mol Graph Model 2017; 73:129-142. [PMID: 28279821 DOI: 10.1016/j.jmgm.2017.02.018] [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: 07/24/2016] [Revised: 01/24/2017] [Accepted: 02/27/2017] [Indexed: 12/31/2022]
Abstract
The dengue envelope β-OG pocket is a crucial hinge for mediating virus-host fusion via conformational changes in the envelope to the fusion-competent form. The β-OG pocket is a small molecule target site for inhibition of virus-host fusion. As of date, the only structure of the β-OG pocket known is of serotype 2. Studies of β-OG inhibition by small molecules primarily target viral serotype 2. Envelope and β-OG sequence alignments, reveal dissimilarities across serotypes. In light of protein sequence-structure-function correlation, sequence variations suggest serotypic variations in β-OG druggability. This, together with the fact that dengue viral proteins do have serotype-specific variations of structure and function, lead to the study of the serotype-specificity of the dengue β-OG ligand binding behaviour. β-OG druggability was compared using comparative models of envelope proteins containing the β-OG pocket in four serotypes of the dengue virus. β-OG ligand binding was found to vary with respect to hydrophobicity, hydrophilicity, hydrogen bonding, van der Waals interactions with ligands and tightness of the binding site. The study also reports serotype-specific virtual leads identified from a library of 9175 alkaloids, using a consensus docking and scoring approach. The docking algorithms of Glide SP and XP, together with the Lamarckian genetic algorithm were employed for consensus docking. For consensus scoring, the Glide empirical score was employed along with the scoring function of AutoDock. A multi-dimensional lead optimisation approach was performed for optimising affinity, ligand efficiency, lipophilic ligand efficiency, ADMET and molecular torsional strains. The study proposes the serotype-specific inhibition of the β-OG for an effective inhibition of virus-host fusion, in contrast to a pan inhibitor.
Collapse
Affiliation(s)
| | - Hirak Jyoti Chakraborty
- Central Inland Fisheries Research Institute, Barrackpore, Kolkata 700120, West Bengal, India.
| | - Abhijit Datta
- Jhargram Raj College, Jhargram 721507, West Bengal, India.
| |
Collapse
|
98
|
Sanders MA, Zhang H, Mladenovic L, Tseng YY, Granneman JG. Molecular Basis of ABHD5 Lipolysis Activation. Sci Rep 2017; 7:42589. [PMID: 28211464 PMCID: PMC5314347 DOI: 10.1038/srep42589] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/11/2017] [Indexed: 12/21/2022] Open
Abstract
Alpha-beta hydrolase domain-containing 5 (ABHD5), the defective gene in human Chanarin-Dorfman syndrome, is a highly conserved regulator of adipose triglyceride lipase (ATGL)-mediated lipolysis that plays important roles in metabolism, tumor progression, viral replication, and skin barrier formation. The structural determinants of ABHD5 lipolysis activation, however, are unknown. We performed comparative evolutionary analysis and structural modeling of ABHD5 and ABHD4, a functionally distinct paralog that diverged from ABHD5 ~500 million years ago, to identify determinants of ABHD5 lipolysis activation. Two highly conserved ABHD5 amino acids (R299 and G328) enabled ABHD4 (ABHD4 N303R/S332G) to activate ATGL in Cos7 cells, brown adipocytes, and artificial lipid droplets. The corresponding ABHD5 mutations (ABHD5 R299N and ABHD5 G328S) selectively disrupted lipolysis without affecting ATGL lipid droplet translocation or ABHD5 interactions with perilipin proteins and ABHD5 ligands, demonstrating that ABHD5 lipase activation could be dissociated from its other functions. Structural modeling placed ABHD5 R299/G328 and R303/G332 from gain-of-function ABHD4 in close proximity on the ABHD protein surface, indicating they form part of a novel functional surface required for lipase activation. These data demonstrate distinct ABHD5 functional properties and provide new insights into the functional evolution of ABHD family members and the structural basis of lipase regulation.
Collapse
Affiliation(s)
- Matthew A. Sanders
- Center for Integrative Metabolic and Endocrine Research Wayne State University School of Medicine, Detroit, MI 48201, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Huamei Zhang
- Center for Integrative Metabolic and Endocrine Research Wayne State University School of Medicine, Detroit, MI 48201, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Ljiljana Mladenovic
- Center for Integrative Metabolic and Endocrine Research Wayne State University School of Medicine, Detroit, MI 48201, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yan Yuan Tseng
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - James G. Granneman
- Center for Integrative Metabolic and Endocrine Research Wayne State University School of Medicine, Detroit, MI 48201, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
99
|
Gholami S, Bordbar AK, Lohrasebi A. Identifying binding modes of two synthetic derivatives of adrenalin to the α2C-adrenoceptor by using molecular modeling; insights into the α2C-adrenoceptor activation. Biophys Chem 2017; 223:17-24. [PMID: 28187351 DOI: 10.1016/j.bpc.2017.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/28/2017] [Accepted: 01/31/2017] [Indexed: 10/20/2022]
Abstract
Although, α2C adrenergic receptor (AR) mediates a number of physiological functions in vivo and has great therapeutic potential, the absence of its crystal structure is a major difficulty in the activation mechanism studies and drug design endeavors. Here, a homology model of α2C AR has been presented by means of multiple sequence alignment. The used templates were the latest crystal structures of the other ARs (Protein Data Bank IDs: 2R4R, 2RH1, 4GPO, 3P0G, 4BVN and 4LDO) that have 38.4% identity with the query. We then conducted docking simulations to understand and analyze the binding of noradrenaline (NOR), and its derivatives, namely arachidonoyl adrenalin (AA-AD) and arachidonoyl noradrenalin (AA-NOR) to the receptor. The existence of H-bonds between the ligands and SER218 residue implies the same binding site of derivatives with respect to the NOR. AA-AD and AA-NOR bind to the receptor with the larger binding affinities. The presence of salt bridge between ARG149 and GLU377 in the free receptor, obtained from molecular dynamics studies proved that the receptor still is in its basal state before binding process take places. The activation process is characterized by increasing in the RMSD values of the backbone receptor in the bound state, increasing the RMSF of the transmembrane involved in the activation process and the disappearance of the ARG149-GLU377 salt bridge.
Collapse
Affiliation(s)
- Samira Gholami
- Department of Chemistry, University of Isfahan, Isfahan 81746-73441, Iran
| | - A Khalegh Bordbar
- Department of Chemistry, University of Isfahan, Isfahan 81746-73441, Iran.
| | - Amir Lohrasebi
- Department of Physics, University of Isfahan, Isfahan 81746-73441, Iran
| |
Collapse
|
100
|
Human Gpn1 purified from bacteria binds guanine nucleotides and hydrolyzes GTP as a protein dimer stabilized by its C-terminal tail. Protein Expr Purif 2017; 132:85-96. [PMID: 28153773 DOI: 10.1016/j.pep.2017.01.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 12/23/2016] [Accepted: 01/25/2017] [Indexed: 12/20/2022]
Abstract
The essential GTPase Gpn1 mediates RNA polymerase II nuclear targeting and controls microtubule dynamics in yeast and human cells by molecular mechanisms still under investigation. Here, we purified human HisGpn1 expressed as a recombinant protein in bacteria E. coli BL-21 (DE3). Affinity purified HisGpn1 eluted from a size exclusion column as a protein dimer, a state conserved after removing the hexa-histidine tail and confirmed by separating HisGpn1 in native gels, and in dynamic light scattering experiments. Human HisGpn1 purity was higher than 95%, molecularly monodisperse and could be concentrated to more than 10 mg/mL without aggregating. Circular dichroism spectra showed that human HisGpn1 was properly folded and displayed a secondary structure rich in alpha helices. HisGpn1 effectively bound GDP and the non-hydrolyzable GTP analogue GMPPCP, and hydrolyzed GTP. We next tested the importance of the C-terminal tail, present in eukaryotic Gpn1 but not in the ancestral archaeal Gpn protein, on HisGpn1 dimer formation. C-terminal deleted human HisGpn1 (HisGpn1ΔC) was also purified as a protein dimer, indicating that the N-terminal GTPase domain contains the interaction surface needed for dimer formation. In contrast to HisGpn1, however, HisGpn1ΔC dimer spontaneously dissociated into monomers. In conclusion, we have developed a method to purify properly folded and functionally active human HisGpn1 from bacteria, and showed that the C-terminal tail, universally conserved in all eukaryotic Gpn1 orthologues, stabilizes the GTPase domain-mediated Gpn1 protein dimer. The availability of recombinant human Gpn1 will open new research avenues to unveil the molecular and pharmacological properties of this essential GTPase.
Collapse
|