Impact of Thiol–Disulfide Balance on the Binding of Covid-19 Spike Protein with Angiotensin-Converting Enzyme 2 Receptor
- Sanchita HatiSanchita HatiDepartment of Chemistry, University of Wisconsin—Eau Claire, Eau Claire, Wisconsin 54701, United StatesMore by Sanchita Hati
- and
- Sudeep Bhattacharyya*Sudeep Bhattacharyya*Email: [email protected]Department of Chemistry, University of Wisconsin—Eau Claire, Eau Claire, Wisconsin 54701, United StatesMore by Sudeep Bhattacharyya
Abstract

The novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has led to an ongoing pandemic of coronavirus disease (COVID-19), which started in 2019. This is a member of Coronaviridae family in the genus Betacoronavirus, which also includes SARS-CoV and Middle East respiratory syndrome coronavirus (MERS-CoV). The angiotensin-converting enzyme 2 (ACE2) is the functional receptor for SARS-CoV and SARS-CoV-2 to enter the host cells. In particular, the interaction of viral spike proteins with ACE2 is a critical step in the viral replication cycle. The receptor-binding domain of the viral spike proteins and ACE2 have several cysteine residues. In this study, the role of thiol–disulfide balance on the interactions between SARS-CoV/CoV-2 spike proteins and ACE2 was investigated using molecular dynamics simulations. The study revealed that the binding affinity was significantly impaired when all of the disulfide bonds of both ACE2 and SARS-CoV/CoV-2 spike proteins were reduced to thiol groups. The impact on the binding affinity was less severe when the disulfide bridges of only one of the binding partners were reduced to thiols. This computational finding possibly provides a molecular basis for the differential COVID-19 cellular recognition due to the oxidative stress.
1. Introduction
2. Results and Discussion
Figure 1

Figure 1. Sequence alignment (generated by Clustal Omega (35)) between the receptor-binding domain of SARS-CoV and SARS-CoV-2 proteins. “*” represents the identical residues, “:” and “.” represent strongly and weakly similar residues, respectively, and gap represents dissimilar residues. The cysteine residues are highlighted in yellow.
Figure 2

Figure 2. Structures of protein complexes: (a) SARS-CoV···ACE2 and (b) SARS-CoV-2···ACE2. All of the disulfide bridges between cysteine residues are shown in green van der Waals (vdW) spheres and thiol groups in cyan licorice.

The cysteine residues are highlighted in yellow and disulfide bonds are shown using black solid lines. The start and end residues in the crystal structures are numbered and highlighted in red. The gray highlighted residues are missing in the crystal structure.
2.1. Structural Changes along the Trajectory
Figure 3

Figure 3. RMSDs, averaged over 1 ns MD simulation, of the protein complexes of ACE2 and SARS-CoV or SARS-CoV-2. Disulfide-containing proteins are referred to as oxidized with a shorthand notation of “ox”, while thiol variants are denoted with “red” notation.
protein systems | number of disulfide moieties remaining | disulfide bridges present | RMSD (Å) | fluctuation (Å) |
---|---|---|---|---|
CoVox···ACE2ox | 6 | CoV: C366:::C419, C467:::C474, ACE2: C133:::C141, C344:::C361, C366:::C419, C530:::C542 | 2.0 | 0.2 |
CoVred···ACE2ox | 4 | ACE2: C133:::C141, C344:::C361, C366:::C419, C530:::C542 | 3.8 | 0.3 |
CoVox···ACE2red | 2 | CoV: C366:::C419, C467:::C474 | 3.1 | 0.3 |
CoVred···ACE2red | 0 | none | 3.6 | 0.2 |
CoV-2ox···ACE2ox | 8 | CoV-2: C336:::C361, C379:::C432, C391:::C525, C480:::C488, ACE2: C133:::C141, C344:::C361, C366:::C419, C530:::C542 | ||
CoV-2red···ACE2ox | 4 | ACE2: C133:::C141, C344:::C361, C366:::C419, C530:::C542 | 2.7 | 0.2 |
CoV-2ox···ACE2red | 4 | CoV-2: C336:::C361, C379:::C432, C391:::C525, C480:::C488 | 2.8 | 0.2 |
CoV-2red···ACE2red | 0 | none | 3.1 | 0.2 |
For each protein system, the “RMSD” column contains the average of the last 5 ns RMSD, and the “fluctuation” column represents the standard deviation computed based on the starting structure as a reference.
2.2. Thermal Fluctuations due to the Cleavage of Disulfide Bridges
Figure 4

Figure 4. Change in residue-level RMSFs of Cα atoms in the oxidized and reduced protein complexes: (a) SARS-CoV···ACE2 and (b) SARS-CoV-2···ACE2. The cysteine residues, for each protein system (labeled in Figure 2), are highlighted as vdW spheres. The backbone as well as the cysteine residues are color-coded, large fluctuations are shown in red, and small fluctuations are in blue. The medium-scale fluctuations are shown in green.
2.3. Binding Study
protein systems | Coulombic interaction free energy ΔCoulG | corrected solvation free energy difference ΔΔsolvGcorr | Gibbs free energy of binding ΔbindG° |
---|---|---|---|
CoVox···ACE2ox | –564.5 | 557.2 | –7.3b |
CoVred···ACE2ox | –673.0 | 661.8 | –11.2 |
CoVox···ACE2red | –696.6 | 693.1 | –3.4 |
CoVred···ACE2red | –528.4 | 580.4 | 52.0 |
CoV-2ox···ACE2ox | –659.6 | 648.2 | –10.4c |
CoV-2red···ACE2ox | –638.2 | 632.2 | –6.0 |
CoV-2ox···ACE2red | –531.9 | 528.1 | –3.8 |
CoV-2red···ACE2red | –655.3 | 712.6 | 57.3 |
All energies are expressed in kcal/mol. An estimated uncertainty of 1–3 kcal/mol was determined for the computed Gibbs free energy.
An experimental value of −10.3 kcal/mol was obtained for SARS-CoV by Lan et al. (34)
2.4. Molecular Basis of the Impaired Binding
Figure 5

Figure 5. Comparison of the conformational change at the protein–protein interface in CoVox···ACE2ox (left) and CoVred···ACE2red (right). CoV and ACE2 subunits in the complex are shown in blue and red colors, respectively, at the center. The structural motif containing the two helices of ACE2 and a β sheet of the CoV (or CoV-2) was monitored before (top) and after (bottom) 20 ns MD simulation. The left and right panels show the difference in conformational changes in the oxidized form (CoVox···ACE2ox) and reduced form (CoVred···ACE2red), respectively.
3. Conclusions
4. Methodology
4.1. Computational Setup
4.2. Molecular Dynamics Simulations
4.3. Binding Free Energy Calculations



Scheme 1

Acknowledgments
We acknowledge computational support from the Blugold Super Computing Cluster (BGSC) of the University of Wisconsin—Eau Claire.
ACE | angiotensin-converting enzyme |
CoV | coronavirus |
MD | molecular dynamics |
RBD | receptor-binding domain |
TABI-PB | treecode-accelerated boundary integral Poisson–Boltzmann |
References
This article references 35 other publications.
- 1Schoeman, D.; Fielding, B. C. Coronavirus envelope protein: current knowledge. Virol. J. 2019, 16, 69 DOI: 10.1186/s12985-019-1182-0Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3M3gtFOrsQ%253D%253D&md5=c3c0865f45fb51cfe809c55d62207d59Coronavirus envelope protein: current knowledgeSchoeman Dewald; Fielding Burtram CVirology journal (2019), 16 (1), 69 ISSN:.BACKGROUND: Coronaviruses (CoVs) primarily cause enzootic infections in birds and mammals but, in the last few decades, have shown to be capable of infecting humans as well. The outbreak of severe acute respiratory syndrome (SARS) in 2003 and, more recently, Middle-East respiratory syndrome (MERS) has demonstrated the lethality of CoVs when they cross the species barrier and infect humans. A renewed interest in coronaviral research has led to the discovery of several novel human CoVs and since then much progress has been made in understanding the CoV life cycle. The CoV envelope (E) protein is a small, integral membrane protein involved in several aspects of the virus' life cycle, such as assembly, budding, envelope formation, and pathogenesis. Recent studies have expanded on its structural motifs and topology, its functions as an ion-channelling viroporin, and its interactions with both other CoV proteins and host cell proteins. MAIN BODY: This review aims to establish the current knowledge on CoV E by highlighting the recent progress that has been made and comparing it to previous knowledge. It also compares E to other viral proteins of a similar nature to speculate the relevance of these new findings. Good progress has been made but much still remains unknown and this review has identified some gaps in the current knowledge and made suggestions for consideration in future research. CONCLUSIONS: The most progress has been made on SARS-CoV E, highlighting specific structural requirements for its functions in the CoV life cycle as well as mechanisms behind its pathogenesis. Data shows that E is involved in critical aspects of the viral life cycle and that CoVs lacking E make promising vaccine candidates. The high mortality rate of certain CoVs, along with their ease of transmission, underpins the need for more research into CoV molecular biology which can aid in the production of effective anti-coronaviral agents for both human CoVs and enzootic CoVs.
- 2Fehr, A. R.; Perlman, S. Coronaviruses: an overview of their replication and pathogenesis. Methods Mol. Biol. 2015, 1282, 1– 23, DOI: 10.1007/978-1-4939-2438-7_1Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xls12ksL8%253D&md5=2e0fc18cd6b7312f0704f933a3614913Coronaviruses: an overview of their replication and pathogenesisFehr, Anthony R.; Perlman, StanleyMethods in Molecular Biology (New York, NY, United States) (2015), 1282 (Coronaviruses), 1-23CODEN: MMBIED; ISSN:1940-6029. (Springer)A review. Coronaviruses (CoVs), enveloped pos.-sense RNA viruses, are characterized by club-like spikes that project from their surface, an unusually large RNA genome, and a unique replication strategy. Coronaviruses cause a variety of diseases in mammals and birds ranging from enteritis in cows and pigs and upper respiratory disease in chickens to potentially lethal human respiratory infections. Here we provide a brief introduction to coronaviruses discussing their replication and pathogenicity, and current prevention and treatment strategies. We also discuss the outbreaks of the highly pathogenic Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) and the recently identified Middle Eastern Respiratory Syndrome Coronavirus (MERS-CoV).
- 3Chen, Y.; Guo, Y.; Pan, Y.; Zhao, Z. J. Structure analysis of the receptor binding of 2019-nCoV. Biochem. Biophys. Res. Commun. 2020, 525, 135– 140, DOI: 10.1016/j.bbrc.2020.02.071Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXjsFygtbs%253D&md5=53f249f15e3558acaf8eec89574bdf18Structure analysis of the receptor binding of 2019-nCoVChen, Yun; Guo, Yao; Pan, Yihang; Zhao, Zhizhuang JoeBiochemical and Biophysical Research Communications (2020), 525 (1), 135-140CODEN: BBRCA9; ISSN:0006-291X. (Elsevier B.V.)2019-nCoV is a newly identified coronavirus with high similarity to SARS-CoV. We performed a structural anal. of the receptor binding domain (RBD) of spike glycoprotein responsible for entry of coronaviruses into host cells. The RBDs from the two viruses share 72% identity in amino acid sequences, and mol. simulation reveals highly similar ternary structures. However, 2019-nCoV has a distinct loop with flexible glycyl residues replacing rigid prolyl residues in SARS-CoV. Mol. modeling revealed that 2019-nCoV RBD has a stronger interaction with angiotensin converting enzyme 2 (ACE2). A unique phenylalanine F486 in the flexible loop likely plays a major role because its penetration into a deep hydrophobic pocket in ACE2. ACE2 is widely expressed with conserved primary structures throughout the animal kingdom from fish, amphibians, reptiles, birds, to mammals. Structural anal. suggests that ACE2 from these animals can potentially bind RBD of 2019-nCoV, making them all possible natural hosts for the virus. 2019-nCoV is thought to be transmitted through respiratory droplets. However, since ACE2 is predominantly expressed in intestines, testis, and kidney, fecal-oral and other routes of transmission are also possible. Finally, antibodies and small mol. inhibitors that can block the interaction of ACE2 with RBD should be developed to combat the virus.
- 4Xu, H.; Zhong, L.; Deng, J.; Peng, J.; Dan, H.; Zeng, X.; Li, T.; Chen, Q. High expression of ACE2 receptor of 2019-nCoV on the epithelial cells of oral mucosa. Int. J. Oral Sci. 2020, 12, 8 DOI: 10.1038/s41368-020-0074-xGoogle Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXksVWnsL4%253D&md5=98de43264b244daee736cfdc99e87aadHigh expression of ACE2 receptor of 2019-nCoV on the epithelial cells of oral mucosaXu, Hao; Zhong, Liang; Deng, Jiaxin; Peng, Jiakuan; Dan, Hongxia; Zeng, Xin; Li, Taiwen; Chen, QianmingInternational Journal of Oral Science (2020), 12 (1), 8CODEN: IJOSFA; ISSN:2049-3169. (Nature Research)It has been reported that ACE2 is the main host cell receptor of 2019-nCoV and plays a crucial role in the entry of virus into the cell to cause the final infection. To investigate the potential route of 2019-nCov infection on the mucosa of oral cavity, bulk RNA-seq profiles from two public databases including The Cancer Genome Atlas (TCGA) and Functional Annotation of The Mammalian Genome Cap Anal. of Gene Expression (FANTOM5 CAGE) dataset were collected. RNA-seq profiling data of 13 organ types with para-carcinoma normal tissues from TCGA and 14 organ types with normal tissues from FANTOM5 CAGE were analyzed in order to explore and validate the expression of ACE2 on the mucosa of oral cavity. Further, single-cell transcriptomes from an independent data generated inhouse were used to identify and confirm the ACE2-expressing cell compn. and proportion in oral cavity. The results demonstrated that the ACE2 expressed on the mucosa of oral cavity. Interestingly, this receptor was highly enriched in epithelial cells of tongue. Preliminarily, those findings have explained the basic mechanism that the oral cavity is a potentially high risk for 2019-nCoV infectious susceptibility and provided a piece of evidence for the future prevention strategy in dental clin. practice as well as daily life.
- 5Ryser, H. J.; Levy, E. M.; Mandel, R.; DiSciullo, G. J. Inhibition of human immunodeficiency virus infection by agents that interfere with thiol-disulfide interchange upon virus-receptor interaction. Proc. Natl. Acad. Sci. U.S.A. 1994, 91, 4559– 4563, DOI: 10.1073/pnas.91.10.4559Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXktVSgs70%253D&md5=772426ff00253d510d64026f556da4f5Inhibition of human immunodeficiency virus infection by agents that interfere with thiol-disulfide interchange upon virus-receptor interactionRyser, Hugues J.-P.; Levy, Elinor M.; Mandel, Richard; DiSciullo, Gino J.Proceedings of the National Academy of Sciences of the United States of America (1994), 91 (10), 4559-63CODEN: PNASA6; ISSN:0027-8424.The cell surface of mammalian cells is capable of reductively cleaving disulfide bonds of exogenous membrane-bound macromols. (for instance, the interchain disulfide of diphtheria toxin), and inhibiting this process with membrane-impermeant sulfhydryl reagents prevents diphtheria toxin cytotoxicity. More recently it was found that the same membrane function can be inhibited by bacitracin, an inhibitor of protein disulfide-isomerase (PDI), and by monoclonal antibodies against PDI, suggesting that PDI catalyzes a thiol-disulfide interchange between its thiols and the disulfides of membrane-bound macromols. The authors provide evidence that the same reductive process plays a role in the penetration of membrane-bound human immunodeficiency virus (HIV) and show that HIV infection of human lymphoid cells is markedly inhibited by the membrane-impermeant sulfhydryl blocker 5,5'-dithiobis(2-nitrobenzoic acid), by bacitracin, and by anti-PDI antibodies. The results imply that HIV and its target cell engage in a thiol-disulfide interchange mediated by PDI and that the redn. of crit. disulfides in viral envelope glycoproteins may be the initial event that triggers conformational changes required for HIV entry and cell infection. These findings suggest addnl. approaches to impede cell infection by HIV.
- 6Gallina, A.; Hanley, T. M.; Mandel, R.; Trahey, M.; Broder, C. C.; Viglianti, G. A.; Ryser, H. J. Inhibitors of protein-disulfide isomerase prevent cleavage of disulfide bonds in receptor-bound glycoprotein 120 and prevent HIV-1 entry. J. Biol. Chem. 2002, 277, 50579– 50588, DOI: 10.1074/jbc.M204547200Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38Xps12ntrw%253D&md5=23bbd63c77a16fb983cd993886822676Inhibitors of Protein-Disulfide Isomerase Prevent Cleavage of Disulfide Bonds in Receptor-bound Glycoprotein 120 and Prevent HIV-1 EntryGallina, Angelo; Hanley, Timothy M.; Mandel, Richard; Trahey, Meg; Broder, Christopher C.; Viglianti, Gregory A.; Ryser, Hugues J.-P.Journal of Biological Chemistry (2002), 277 (52), 50579-50588CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)We previously reported that monoclonal antibodies to protein-disulfide isomerase (PDI) and other membrane-impermeant PDI inhibitors prevented HIV-1 infection. PDI is present at the surface of HIV-1 target cells and reduces disulfide bonds in a model peptide attached to the cell membrane. Here we show that sol. PDI cleaves disulfide bonds in recombinant envelope glycoprotein gp120 and that gp120 bound to the surface receptor CD4 undergoes a disulfide redn. that is prevented by PDI inhibitors. Concns. of inhibitors that prevent this redn. and inhibit the cleavage of surface-bound disulfide conjugate prevent infection at the level of HIV-1 entry. The entry of HIV-1 strains differing in their coreceptor specificities is similarly inhibited, and so is the redn. of gp120 bound to CD4 of coreceptor-neg. cells. PDI inhibitors also prevent HIV envelope-mediated cell-cell fusion but have no effect on the entry of HIV-1 pseudo-typed with murine leukemia virus envelope. Importantly, PDI coppts. with both sol. and cellular CD4. We propose that a PDI·CD4 assocn. at the cell surface enables PDI to reach CD4-bound virus and to reduce disulfide bonds present in the domain of gp120 that binds to CD4. Conformational changes resulting from the opening of gp120-disulfide loops may drive the processes of virus-cell and cell-cell fusion. The biochem. events described identify new potential targets for anti-HIV agents.
- 7Lavillette, D.; Barbouche, R.; Yao, Y.; Boson, B.; Cosset, F. L.; Jones, I. M.; Fenouillet, E. Significant redox insensitivity of the functions of the SARS-CoV spike glycoprotein: comparison with HIV envelope. J. Biol. Chem. 2006, 281, 9200– 9204, DOI: 10.1074/jbc.M512529200Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XjtVSrsrs%253D&md5=2b4152b59dc1dbe5d4a2f1e1b69bdafeSignificant Redox Insensitivity of the Functions of the SARS-CoV Spike Glycoprotein: Comparison with HIV envelopeLavillette, Dimitri; Barbouche, Rym; Yao, Yongxiu; Boson, Bertrand; Cosset, Francois-Loic; Jones, Ian M.; Fenouillet, EmmanuelJournal of Biological Chemistry (2006), 281 (14), 9200-9204CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)The capacity of the surface glycoproteins of enveloped viruses to mediate virus/cell binding and membrane fusion requires a proper thiol/disulfide balance. Chem. manipulation of their redox state using reducing agents or free sulfhydryl reagents affects virus/cell interaction. Conversely, natural thiol/disulfide rearrangements often occur during the cell interaction to trigger fusogenicity, hence the virus entry. We examd. the relationship between the redox state of the 20 cysteine residues of the SARS-CoV (severe acute respiratory syndrome coronavirus) Spike glycoprotein S1 subdomain and its functional properties. Mature S1 exhibited ∼4 unpaired cysteines, and chem. reduced S1 displaying up to ∼6 addnl. unpaired cysteines still bound ACE2 and enabled fusion. In addn., virus/cell membrane fusion occurred in the presence of sulfhydryl-blocking reagents and oxidoreductase inhibitors. Thus, in contrast to various viruses including HIV (human immunodeficiency virus) examd. in parallel, the functions of the SARS-CoV Spike glycoprotein exhibit a significant and surprising independence of redox state, which may contribute to the wide host range of the virus. These data suggest clues for molecularly engineering vaccine immunogens.
- 8Pan, S.; Chen, H. H.; Correia, C.; Dai, H.; Witt, T. A.; Kleppe, L. S.; Burnett, J. C., Jr.; Simari, R. D. Cell surface protein disulfide isomerase regulates natriuretic peptide generation of cyclic guanosine monophosphate. PLoS One 2014, 9, e112986 DOI: 10.1371/journal.pone.0112986Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitV2htb7L&md5=2b7f451941a888a4e0b170bf8f510138Cell surface protein disulfide isomerase regulates natriuretic peptide generation of cyclic guanosine monophosphatePan, Shuchong; Chen, Horng H.; Correia, Cristina; Dai, Haiming; Witt, Tyra A.; Kleppe, Laurel S.; Burnett, John C., Jr.; Simari, Robert D.PLoS One (2014), 9 (11), e112986/1-e112986/14, 14 pp.CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)Rationale: The family of natriuretic peptides (NPs), including atrial natriuretic peptide (ANP), B-type natriuretic peptide (BNP), and C-type natriuretic peptide (CNP), exert important and diverse actions for cardiovascular and renal homeostasis. The autocrine and paracrine functions of the NPs are primarily mediated through the cellular membrane bound guanylyl cyclase-linked receptors GC-A (NPR-A) and GC-B (NPR-B). As the ligands and receptors each contain disulfide bonds, a regulatory role for the cell surface protein disulfide isomerase (PDI) was investigated. Objective: We utilized complementary in vitro and in vivo models to det. the potential role of PDI in regulating the ability of the NPs to generate its second messenger, cyclic guanosine monophosphate. Methods and Results: Inhibition of PDI attenuated the ability of ANP, BNP and CNP to generate cGMP in human mesangial cells (HMCs), human umbilical vein endothelial cells (HUVECs), and human aortic smooth muscle cells (HASMCs), each of which were shown to express PDI. In LLC-PK1 cells, where PDI expression was undetectable by immunoblotting, PDI inhibition had a minimal effect on cGMP generation. Addn. of PDI to cultured LLC-PK1 cells increased intracellular cGMP generation mediated by ANP. Inhibition of PDI in vivo attenuated NP-mediated generation of cGMP by ANP. Surface Plasmon Resonance demonstrated modest and differential binding of the natriuretic peptides with immobilized PDI in a cell free system. However, PDI was shown to co-localize on the surface of cells with GC-A and GC-B by co-immunoprecpitation and immunohistochem. Conclusion: These data demonstrate for the first time that cell surface PDI expression and function regulate the capacity of natriuretic peptides to generate cGMP through interaction with their receptors.
- 9Mathys, L.; Balzarini, J. The role of cellular oxidoreductases in viral entry and virus infection-associated oxidative stress: potential therapeutic applications. Expert Opin. Ther. Targets 2016, 20, 123– 143, DOI: 10.1517/14728222.2015.1068760Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslKltbs%253D&md5=392ee62019de0363448d2b5fe86ac408The role of cellular oxidoreductases in viral entry and virus infection-associated oxidative stress: potential therapeutic applicationsMathys, Leen; Balzarini, JanExpert Opinion on Therapeutic Targets (2016), 20 (1), 123-143CODEN: EOTTAO; ISSN:1472-8222. (Taylor & Francis Ltd.)Cellular oxidoreductases catalyze thiol/disulfide exchange reactions in susceptible proteins and contribute to the cellular defense against oxidative stress. Oxidoreductases and oxidative stress are also involved in viral infections. In this overview, different aspects of the role of cellular oxidoreductases and oxidative stress during viral infections are discussed from a chemotherapeutic viewpoint. Entry of enveloped viruses into their target cells is triggered by the interaction of viral envelope glycoproteins with cellular (co)receptor(s) and depends on obligatory conformational changes in these viral envelope glycoproteins and/or cellular receptors. For some viruses, these conformational changes are mediated by cell surface-assocd. cellular oxidoreductases, which mediate disulfide bridge redns. in viral envelope glycoprotein(s). Therefore, targeting these oxidoreductases using oxidoreductase inhibitors might yield an interesting strategy to block viral entry of these viruses. Furthermore, since viral infections are often assocd. with systemic oxidative stress, contributing to disease progression, the enhancement of the cellular antioxidant defense systems might have potential as an adjuvant antiviral strategy, slowing down disease progression. Promising antiviral data were obtained for both strategies. However, potential pitfalls have also been identified for these strategies, indicating that it is important to carefully assess the benefits vs. risks of these antiviral strategies.
- 10Camini, F. C.; da Silva Caetano, C. C.; Almeida, L. T.; de Brito Magalhães, C. L. Implications of oxidative stress on viral pathogenesis. Arch. Virol. 2017, 162, 907– 917, DOI: 10.1007/s00705-016-3187-yGoogle Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtVOntg%253D%253D&md5=8acc801377bd621b2ea2fc12b5178fd7Implications of oxidative stress on viral pathogenesisCamini, Fernanda Caetano; Caetano, Camila Carla da Silva; Almeida, Leticia Trindade; Magalhaes, Cintia Lopes de BritoArchives of Virology (2017), 162 (4), 907-917CODEN: ARVIDF; ISSN:0304-8608. (Springer-Verlag GmbH)Reactive species are frequently formed after viral infections. Antioxidant defences, including enzymic and non-enzymic components, protect against reactive species, but sometimes these defences are not completely adequate. An imbalance in the prodn. of reactive species and the body's inability to detoxify these reactive species is referred to as oxidative stress. The aim of this review is to analyze the role of oxidative stress in the pathogenesis of viral infections and highlight some major therapeutic approaches that have gained importance, with regards to controlling virus-induced oxidative injury. Attention will be focused on DNA viruses (papillomaviruses, hepadnaviruses), RNA viruses (flaviviruses, orthomyxoviruses, paramyxoviruses, togaviruses) and retroviruses (human immunodeficiency virus). In general, viruses cause an imbalance in the cellular redox environment, which depending on the virus and the cell can result in different responses, e.g. cell signaling, antioxidant defences, reactive species, and other processes. Therefore, the modulation of reactive species prodn. and oxidative stress potentially represents a novel pharmacol. approach for reducing the consequences of viral pathogenesis.
- 11Fenouillet, E.; Barbouche, R.; Jones, I. M. Cell entry by enveloped viruses: redox considerations for HIV and SARS-coronavirus. Antioxid. Redox Signaling 2007, 9, 1009– 1034, DOI: 10.1089/ars.2007.1639Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXot1Wktrk%253D&md5=14c6275868393216f703e406135af1a1Cell entry by enveloped viruses: redox considerations for HIV and SARS-coronavirusFenouillet, Emmanuel; Barbouche, Rym; Jones, Ian M.Antioxidants & Redox Signaling (2007), 9 (8), 1009-1034CODEN: ARSIF2; ISSN:1523-0864. (Mary Ann Liebert, Inc.)A review. For enveloped viruses, genome entry into the target cell involves two major steps: virion binding to the cell-surface receptor and fusion of the virion and cell membranes. Virus-cell membrane fusion is mediated by the virus envelope complex, and its fusogenicity is the result of an active virus-cell interaction process that induces conformation changes within the envelope. For some viruses, such as influenza, exposure to an acidic milieu within the cell during the early steps of infection triggers the necessary structural changes. However, for other pathogens which are not exposed to such environmental stress, activation of fusogenicity can result from precise thiol/disulfide rearrangements mediated by either an endogenous redox autocatalytic isomerase or a cell-assocd. oxidoreductase. Study of the activation of HIV envelope fusogenicity has revealed new knowledge about how redox changes within a viral envelope trigger fusion. We discuss these findings and their implication for anti-HIV therapy. In addn., to compare and contrast the situation outlined for HIV with an enveloped virus that can fuse with the cell plasma membrane independent of the redox status of its envelope protein, we review parallel data obtained on SARS coronavirus entry.
- 12Yi, M. C.; Khosla, C. Thiol-Disulfide Exchange Reactions in the Mammalian Extracellular Environment. Annu. Rev. Chem. Biomol. Eng. 2016, 7, 197– 222, DOI: 10.1146/annurev-chembioeng-080615-033553Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XltFGrsLs%253D&md5=4c2bb579953388a3de9ab8c86c606a22Thiol-Disulfide Exchange Reactions in the Mammalian Extracellular EnvironmentYi, Michael C.; Khosla, ChaitanAnnual Review of Chemical and Biomolecular Engineering (2016), 7 (), 197-222CODEN: ARCBCY; ISSN:1947-5438. (Annual Reviews)Disulfide bonds represent versatile posttranslational modifications whose roles encompass the structure, catalysis, and regulation of protein function. Due to the oxidizing nature of the extracellular environment, disulfide bonds found in secreted proteins were once believed to be inert. This notion has been challenged by the discovery of redox-sensitive disulfides that, once cleaved, can lead to changes in protein activity. These functional disulfides are twisted into unique configurations, leading to high strain and potential energy. In some cases, cleavage of these disulfides can lead to a gain of function in protein activity. Thus, these motifs can be referred to as switches. We describe the couples that control redox in the extracellular environment, examine several examples of proteins with switchable disulfides, and discuss the potential applications of disulfides in mol. biol.
- 13Li, F. Structural analysis of major species barriers between humans and palm civets for severe acute respiratory syndrome coronavirus infections. J. Virol. 2008, 82, 6984– 6991, DOI: 10.1128/JVI.00442-08Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXosFWmsro%253D&md5=5df8566921a86428b63d3e3b261896b3Structural analysis of major species barriers between humans and palm civets for severe acute respiratory syndrome coronavirus infectionsLi, FangJournal of Virology (2008), 82 (14), 6984-6991CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)It is believed that a novel coronavirus, severe acute respiratory syndrome coronavirus (SARS-CoV), was passed from palm civets to humans and caused the epidemic of SARS in 2002 to 2003. The major species barriers between humans and civets for SARS-CoV infections are the specific interactions between a defined receptor-binding domain (RBD) on a viral spike protein and its host receptor, angiotensin-converting enzyme 2 (ACE2). In this study a chimeric ACE2 bearing the crit. N-terminal helix from civet and the remaining peptidase domain from human was constructed, and it was shown that this construct has the same receptor activity as civet ACE2. In addn., crystal structures of the chimeric ACE2 complexed with RBDs from various human and civet SARS-CoV strains were detd. These structures, combined with a previously detd. structure of human ACE2 complexed with the RBD from a human SARS-CoV strain, have revealed a structural basis for understanding the major species barriers between humans and civets for SARS-CoV infections. They show that the major species barriers are detd. by interactions between four ACE2 residues (residues 31, 35, 38, and 353) and two RBD residues (residues 479 and 487), that early civet SARS-CoV isolates were prevented from infecting human cells due to imbalanced salt bridges at the hydrophobic virus/receptor interface, and that SARS-CoV has evolved to gain sustained infectivity for human cells by eliminating unfavorable free charges at the interface through stepwise mutations at positions 479 and 487. These results enhance our understanding of host adaptations and cross-species infections of SARS-CoV and other emerging animal viruses.
- 14Wrapp, D.; Wang, N.; Corbett, K. S.; Goldsmith, J. A.; Hsieh, C. L.; Abiona, O.; Graham, B. S.; McLellan, J. S. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science 2020, 367, 1260– 1263, DOI: 10.1126/science.abb2507Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXkvFemt70%253D&md5=27d08cbb9a43d1da051a8a92a9f68aa5Cryo-EM structure of the 2019-nCoV spike in the prefusion conformationWrapp, Daniel; Wang, Nianshuang; Corbett, Kizzmekia S.; Goldsmith, Jory A.; Hsieh, Ching-Lin; Abiona, Olubukola; Graham, Barney S.; McLellan, Jason S.Science (Washington, DC, United States) (2020), 367 (6483), 1260-1263CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)The outbreak of a novel coronavirus (2019-nCoV) represents a pandemic threat that has been declared a public health emergency of international concern. The CoV spike (S) glycoprotein is a key target for vaccines, therapeutic antibodies, and diagnostics. To facilitate medical countermeasure development, we detd. a 3.5-angstrom-resoln. cryo-electron microscopy structure of the 2019-nCoV S trimer in the prefusion conformation. The predominant state of the trimer has one of the three receptor-binding domains (RBDs) rotated up in a receptor-accessible conformation. We also provide biophys. and structural evidence that the 2019-nCoV S protein binds angiotensin-converting enzyme 2 (ACE2) with higher affinity than does severe acute respiratory syndrome (SARS)-CoV S. Addnl., we tested several published SARS-CoV RBD-specific monoclonal antibodies and found that they do not have appreciable binding to 2019-nCoV S, suggesting that antibody cross-reactivity may be limited between the two RBDs. The structure of 2019-nCoV S should enable the rapid development and evaluation of medical countermeasures to address the ongoing public health crisis.
- 15Wang, Q.; Zhang, Y.; Wu, L.; Niu, S.; Song, C.; Zhang, Z.; Lu, G.; Qiao, C.; Hu, Y.; Yuen, K. Y.; Wang, Q.; Zhou, H.; Yan, J.; Qi, J. Structural and Functional Basis of SARS-CoV-2 Entry by Using Human ACE2. Cell 2020, 181, 894– 904.e9, DOI: 10.1016/j.cell.2020.03.045Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXmvFGrur4%253D&md5=188108e44f104860d4a2b83707ce7230Structural and Functional Basis of SARS-CoV-2 Entry by Using Human ACE2Wang, Qihui; Zhang, Yanfang; Wu, Lili; Niu, Sheng; Song, Chunli; Zhang, Zengyuan; Lu, Guangwen; Qiao, Chengpeng; Hu, Yu; Yuen, Kwok-Yung; Wang, Qisheng; Zhou, Huan; Yan, Jinghua; Qi, JianxunCell (Cambridge, MA, United States) (2020), 181 (4), 894-904.e9CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The recent emergence of a novel coronavirus (SARS-CoV-2) in China has caused significant public health concerns. Recently, ACE2 was reported as an entry receptor for SARS-CoV-2. In this study, we present the crystal structure of the C-terminal domain of SARS-CoV-2 (SARS-CoV-2-CTD) spike (S) protein in complex with human ACE2 (hACE2), which reveals a hACE2-binding mode similar overall to that obsd. for SARS-CoV. However, at. details at the binding interface demonstrate that key residue substitutions in SARS-CoV-2-CTD slightly strengthen the interaction and lead to higher affinity for receptor binding than SARS-RBD. Addnl., a panel of murine monoclonal antibodies (mAbs) and polyclonal antibodies (pAbs) against SARS-CoV-S1/receptor-binding domain (RBD) were unable to interact with the SARS-CoV-2 S protein, indicating notable differences in antigenicity between SARS-CoV and SARS-CoV-2. These findings shed light on the viral pathogenesis and provide important structural information regarding development of therapeutic countermeasures against the emerging virus.
- 16Yan, R.; Zhang, Y.; Li, Y.; Xia, L.; Guo, Y.; Zhou, Q. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science 2020, 367, 1444– 1448, DOI: 10.1126/science.abb2762Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXlslymsLo%253D&md5=ff4dfdfc646ea878cfb325019160e94aStructural basis for the recognition of SARS-CoV-2 by full-length human ACE2Yan, Renhong; Zhang, Yuanyuan; Li, Yaning; Xia, Lu; Guo, Yingying; Zhou, QiangScience (Washington, DC, United States) (2020), 367 (6485), 1444-1448CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)Angiotensin-converting enzyme 2 (ACE2) is the cellular receptor for severe acute respiratory syndrome coronavirus (SARS-CoV) and the new coronavirus (SARS-CoV-2) that is causing the serious coronavirus disease 2019 (COVID-19) epidemic. Here, we present cryo-electron microscopy structures of full-length human ACE2 in the presence of the neutral amino acid transporter B0AT1 with or without the receptor binding domain (RBD) of the surface spike glycoprotein (S protein) of SARS-CoV-2, both at an overall resoln. of 2.9 angstroms, with a local resoln. of 3.5 angstroms at the ACE2-RBD interface. The ACE2-B0AT1 complex is assembled as a dimer of heterodimers, with the collectrin-like domain of ACE2 mediating homodimerization. The RBD is recognized by the extracellular peptidase domain of ACE2 mainly through polar residues. These findings provide important insights into the mol. basis for coronavirus recognition and infection.
- 17Adams, L. M.; Andrews, R. J.; Hu, Q. H.; Schmit, H. L.; Hati, S.; Bhattacharyya, S. Crowder-Induced Conformational Ensemble Shift in Escherichia coli Prolyl-tRNA Synthetase. Biophys. J. 2019, 117, 1269– 1284, DOI: 10.1016/j.bpj.2019.08.033Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhslyisLzJ&md5=c2c9242b3ced38f0814a96f577208347Crowder-Induced Conformational Ensemble Shift in Escherichia coli Prolyl-tRNA SynthetaseAdams, Lauren M.; Andrews, Ryan J.; Hu, Quin H.; Schmit, Heidi L.; Hati, Sanchita; Bhattacharyya, SudeepBiophysical Journal (2019), 117 (7), 1269-1284CODEN: BIOJAU; ISSN:0006-3495. (Cell Press)The effect of mol. crowding on the structure and function of Escherichia coli prolyl-tRNA synthetase (Ec ProRS), a member of the aminoacyl-tRNA synthetase family, has been investigated using a combined exptl. and theor. method. Ec ProRS is a multidomain enzyme; coupled-domain dynamics are essential for efficient catalysis. To gain insight into the mechanistic detail of the crowding effect, kinetic studies were conducted with varying concns. and sizes of crowders. In parallel, spectroscopic and quantum chem. studies were employed to probe the "soft interactions" between crowders and protein side chains. Finally, the dynamics of the dimeric protein was examd. in the presence of crowders using a long-duration (70 ns) classical mol. dynamic simulations. The results of the simulations revealed a shift in the conformational ensemble, which is consistent with the preferential exclusion of cosolutes. The "soft interactions" model of the crowding effect also explained the alteration in kinetic parameters. In summary, the study found that the effects of mol. crowding on both conformational dynamics and catalytic function are correlated in the multidomain Ec ProRS, an enzyme that is central to protein synthesis in all living cells. This study affirmed that large and small cosolutes have considerable impacts on the structure, dynamics, and function of modular proteins and therefore must be considered for stabilizing protein-based pharmaceuticals and industrial enzymes.
- 18Bechtel, T. J.; Weerapana, E. From structure to redox: The diverse functional roles of disulfides and implications in disease. Proteomics 2017, 17, 1600391 DOI: 10.1002/pmic.201600391Google ScholarThere is no corresponding record for this reference.
- 19Ballatori, N.; Krance, S. M.; Marchan, R.; Hammond, C. L. Plasma membrane glutathione transporters and their roles in cell physiology and pathophysiology. Mol. Aspects Med. 2009, 30, 13– 28, DOI: 10.1016/j.mam.2008.08.004Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXltFelurc%253D&md5=43e9fd45d618eecfd3b2c73780b0f833Plasma membrane glutathione transporters and their roles in cell physiology and pathophysiologyBallatori, Nazzareno; Krance, Suzanne M.; Marchan, Rosemarie; Hammond, Christine L.Molecular Aspects of Medicine (2009), 30 (1-2), 13-28CODEN: MAMED5; ISSN:0098-2997. (Elsevier B.V.)A review. Reduced glutathione (GSH) is crit. for many cellular processes, and both its intracellular and extracellular concns. are tightly regulated. Intracellular GSH levels are regulated by two main mechanisms: by adjusting the rates of synthesis and of export from cells. Some of the proteins responsible for GSH export from mammalian cells have recently been identified, and there is increasing evidence that these GSH exporters are multispecific and multifunctional, regulating a no. of key biol. processes. In particular, some of the multidrug resistance-assocd. proteins (Mrp/Abcc) appear to mediate GSH export and homeostasis. The Mrp proteins mediate not only GSH efflux, but they also export oxidized glutathione derivs. (e.g., glutathione disulfide (GSSG), S-nitrosoglutathione (GS-NO), and glutathione-metal complexes), as well as other glutathione S-conjugates. The ability to export both GSH and oxidized derivs. of GSH, endows these transporters with the capacity to directly regulate the cellular thiol-redox status, and therefore the ability to influence many key signaling and biochem. pathways. Among the many processes that are influenced by the GSH transporters are apoptosis, cell proliferation, and cell differentiation. This report summarizes the evidence that Mrps contribute to the regulation of cellular GSH levels and the thiol-redox state, and thus to the many biochem. processes that are influenced by this tripeptide.
- 20Xia, H.; Suda, S.; Bindom, S.; Feng, Y.; Gurley, S. B.; Seth, D.; Navar, L. G.; Lazartigues, E. ACE2-mediated reduction of oxidative stress in the central nervous system is associated with improvement of autonomic function. PLoS One 2011, 6, e22682 DOI: 10.1371/journal.pone.0022682Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtVOisr%252FP&md5=8a10eafb719a132285886913f5c245c0ACE2-mediated reduction of oxidative stress in the central nervous system is associated with improvement of autonomic functionXia, Huijing; Suda, Sonia; Bindom, Sharell; Feng, Yumei; Gurley, Susan B.; Seth, Dale; Navar, L. Gabriel; Lazartigues, EricPLoS One (2011), 6 (7), e22682CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)Oxidative stress in the central nervous system mediates the increase in sympathetic tone that precedes the development of hypertension. We hypothesized that by transforming Angiotensin-II (AngII) into Ang-(1-7), ACE2 might reduce AngII-mediated oxidative stress in the brain and prevent autonomic dysfunction. To test this hypothesis, a relationship between ACE2 and oxidative stress was first confirmed in a mouse neuroblastoma cell line (Neuro2A cells) treated with AngII and infected with Ad-hACE2. ACE2 overexpression resulted in a redn. of reactive oxygen species (ROS) formation. In vivo, ACE2 knockout (ACE2-/y) mice and non-transgenic (NT) littermates were infused with AngII (10 days) and infected with Ad-hACE2 in the paraventricular nucleus (PVN). Baseline blood pressure (BP), AngII and brain ROS levels were not different between young mice (12 wk). However, cardiac sympathetic tone, brain NADPH oxidase and SOD activities were significantly increased in ACE2-/y. Post infusion, plasma and brain AngII levels were also significantly higher in ACE2-/y, although BP was similarly increased in both genotypes. ROS formation in the PVN and RVLM was significantly higher in ACE2-/y mice following AngII infusion. Similar phenotypes, i.e. increased oxidative stress, exacerbated dysautonomia and hypertension, were also obsd. on baseline in mature ACE2-/y mice (48 wk). ACE2 gene therapy to the PVN reduced AngII-mediated increase in NADPH oxidase activity and normalized cardiac dysautonomia in ACE2-/y mice. Altogether, these data indicate that ACE2 gene deletion promotes age-dependent oxidative stress, autonomic dysfunction and hypertension, while PVN-targeted ACE2 gene therapy decreases ROS formation via NADPH oxidase inhibition and improves autonomic function. Accordingly, ACE2 could represent a new target for the treatment of hypertension-assocd. dysautonomia and oxidative stress.
- 21Humphrey, W.; Dalke, A.; Schulten, K. VMD: visual molecular dynamics. J. Mol. Graphics 1996, 14, 33– 38, DOI: 10.1016/0263-7855(96)00018-5Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28Xis12nsrg%253D&md5=1e3094ec3151fb85c5ff05f8505c78d5VDM: visual molecular dynamicsHumphrey, William; Dalke, Andrew; Schulten, KlausJournal of Molecular Graphics (1996), 14 (1), 33-8, plates, 27-28CODEN: JMGRDV; ISSN:0263-7855. (Elsevier)VMD is a mol. graphics program designed for the display and anal. of mol. assemblies, in particular, biopolymers such as proteins and nucleic acids. VMD can simultaneously display any no. of structures using a wide variety of rendering styles and coloring methods. Mols. are displayed as one or more "representations," in which each representation embodies a particular rendering method and coloring scheme for a selected subset of atoms. The atoms displayed in each representation are chosen using an extensive atom selection syntax, which includes Boolean operators and regular expressions. VMD provides a complete graphical user interface for program control, as well as a text interface using the Tcl embeddable parser to allow for complex scripts with variable substitution, control loops, and function calls. Full session logging is supported, which produces a VMD command script for later playback. High-resoln. raster images of displayed mols. may be produced by generating input scripts for use by a no. of photorealistic image-rendering applications. VMD has also been expressly designed with the ability to animate mol. dynamics (MD) simulation trajectories, imported either from files or from a direct connection to a running MD simulation. VMD is the visualization component of MDScope, a set of tools for interactive problem solving in structural biol., which also includes the parallel MD program NAMD, and the MDCOMM software used to connect the visualization and simulation programs, VMD is written in C++, using an object-oriented design; the program, including source code and extensive documentation, is freely available via anonymous ftp and through the World Wide Web.
- 22Guvench, O.; Hatcher, E.; Venable, R. M.; Pastor, R. W.; MacKerell, A. D. CHARMM Additive All-Atom Force Field for Glycosidic Linkages between Hexopyranoses. J. Chem. Theory Comput. 2009, 5, 2353– 2370, DOI: 10.1021/ct900242eGoogle Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtVartbrM&md5=575b6daab16fbafc4d00ee445f16328dCHARMM Additive All-Atom Force Field for Glycosidic Linkages between HexopyranosesGuvench, Olgun; Hatcher, Elizabeth; Venable, Richard M.; Pastor, Richard W.; MacKerell, Alexander D., Jr.Journal of Chemical Theory and Computation (2009), 5 (9), 2353-2370CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)We present an extension of the CHARMM hexopyranose monosaccharide additive all-atom force field to enable modeling of glycosidic-linked hexopyranose polysaccharides. The new force field parameters encompass 1-1, 1-2, 1-3, 1-4, and 1-6 hexopyranose glycosidic linkages, as well as O-methylation at the C1 anomeric carbon, and are developed to be consistent with the CHARMM all-atom biomol. force fields for proteins, nucleic acids, and lipids. The parameters are developed in a hierarchical fashion using model compds. contg. the key atoms in the full carbohydrates, in particular O-methyl-tetrahydropyran and glycosidic-linked dimers consisting of two mols. of tetrahyropyran or one mol. of tetrahydropyran and one of cyclohexane. Target data for parameter optimization include full two-dimensional energy surfaces defined by the Φ/Ψ glycosidic dihedral angles in the disaccharide analogs, as detd. by quantum mech. MP2/cc-pVTZ single point energies on MP2/6-31G(d) optimized structures (MP2/cc-pVTZ//MP2/6-31G(d)). In order to achieve balanced, transferable dihedral parameters for the Φ/Ψ glycosidic dihedral angles, surfaces for all possible chiralities at the ring carbon atoms involved in the glycosidic linkages are considered, resulting in over 5000 MP2/cc-pVTZ//MP2/6-31G(d) conformational energies. Also included as target data are vibrational frequencies, pair interaction energies and distances with water mols., and intramol. geometries including distortion of the glycosidic valence angle as a function of the glycosidic dihedral angles. The model compd. optimized force field parameters are validated on full disaccharides through the comparison of mol. dynamics results to available exptl. data. Good agreement is achieved with expt. for a variety of properties including crystal cell parameters and intramol. geometries, aq. densities, and aq. NMR coupling consts. assocd. with the glycosidic linkage. The newly developed parameters allow for the modeling of linear, branched, and cyclic hexopyranose glycosides both alone and in heterogeneous systems including proteins, nucleic acids, and/or lipids when combined with existing CHARMM biomol. force fields.
- 23Raman, E. P.; Guvench, O.; MacKerell, A. D., Jr. CHARMM additive all-atom force field for glycosidic linkages in carbohydrates involving furanoses. J. Phys. Chem. B 2010, 114, 12981– 12994, DOI: 10.1021/jp105758hGoogle Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtFKlu73F&md5=a4619fcb09cafebb3f84a3d7f59ea999CHARMM Additive All-Atom Force Field for Glycosidic Linkages in Carbohydrates Involving FuranosesRaman, E. Prabhu; Guvench, Olgun; MacKerell, Alexander D., Jr.Journal of Physical Chemistry B (2010), 114 (40), 12981-12994CODEN: JPCBFK; ISSN:1520-6106. (American Chemical Society)Presented is an extension of the CHARMM additive carbohydrate all-atom force field to enable modeling of polysaccharides contg. furanose sugars. The new force field parameters encompass 1 ↔ 2, 1 → 3, 1 → 4, and 1 → 6 pyranose-furanose linkages and 2 → 1 and 2 → 6 furanose-furanose linkages, building on existing hexopyranose and furanose monosaccharide parameters. The model compds. were chosen to be monomers or glycosidic-linked dimers of tetrahydropyran (THP) and THF as to contain the key atoms in full carbohydrates. Target data for optimization included two-dimensional quantum mech. (QM) potential energy scans of the Φ/Ψ glycosidic dihedral angles, with geometry optimization at the MP2/6-31G(d) level followed by MP2/cc-pVTZ single-point energies. All possible chiralities of the model compds. at the linkage carbons were considered, and for each geometry, the THF ring was constrained to the favorable south or north conformations. Target data also included QM vibrational frequencies and pair interaction energies and distances with water mols. Force field validation included comparison of computed crystal properties, aq. soln. densities, and NMR J-coupling consts. to exptl. ref. values. Simulations of infinite crystals showed good agreement with exptl. values for intramol. geometries as well as for crystal unit cell parameters. Addnl., aq. soln. densities and available NMR data were reproduced to a high degree of accuracy, thus validating the hierarchically optimized parameters in both cryst. and aq. condensed phases. The newly developed parameters allow for the modeling of linear, branched, and cyclic pyranose/furanose polysaccharides both alone and in heterogeneous systems including proteins, nucleic acids, and/or lipids when combined with existing additive CHARMM biomol. force fields.
- 24Guvench, O.; Mallajosyula, S. S.; Raman, E. P.; Hatcher, E.; Vanommeslaeghe, K.; Foster, T. J.; Jamison, F. W., II; Mackerell, A. D., Jr. CHARMM additive all-atom force field for carbohydrate derivatives and its utility in polysaccharide and carbohydrate-protein modeling. J. Chem. Theory Comput. 2011, 7, 3162– 3180, DOI: 10.1021/ct200328pGoogle Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtFSru77P&md5=fc3c05ef4c95895ee3e7b08472ef3554CHARMM Additive All-Atom Force Field for Carbohydrate Derivatives and Its Utility in Polysaccharide and Carbohydrate-Protein ModelingGuvench, Olgun; Mallajosyula, Sairam S.; Raman, E. Prabhu; Hatcher, Elizabeth; Vanommeslaeghe, Kenno; Foster, Theresa J.; Jamison, Francis W.; MacKerell, Alexander D.Journal of Chemical Theory and Computation (2011), 7 (10), 3162-3180CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Monosaccharide derivs. such as xylose, fucose, N-acetylglucosamine (GlcNAc), N-acetylgalactosamine (GlaNAc), glucuronic acid, iduronic acid, and N-acetylneuraminic acid (Neu5Ac) are important components of eukaryotic glycans. The present work details the development of force-field parameters for these monosaccharides and their covalent connections to proteins via O linkages to serine or threonine side chains and via N linkages to asparagine side chains. The force field development protocol was designed to explicitly yield parameters that are compatible with the existing CHARMM additive force field for proteins, nucleic acids, lipids, carbohydrates, and small mols. Therefore, when combined with previously developed parameters for pyranose and furanose monosaccharides, for glycosidic linkages between monosaccharides, and for proteins, the present set of parameters enables the mol. simulation of a wide variety of biol. important mols. such as complex carbohydrates and glycoproteins. Parametrization included fitting to quantum mech. (QM) geometries and conformational energies of model compds., as well as to QM pair interaction energies and distances of model compds. with water. Parameters were validated in the context of crystals of relevant monosaccharides, as well NMR and/or x-ray crystallog. data on larger systems including oligomeric hyaluronan, sialyl Lewis X, O- and N-linked glycopeptides, and a lectin:sucrose complex. As the validated parameters are an extension of the CHARMM all-atom additive biomol. force field, they further broaden the types of heterogeneous systems accessible with a consistently developed force-field model.
- 25Best, R. B.; Zhu, X.; Shim, J.; Lopes, P. E. M.; Mittal, J.; Feig, M.; MacKerell, A. D. Optimization of the additive CHARMM all-atom protein force field targeting improved sampling of the backbone φ, ψ and side-chain χ(1) and χ(2) dihedral angles. J. Chem. Theory Comput. 2012, 8, 3257– 3273, DOI: 10.1021/ct300400xGoogle Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtVKqurfP&md5=9a48a0c5770fb1e887c3bb34d45b1354Optimization of the Additive CHARMM All-Atom Protein Force Field Targeting Improved Sampling of the Backbone .vphi., ψ and Side-Chain χ1 and χ2 Dihedral AnglesBest, Robert B.; Zhu, Xiao; Shim, Jihyun; Lopes, Pedro E. M.; Mittal, Jeetain; Feig, Michael; MacKerell, Alexander D.Journal of Chemical Theory and Computation (2012), 8 (9), 3257-3273CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)While the quality of the current CHARMM22/CMAP additive force field for proteins has been demonstrated in a large no. of applications, limitations in the model with respect to the equil. between the sampling of helical and extended conformations in folding simulations have been noted. To overcome this, as well as make other improvements in the model, we present a combination of refinements that should result in enhanced accuracy in simulations of proteins. The common (non-Gly, -Pro) backbone CMAP potential has been refined against exptl. soln. NMR data for weakly structured peptides, resulting in a rebalancing of the energies of the α-helix and extended regions of the Ramachandran map, correcting the α-helical bias of CHARMM22/CMAP. The Gly and Pro CMAPs have been refitted to more accurate quantum-mech. energy surfaces. Side-chain torsion parameters have been optimized by fitting to backbone-dependent quantum-mech. energy surfaces, followed by addnl. empirical optimization targeting NMR scalar couplings for unfolded proteins. A comprehensive validation of the revised force field was then performed against a collection of exptl. data: (i) comparison of simulations of eight proteins in their crystal environments with crystal structures; (ii) comparison with backbone scalar couplings for weakly structured peptides; (iii) comparison with NMR residual dipolar couplings and scalar couplings for both backbone and side-chains in folded proteins; (iv) equil. folding of mini-proteins. The results indicate that the revised CHARMM 36 parameters represent an improved model for modeling and simulation studies of proteins, including studies of protein folding, assembly, and functionally relevant conformational changes.
- 26Mallajosyula, S. S.; Guvench, O.; Hatcher, E.; Mackerell, A. D., Jr. CHARMM Additive All-Atom Force Field for Phosphate and Sulfate Linked to Carbohydrates. J. Chem. Theory Comput. 2012, 8, 759– 776, DOI: 10.1021/ct200792vGoogle Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs1Oju7zP&md5=8350bcdedba6a5e80675f9cae77dfc7eCHARMM Additive All-Atom Force Field for Phosphate and Sulfate Linked to CarbohydratesMallajosyula, Sairam S.; Guvench, Olgun; Hatcher, Elizabeth; MacKerell, Alexander D.Journal of Chemical Theory and Computation (2012), 8 (2), 759-776CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Presented is an extension of the CHARMM additive all-atom carbohydrate force field to enable the modeling of phosphate and sulfate linked to carbohydrates. The parameters are developed in a hierarchical fashion using model compds. contg. the key atoms in the full carbohydrates. Target data for parameter optimization included full two-dimensional energy surfaces defined by the glycosidic dihedral angle pairs in the phosphate/sulfate model compd. analogs of hexopyranose monosaccharide phosphates and sulfates, as detd. by quantum mech. (QM) MP2/cc-pVTZ single point energies on MP2/6-31+G(d) optimized structures. To achieve balanced, transferable dihedral parameters for the dihedral angles, surfaces for all possible anomeric and conformational states were included during the parametrization process. To model physiol. relevant systems, both the mono- and dianionic charged states were studied for the phosphates. This resulted in over 7000 MP2/cc-pVTZ//MP2/6-31G+(d) model compd. conformational energies which, supplemented with QM geometries, were the main target data for the parametrization. Parameters were validated against crystals of relevant monosaccharide derivs. obtained from the Cambridge Structural Database (CSD) and larger systems, inositol-(tri/tetra/penta) phosphates noncovalently bound to the pleckstrin homol. (PH) domain and oligomeric chondroitin sulfate in soln. and in complex with cathepsin K protein.
- 27Essmann, U.; Perera, L.; Berkowitz, M. L. A smooth particle mesh Ewald method. J. Chem. Phys. 1995, 103, 8577, DOI: 10.1063/1.470117Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXptlehtrw%253D&md5=092a679dd3bee08da28df41e302383a7A smooth particle mesh Ewald methodEssmann, Ulrich; Perera, Lalith; Berkowitz, Max L.; Darden, Tom; Lee, Hsing; Pedersen, Lee G.Journal of Chemical Physics (1995), 103 (19), 8577-93CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)The previously developed particle mesh Ewald method is reformulated in terms of efficient B-spline interpolation of the structure factors. This reformulation allows a natural extension of the method to potentials of the form 1/rp with p ≥ 1. Furthermore, efficient calcn. of the virial tensor follows. Use of B-splines in the place of Lagrange interpolation leads to analytic gradients as well as a significant improvement in the accuracy. The authors demonstrate that arbitrary accuracy can be achieved, independent of system size N, at a cost that scales as N log(N). For biomol. systems with many thousands of atoms and this method permits the use of Ewald summation at a computational cost comparable to that of a simple truncation method of 10 Å or less.
- 28Jurrus, E.; Engel, D.; Star, K.; Monson, K.; Brandi, J.; Felberg, L. E.; Brookes, D. H.; Wilson, L.; Chen, J.; Liles, K.; Chun, M.; Li, P.; Gohara, D. W.; Dolinsky, T.; Konecny, R.; Koes, D. R.; Nielsen, J. E.; Head-Gordon, T.; Geng, W.; Krasny, R.; Wei, G. W.; Holst, M. J.; McCammon, J. A.; Baker, N. A. Improvements to the APBS biomolecular solvation software suite. Protein Sci. 2018, 27, 112– 128, DOI: 10.1002/pro.3280Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhslSkt7vI&md5=99651d125e38f4a85d453fecf0f71652Improvements to the APBS biomolecular solvation software suiteJurrus, Elizabeth; Engel, Dave; Star, Keith; Monson, Kyle; Brandi, Juan; Felberg, Lisa E.; Brookes, David H.; Wilson, Leighton; Chen, Jiahui; Liles, Karina; Chun, Minju; Li, Peter; Gohara, David W.; Dolinsky, Todd; Konecny, Robert; Koes, David R.; Nielsen, Jens Erik; Head-Gordon, Teresa; Geng, Weihua; Krasny, Robert; Wei, Guo-Wei; Holst, Michael J.; McCammon, J. Andrew; Baker, Nathan A.Protein Science (2018), 27 (1), 112-128CODEN: PRCIEI; ISSN:1469-896X. (Wiley-Blackwell)The Adaptive Poisson-Boltzmann Solver (APBS) software was developed to solve the equations of continuum electrostatics for large biomol. assemblages that have provided impact in the study of a broad range of chem., biol., and biomedical applications. APBS addresses the three key technol. challenges for understanding solvation and electrostatics in biomedical applications: accurate and efficient models for biomol. solvation and electrostatics, robust and scalable software for applying those theories to biomol. systems, and mechanisms for sharing and analyzing biomol. electrostatics data in the scientific community. To address new research applications and advancing computational capabilities, we have continually updated APBS and its suite of accompanying software since its release in 2001. In this article, we discuss the models and capabilities that have recently been implemented within the APBS software package including a Poisson-Boltzmann anal. and a semi-anal. solver, an optimized boundary element solver, a geometry-based geometric flow solvation model, a graph theory-based algorithm for detg. pKa values, and an improved web-based visualization tool for viewing electrostatics.
- 29Dolinsky, T. J.; Czodrowski, P.; Li, H.; Nielsen, J. E.; Jensen, J. H.; Klebe, G.; Baker, N. A. PDB2PQR: expanding and upgrading automated preparation of biomolecular structures for molecular simulations. Nucleic Acids Res. 2007, 35, W522– W525, DOI: 10.1093/nar/gkm276Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2svhsVGltQ%253D%253D&md5=18883d2f9a6f30dba143a4a2ca80b0d8PDB2PQR: expanding and upgrading automated preparation of biomolecular structures for molecular simulationsDolinsky Todd J; Czodrowski Paul; Li Hui; Nielsen Jens E; Jensen Jan H; Klebe Gerhard; Baker Nathan ANucleic acids research (2007), 35 (Web Server issue), W522-5 ISSN:.Real-world observable physical and chemical characteristics are increasingly being calculated from the 3D structures of biomolecules. Methods for calculating pK(a) values, binding constants of ligands, and changes in protein stability are readily available, but often the limiting step in computational biology is the conversion of PDB structures into formats ready for use with biomolecular simulation software. The continued sophistication and integration of biomolecular simulation methods for systems- and genome-wide studies requires a fast, robust, physically realistic and standardized protocol for preparing macromolecular structures for biophysical algorithms. As described previously, the PDB2PQR web server addresses this need for electrostatic field calculations (Dolinsky et al., Nucleic Acids Research, 32, W665-W667, 2004). Here we report the significantly expanded PDB2PQR that includes the following features: robust standalone command line support, improved pK(a) estimation via the PROPKA framework, ligand parameterization via PEOE_PB charge methodology, expanded set of force fields and easily incorporated user-defined parameters via XML input files, and improvement of atom addition and optimization code. These features are available through a new web interface (http://pdb2pqr.sourceforge.net/), which offers users a wide range of options for PDB file conversion, modification and parameterization.
- 30Johnson, J. M.; Sanford, B. L.; Strom, A. M.; Tadayon, S. N.; Lehman, B. P.; Zirbes, A. M.; Bhattacharyya, S.; Musier-Forsyth, K.; Hati, S. Multiple pathways promote dynamical coupling between catalytic domains in Escherichia coli prolyl-tRNA synthetase. Biochemistry 2013, 52, 4399– 4412, DOI: 10.1021/bi400079hGoogle Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXosFygsL8%253D&md5=e4130c42e5a6c34a1f9f15135222f6ceMultiple Pathways Promote Dynamical Coupling between Catalytic Domains in Escherichia coli Prolyl-tRNA SynthetaseJohnson, James M.; Sanford, Brianne L.; Strom, Alexander M.; Tadayon, Stephanie N.; Lehman, Brent P.; Zirbes, Arrianna M.; Bhattacharyya, Sudeep; Musier-Forsyth, Karin; Hati, SanchitaBiochemistry (2013), 52 (25), 4399-4412CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)Aminoacyl-tRNA synthetases are multidomain enzymes that catalyze covalent attachment of amino acids to their cognate tRNA. Cross-talk between functional domains is a prerequisite for this process. In this study, we investigate the mol. mechanism of site-to-site communication in Escherichia coli prolyl-tRNA synthetase (Ec ProRS). Earlier studies have demonstrated that evolutionarily conserved and/or co-evolved residues that are engaged in correlated motion are crit. for the propagation of functional conformational changes from one site to another in modular proteins. Here, mol. simulation and bioinformatics-based anal. were performed to identify dynamically coupled and evolutionarily constrained residues that form contiguous pathways of residue-residue interactions between the aminoacylation and editing domains of Ec ProRS. The results of this study suggest that multiple pathways exist between these two domains to maintain the dynamic coupling essential for enzyme function. Moreover, residues in these interaction networks are generally highly conserved. Site-directed changes of on-pathway residues have a significant impact on enzyme function and dynamics, suggesting that any perturbation along these pathways disrupts the native residue-residue interactions that are required for effective communication between the two functional domains. Free energy anal. revealed that communication between residues within a pathway and cross-talk between pathways are important for coordinating functions of different domains of Ec ProRS for efficient catalysis.
- 31Verlet, L. Computer “experiments” on classic fluids. I. Thermodynamical properties of Lennard-Jones molecules. Phys. Rev. 1967, 159, 98– 103, DOI: 10.1103/PhysRev.159.98Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaF2sXks1Orsrc%253D&md5=4dbb891023a4169feaa95513ce8075a2Computer "experiments" on classical fluids. I. Thermodynamical properties of Lennard-Jones moleculesVerlet, LoupPhysical Review (1967), 159 (1), 98-103CODEN: PHRVAO; ISSN:0031-899X.The equation of motion of a system of 864 particles interacting through a Lennard-Jones potential was integrated for various values of the temp. and d., relative, generally, to a fluid state. The equil. properties agree with the corresponding properties of Ar. The equil. state of Ar can be described through a 2-body potential.
- 32Phillips, J. C.; Braun, R.; Wang, W.; Gumbart, J.; Tajkhorshid, E.; Villa, E.; Chipot, C.; Skeel, R. D.; Kalé, L.; Schulten, K. Scalable molecular dynamics with NAMD. J. Comput. Chem. 2005, 26, 1781– 1802, DOI: 10.1002/jcc.20289Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXht1SlsbbJ&md5=189051128443b547f4300a1b8fb0e034Scalable molecular dynamics with NAMDPhillips, James C.; Braun, Rosemary; Wang, Wei; Gumbart, James; Tajkhorshid, Emad; Villa, Elizabeth; Chipot, Christophe; Skeel, Robert D.; Kale, Laxmikant; Schulten, KlausJournal of Computational Chemistry (2005), 26 (16), 1781-1802CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)NAMD is a parallel mol. dynamics code designed for high-performance simulation of large biomol. systems. NAMD scales to hundreds of processors on high-end parallel platforms, as well as tens of processors on low-cost commodity clusters, and also runs on individual desktop and laptop computers. NAMD works with AMBER and CHARMM potential functions, parameters, and file formats. This article, directed to novices as well as experts, first introduces concepts and methods used in the NAMD program, describing the classical mol. dynamics force field, equations of motion, and integration methods along with the efficient electrostatics evaluation algorithms employed and temp. and pressure controls used. Features for steering the simulation across barriers and for calcg. both alchem. and conformational free energy differences are presented. The motivations for and a roadmap to the internal design of NAMD, implemented in C++ and based on Charm++ parallel objects, are outlined. The factors affecting the serial and parallel performance of a simulation are discussed. Finally, typical NAMD use is illustrated with representative applications to a small, a medium, and a large biomol. system, highlighting particular features of NAMD, for example, the Tcl scripting language. The article also provides a list of the key features of NAMD and discusses the benefits of combining NAMD with the mol. graphics/sequence anal. software VMD and the grid computing/collab. software BioCoRE. NAMD is distributed free of charge with source code at www.ks.uiuc.edu.
- 33Geng, W.; Krasny, R. A treecode-accelerated boundary integral Poisson–Boltzmann solver for electrostatics of solvated biomolecules. J. Comput. Phys. 2013, 247, 62– 78, DOI: 10.1016/j.jcp.2013.03.056Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXosFyltbY%253D&md5=4959c5c5c74974ac8167c78a00042842A treecode-accelerated boundary integral Poisson-Boltzmann solver for electrostatics of solvated biomoleculesGeng, Weihua; Krasny, RobertJournal of Computational Physics (2013), 247 (), 62-78CODEN: JCTPAH; ISSN:0021-9991. (Elsevier Inc.)We present a treecode-accelerated boundary integral (TABI) solver for electrostatics of solvated biomols. described by the linear Poisson-Boltzmann equation. The method employs a well-conditioned boundary integral formulation for the electrostatic potential and its normal deriv. on the mol. surface. The surface is triangulated and the integral equations are discretized by centroid collocation. The linear system is solved by GMRES iteration and the matrix-vector product is carried out by a Cartesian treecode which reduces the cost from O(N2) to O(NlogN), where N is the no. of faces in the triangulation. The TABI solver is applied to compute the electrostatic solvation energy in two cases, the Kirkwood sphere and a solvated protein. We present the error, CPU time, and memory usage, and compare results for the Poisson-Boltzmann and Poisson equations. We show that the treecode approxn. error can be made smaller than the discretization error, and we compare two versions of the treecode, one with uniform clusters and one with non-uniform clusters adapted to the mol. surface. For the protein test case, we compare TABI results with those obtained using the grid-based APBS code, and we also present parallel TABI simulations using up to eight processors. We find that the TABI solver exhibits good serial and parallel performance combined with relatively simple implementation, efficient memory usage, and geometric adaptability.
- 34Lan, J.; Ge, J.; Yu, J.; Shan, S.; Zhou, H.; Fan, S.; Zhang, Q.; Shi, X.; Wang, Q.; Zhang, L.; Wang, X. Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature 2020, 581, 215– 220, DOI: 10.1038/s41586-020-2180-5Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXoslOqtL8%253D&md5=279c60143e8e5eb505457e0778baa8efStructure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptorLan, Jun; Ge, Jiwan; Yu, Jinfang; Shan, Sisi; Zhou, Huan; Fan, Shilong; Zhang, Qi; Shi, Xuanling; Wang, Qisheng; Zhang, Linqi; Wang, XinquanNature (London, United Kingdom) (2020), 581 (7807), 215-220CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Abstr.: A new and highly pathogenic coronavirus (severe acute respiratory syndrome coronavirus-2, SARS-CoV-2) caused an outbreak in Wuhan city, Hubei province, China, starting from Dec. 2019 that quickly spread nationwide and to other countries around the world1-3. Here, to better understand the initial step of infection at an at. level, we detd. the crystal structure of the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 bound to the cell receptor ACE2. The overall ACE2-binding mode of the SARS-CoV-2 RBD is nearly identical to that of the SARS-CoV RBD, which also uses ACE2 as the cell receptor4. Structural anal. identified residues in the SARS-CoV-2 RBD that are essential for ACE2 binding, the majority of which either are highly conserved or share similar side chain properties with those in the SARS-CoV RBD. Such similarity in structure and sequence strongly indicate convergent evolution between the SARS-CoV-2 and SARS-CoV RBDs for improved binding to ACE2, although SARS-CoV-2 does not cluster within SARS and SARS-related coronaviruses1-3,5. The epitopes of two SARS-CoV antibodies that target the RBD are also analyzed for binding to the SARS-CoV-2 RBD, providing insights into the future identification of cross-reactive antibodies.
- 35Madeira, F.; Park, Y. M.; Lee, J.; Buso, N.; Gur, T.; Madhusoodanan, N.; Basutkar, P.; Tivey, A. R. N.; Potter, S. C.; Finn, R. D.; Lopez, R. The EMBL-EBI search and sequence analysis tools APIs in 2019. Nucleic Acids Res. 2019, 47, W636– W641, DOI: 10.1093/nar/gkz268Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXktVyitbY%253D&md5=ece6310cea25d80c352cbff7ec5d0661The EMBL-EBI search and sequence analysis tools APIs in 2019Madeira, Fabio; Park, Young Mi; Lee, Joon; Buso, Nicola; Gur, Tamer; Madhusoodanan, Nandana; Basutkar, Prasad; Tivey, Adrian R. N.; Potter, Simon C.; Finn, Robert D.; Lopez, RodrigoNucleic Acids Research (2019), 47 (W1), W636-W641CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)The EMBL-EBI provides free access to popular bioinformatics sequence anal. applications as well as to a full-featured text search engine with powerful cross-referencing and data retrieval capabilities. Access to these services is provided via user-friendly web interfaces and via established RESTful and SOAP Web Services APIs (https://www.ebi.ac.uk/seqdb/confluence/display/JDSAT/EMBL-EBI+Web+Services+APIs+-+Data+Retrieval). Both systems have been developed with the same core principles that allow them to integrate an ever-increasing vol. of biol. data, making them an integral part of many popular data resources provided at the EMBL-EBI. Here, we describe the latest improvements made to the frameworks which enhance the interconnectivity between public EMBL-EBI resources and ultimately enhance biol. data discoverability, accessibility, interoperability and reusability.
Cited By
This article is cited by 109 publications.
- Aurelia Magdalena Pisoschi, Florin Iordache, Loredana Stanca, Iuliana Gajaila, Oana Margarita Ghimpeteanu, Ovidiu Ionut Geicu, Liviu Bilteanu, Andreea Iren Serban. Antioxidant, Anti-inflammatory, and Immunomodulatory Roles of Nonvitamin Antioxidants in Anti-SARS-CoV-2 Therapy. Journal of Medicinal Chemistry 2022, 65
(19)
, 12562-12593. https://doi.org/10.1021/acs.jmedchem.2c01134
- Kaifu Gao, Rui Wang, Jiahui Chen, Limei Cheng, Jaclyn Frishcosy, Yuta Huzumi, Yuchi Qiu, Tom Schluckbier, Xiaoqi Wei, Guo-Wei Wei. Methodology-Centered Review of Molecular Modeling, Simulation, and Prediction of SARS-CoV-2. Chemical Reviews 2022, 122
(13)
, 11287-11368. https://doi.org/10.1021/acs.chemrev.1c00965
- Manoj K. Kolel-Veetil, Aayush Kant, Vivek B. Shenoy, Markus J. Buehler. SARS-CoV-2 Infection−Of Music and Mechanics of Its Spikes! A Perspective. ACS Nano 2022, 16
(5)
, 6949-6955. https://doi.org/10.1021/acsnano.1c11491
- Carl J. Fossum, Bethany F. Laatsch, Harrison R. Lowater, Alex W. Narkiewicz-Jodko, Leo Lonzarich, Sanchita Hati, Sudeep Bhattacharyya. Pre-Existing Oxidative Stress Creates a Docking-Ready Conformation of the SARS-CoV-2 Receptor-Binding Domain. ACS Bio & Med Chem Au 2022, 2
(1)
, 84-93. https://doi.org/10.1021/acsbiomedchemau.1c00040
- Maryam Ghasemitarei, Angela Privat-Maldonado, Maksudbek Yusupov, Shadi Rahnama, Annemie Bogaerts, Mohammad Reza Ejtehadi. Effect of Cysteine Oxidation in SARS-CoV-2 Receptor-Binding Domain on Its Interaction with Two Cell Receptors: Insights from Atomistic Simulations. Journal of Chemical Information and Modeling 2022, 62
(1)
, 129-141. https://doi.org/10.1021/acs.jcim.1c00853
- Eric J Niesor, Guy Boivin, Eric Rhéaume, Rong Shi, Véronique Lavoie, Nathalie Goyette, Marie-Eve Picard, Anne Perez, Fouzia Laghrissi-Thode, Jean-Claude Tardif. Inhibition of the 3CL Protease and SARS-CoV-2 Replication by Dalcetrapib. ACS Omega 2021, 6
(25)
, 16584-16591. https://doi.org/10.1021/acsomega.1c01797
- Christoph Gstöttner, Tao Zhang, Anja Resemann, Sophia Ruben, Stuart Pengelley, Detlev Suckau, Tim Welsink, Manfred Wuhrer, Elena Domínguez-Vega. Structural and Functional Characterization of SARS-CoV-2 RBD Domains Produced in Mammalian Cells. Analytical Chemistry 2021, 93
(17)
, 6839-6847. https://doi.org/10.1021/acs.analchem.1c00893
- Yi Y. Zuo, William E. Uspal, Tao Wei. Airborne Transmission of COVID-19: Aerosol Dispersion, Lung Deposition, and Virus-Receptor Interactions. ACS Nano 2020, 14
(12)
, 16502-16524. https://doi.org/10.1021/acsnano.0c08484
- Aftab Ahmad, Muhammad Afzaal, Farhan Saeed, Shinawar Waseem Ali, Ali Imran, Syed Yaseen Raza Zaidi, Muhammad Awais Saleem, Muzzamal Hussain, Entessar Al Jbawi. A comprehensive review of the therapeutic potential of citrus bioflavonoid hesperidin against lifestyle-related disorders. Cogent Food & Agriculture 2023, 9
(1)
https://doi.org/10.1080/23311932.2023.2226427
- M. Abou-Hamdan, R. Saleh, S. Mani, P. Dournaud, M. Metifiot, M. L. Blondot, M. L. Andreola, F. Abdel-sater, M. De Reggi, P. Gressens, M. Laforge. Potential antiviral effects of pantethine against SARS-CoV-2. Scientific Reports 2023, 13
(1)
https://doi.org/10.1038/s41598-023-29245-0
- Roger J. Grand. SARS-CoV-2 and the DNA damage response. Journal of General Virology 2023, 104
(11)
https://doi.org/10.1099/jgv.0.001918
- Jennyfer Bejoy, Charlye I. Williams, Hattie J. Cole, Shavaiz Manzoor, Parsa Davoodi, Jacqueline I. Battaile, Arjun Kaushik, Sofia I. Nikolaienko, Tinatin I. Brelidze, Sergiy G. Gychka, Yuichiro J. Suzuki. Effects of spike proteins on angiotensin converting enzyme 2 (ACE2). Archives of Biochemistry and Biophysics 2023, 748 , 109769. https://doi.org/10.1016/j.abb.2023.109769
- Jonathan C. Luque-Ceballos, Penélope Rodríguez-Zamora, Juan Carlos López-Olivos, Ignacio L. Garzón. Revisiting the scavenging activity of glutathione: Free radicals diversity and reaction mechanisms. Computational and Theoretical Chemistry 2023, 1227 , 114227. https://doi.org/10.1016/j.comptc.2023.114227
- Audry Peredo-Lovillo, Haydee Eliza Romero-Luna, Naida Juárez-Trujillo, Maribel Jiménez-Fernández. Antimicrobial efficiency of chlorine dioxide and its potential use as anti-SARS-CoV-2 agent: mechanisms of action and interactions with gut microbiota. Journal of Applied Microbiology 2023, 134
(7)
https://doi.org/10.1093/jambio/lxad133
- Yifan Zhao, Xiaorong Han, Cheng Li, Yucheng Liu, Jiayu Cheng, Binay Kumar Adhikari, Yonggang Wang. COVID-19 and the cardiovascular system: a study of pathophysiology and interpopulation variability. Frontiers in Microbiology 2023, 14 https://doi.org/10.3389/fmicb.2023.1213111
- Bushra R Hade, Raheem Al-Mammori, Talat Tariq Khalil. ESTIMATION OF OXIDATIVE STRESS AND ANTIOXIDANT CAPACITY IN COVID-19 PATIENTS, CURED PATIENTS AND PERSONS TAKING VACCINE. Military Medical Science Letters 2023, 92
(2)
, 122-127. https://doi.org/10.31482/mmsl.2022.031
- Anna S. Tolmacheva, Margarita K. Onvumere, Sergey E. Sedykh, Anna M. Timofeeva, Georgy A. Nevinsky. Catalase Activity of IgGs of Patients Infected with SARS-CoV-2. International Journal of Molecular Sciences 2023, 24
(12)
, 10081. https://doi.org/10.3390/ijms241210081
- Laurent Schwartz, Manuel Aparicio-Alonso, Marc Henry, Miroslav Radman, Romain Attal, Ashraf Bakkar. Toxicity of the spike protein of COVID-19 is a redox shift phenomenon: A novel therapeutic approach. Free Radical Biology and Medicine 2023, 118 https://doi.org/10.1016/j.freeradbiomed.2023.05.034
- Iñaki Milton-Laskibar, Jenifer Trepiana, María Teresa Macarulla, Saioa Gómez-Zorita, Laura Arellano-García, Alfredo Fernández-Quintela, María P. Portillo. Potential usefulness of Mediterranean diet polyphenols against COVID-19-induced inflammation: a review of the current knowledge. Journal of Physiology and Biochemistry 2023, 79
(2)
, 371-382. https://doi.org/10.1007/s13105-022-00926-0
- Didem Barlak Keti, Sabahattin Muhtaroglu, Orhan Yildiz, Hatice Saraçoglu. Prognostic value of total thiol and D-dimer in patients hospitalized with COVID-19. Turkish Journal of Biochemistry 2023, 48
(2)
, 209-214. https://doi.org/10.1515/tjb-2022-0171
- Arun K. Singh. Nanostructured coatings based on metallic nanoparticles as viral entry inhibitor to combat COVID-19. Sustainable Materials and Technologies 2023, 35 , e00544. https://doi.org/10.1016/j.susmat.2022.e00544
- İrfan BİNİCİ, Hamit Hakan ALP, Zübeyir HUYUT, Esra GÜRBÜZ, Hülya GÜNBATAR, Şükrü AKMEŞE, Mustafa Kasım KARAHOCAGİL. The Status of Antioxidants and Oxidative Damage in Patients with COVID-19. Ahi Evran Medical Journal 2023, https://doi.org/10.46332/aemj.1152479
- Teruo Kusano, Tomoko Nishino, Ken Okamoto, Russ Hille, Takeshi Nishino. The mechanism and significance of the conversion of xanthine dehydrogenase to xanthine oxidase in mammalian secretory gland cells. Redox Biology 2023, 59 , 102573. https://doi.org/10.1016/j.redox.2022.102573
- Annamaria Vernone, Loredana Bergandi, Simone Pernice, Gianpiero Pescarmona, Francesca Silvagno. How the Competition for Cysteine May Promote Infection of SARS-CoV-2 by Triggering Oxidative Stress. Antioxidants 2023, 12
(2)
, 483. https://doi.org/10.3390/antiox12020483
- Alessandra Fraternale, Marta De Angelis, Riccardo De Santis, Donatella Amatore, Sofia Masini, Francesca Monittola, Michele Menotta, Federica Biancucci, Francesca Bartoccini, Michele Retini, Valentina Fiori, Raoul Fioravanti, Fabio Magurano, Laura Chiarantini, Florigio Lista, Giovanni Piersanti, Anna T. Palamara, Lucia Nencioni, Mauro Magnani, Rita Crinelli. Targeting
SARS‐CoV
‐2 by synthetic dual‐acting thiol compounds that inhibit Spike/
ACE2
interaction and viral protein production. The FASEB Journal 2023, 37
(2)
https://doi.org/10.1096/fj.202201157RR
- George J. Dugbartey, Karl K. Alornyo, Vincent Boima, Sampson Antwi, Alp Sener. Hydrogen Sulfide as a Potential Therapy for COVID-19-Associated Nephropathy. 2023, 119-142. https://doi.org/10.1007/978-3-031-44041-0_6
- Djurdja Jerotic, Jovan Ranin, Zoran Bukumiric, Tatjana Djukic, Vesna Coric, Ana Savic-Radojevic, Nevena Todorovic, Milika Asanin, Marko Ercegovac, Ivana Milosevic, Marija Pljesa-Ercegovac, Goran Stevanovic, Marija Matic, Tatjana Simic. SOD2
rs4880 and
GPX1
rs1050450 polymorphisms do not confer risk of COVID-19, but influence inflammation or coagulation parameters in Serbian cohort. Redox Report 2022, 27
(1)
, 85-91. https://doi.org/10.1080/13510002.2022.2057707
- Volga Kojasoy, Dean J. Tantillo. Impacts of noncovalent interactions involving sulfur atoms on protein stability, structure, folding, and bioactivity. Organic & Biomolecular Chemistry 2022, 21
(1)
, 11-23. https://doi.org/10.1039/D2OB01602H
- Betony Adams, Ilya Sinayskiy, Rienk van Grondelle, Francesco Petruccione. Quantum tunnelling in the context of SARS-CoV-2 infection. Scientific Reports 2022, 12
(1)
https://doi.org/10.1038/s41598-022-21321-1
- Michal Kluknavsky, Andrea Micurova, Martina Cebova, Ezgi Şaman, Sona Cacanyiova, Iveta Bernatova. MLN-4760 Induces Oxidative Stress without Blood Pressure and Behavioural Alterations in SHRs: Roles of Nfe2l2 Gene, Nitric Oxide and Hydrogen Sulfide. Antioxidants 2022, 11
(12)
, 2385. https://doi.org/10.3390/antiox11122385
- Sibei Lei, Xiaohua Chen, Jieping Wu, Xingmei Duan, Ke Men. Small molecules in the treatment of COVID-19. Signal Transduction and Targeted Therapy 2022, 7
(1)
https://doi.org/10.1038/s41392-022-01249-8
- Vineeta Chattree, Kamana Singh, Kanishk Singh, Aayush Goel, Amritaparna Maity, Asif Lone. A comprehensive review on modulation of
SIRT1
signaling pathways in the immune system of
COVID
‐19 patients by phytotherapeutic melatonin and epigallocatechin‐3‐gallate. Journal of Food Biochemistry 2022, 46
(12)
https://doi.org/10.1111/jfbc.14259
- Lorenzo Mancini, Marco Paolantoni, Aurélie Schoubben, Maurizio Ricci. Development of spray-dried N-acetylcysteine dry powder for inhalation. International Journal of Pharmaceutics 2022, 13 , 122550. https://doi.org/10.1016/j.ijpharm.2022.122550
- Tarik Acar, Birsen Ertekin, Mehmet Yortanli. Value of thiol and ischemia modified albumin (IMA) in predicting mortality in serious COVID-19 pneumonia. Heliyon 2022, 8
(12)
, e12514. https://doi.org/10.1016/j.heliyon.2022.e12514
- Mikhail V. Osikov, Vladimir N. Antonov, Semen O. Zotov, Galina L. Ignatova. The role of redox status in platelet dysfunction in severe COVID-19-associated pneumonia. HERALD of North-Western State Medical University named after I.I. Mechnikov 2022, 14
(3)
, 69-78. https://doi.org/10.17816/mechnikov109076
- Jaber Dehghani, Ali Movafeghi, Elodie Mathieu-Rivet, Narimane Mati-Baouche, Sébastien Calbo, Patrice Lerouge, Muriel Bardor. Microalgae as an Efficient Vehicle for the Production and Targeted Delivery of Therapeutic Glycoproteins against SARS-CoV-2 Variants. Marine Drugs 2022, 20
(11)
, 657. https://doi.org/10.3390/md20110657
- Andrea Bianco, Stefano Conte, Domenica Francesca Mariniello, Valentino Allocca, Maria Gabriella Matera, Vito D’Agnano, Luigi Lanata, Mario Cazzola, Fabio Perrotta. Mucolytic and Antioxidant Properties of Carbocysteine as a Strategy in COVID-19 Therapy. Life 2022, 12
(11)
, 1824. https://doi.org/10.3390/life12111824
- Carlos A. Labarrere, Ghassan S. Kassab. Glutathione deficiency in the pathogenesis of SARS-CoV-2 infection and its effects upon the host immune response in severe COVID-19 disease. Frontiers in Microbiology 2022, 13 https://doi.org/10.3389/fmicb.2022.979719
- Tanya Kadiyska, Ivan Tourtourikov, Kristiyan Dabchev, Radostina Cherneva, Nikolay Stoynev, Radka Hadjiolova, Vanyo Mitev, Demetrios Spandidos, Maria Adamaki, Vassilis Zoumpourlis. Role of endothelial dysfunction in the severity of COVID‑19 infection (Review). Molecular Medicine Reports 2022, 26
(5)
https://doi.org/10.3892/mmr.2022.12867
- Sebastiano La Maestra, Silvano Garibaldi, Roumen Balansky, Francesco D’Agostini, Rosanna T. Micale, Silvio De Flora. Inhibition of the Cell Uptake of Delta and Omicron SARS-CoV-2 Pseudoviruses by N-Acetylcysteine Irrespective of the Oxidoreductive Environment. Cells 2022, 11
(20)
, 3313. https://doi.org/10.3390/cells11203313
- Alec J. Wozney, Macey A. Smith, Mobeen Abdrabbo, Cole M. Birch, Kelsey A. Cicigoi, Connor C. Dolan, Audrey E. L. Gerzema, Abby Hansen, Ethan J. Henseler, Ben LaBerge, Caterra M. Leavens, Christine N. Le, Allison C. Lindquist, Rikaela K. Ludwig, Maggie G. O’Reilly, Jacob H. Reynolds, Brandon A. Sherman, Hunter W. Sillman, Michael A. Smith, Marissa J. Snortheim, Levi M. Svaren, Emily C. Vanderpas, Aidan Voon, Miles J. Wackett, Moriah M. Weiss, Sanchita Hati, Sudeep Bhattacharyya. Evolution of Stronger SARS-CoV-2 Variants as Revealed Through the Lens of Molecular Dynamics Simulations. The Protein Journal 2022, 41
(4-5)
, 444-456. https://doi.org/10.1007/s10930-022-10065-6
- Mahnaz D. Gholami, Kristyan Guppy-Coles, Serena Nihal, Daman Langguth, Prashant Sonar, Godwin A. Ayoko, Chamindie Punyadeera, Emad L. Izake. A paper-based optical sensor for the screening of viruses through the cysteine residues of their surface proteins: A proof of concept on the detection of coronavirus infection. Talanta 2022, 248 , 123630. https://doi.org/10.1016/j.talanta.2022.123630
- Christina Papanikolaou, Vasiliki Rapti, Dimitris Stellas, Dimitra Stefanou, Konstantinos Syrigos, George Pavlakis, Vassilis Souliotis. Delineating the SARS-CoV-2 Induced Interplay between the Host Immune System and the DNA Damage Response Network. Vaccines 2022, 10
(10)
, 1764. https://doi.org/10.3390/vaccines10101764
- Kritika Khanna, Wilfred W. Raymond, Jing Jin, Annabelle R. Charbit, Irina Gitlin, Monica Tang, Adam D. Werts, Edward G. Barrett, Jason M. Cox, Sharla M. Birch, Rachel Martinelli, Hannah S. Sperber, Sergej Franz, Thomas Duff, Markus Hoffmann, Anne Marie Healy, Stefan Oscarson, Stefan Pöhlmann, Satish K. Pillai, Graham Simmons, John V. Fahy. Exploring antiviral and anti-inflammatory effects of thiol drugs in COVID-19. American Journal of Physiology-Lung Cellular and Molecular Physiology 2022, 323
(3)
, L372-L389. https://doi.org/10.1152/ajplung.00136.2022
- Biju Bhargavan, Georgette D. Kanmogne. SARS-CoV-2 Spike Proteins and Cell–Cell Communication Inhibits TFPI and Induces Thrombogenic Factors in Human Lung Microvascular Endothelial Cells and Neutrophils: Implications for COVID-19 Coagulopathy Pathogenesis. International Journal of Molecular Sciences 2022, 23
(18)
, 10436. https://doi.org/10.3390/ijms231810436
- Sergei Onikienko, Maxim Vinokurov, Marina Yurinskaya, Alexander Zemlyanoi, Sergei Abkin, Elvira Shaykhutdinova, Victor Palikov, Alexander Ivanov, Olga Smirnova, Irina Fedyakina, Natalia Bychkova, Olga Zatsepina, David Garbuz, Michael Evgen’ev. The Effects of H2S and Recombinant Human Hsp70 on Inflammation Induced by SARS and Other Agents In Vitro and In Vivo. Biomedicines 2022, 10
(9)
, 2155. https://doi.org/10.3390/biomedicines10092155
- Jess Thoene, Robert F. Gavin, Aaron Towne, Lauren Wattay, Maria Grazia Ferrari, Jennifer Navarrete, Ranajit Pal. In vitro activity of cysteamine against SARS-CoV-2 variants. Molecular Genetics and Metabolism 2022, 137
(1-2)
, 192-200. https://doi.org/10.1016/j.ymgme.2022.08.009
- Moua Yang. Redox stress in COVID-19: Implications for hematologic disorders. Best Practice & Research Clinical Haematology 2022, 35
(3)
, 101373. https://doi.org/10.1016/j.beha.2022.101373
- Keisuke Minami, Maki Minakawa, Yasuhiro Uozumi. Preparation of Benzothiazoles and Heterocyclic Spiro Compounds Through Cu‐catalyzed S−S Bond Cleavage and C−S Bond Formation. Asian Journal of Organic Chemistry 2022, 11
(9)
https://doi.org/10.1002/ajoc.202200211
- Tobias Sinnberg, Christa Lichtensteiger, Katharina Hill-Mündel, Christian Leischner, Heike Niessner, Christian Busch, Olga Renner, Nina Wyss, Lukas Flatz, Ulrich M. Lauer, Ludwig E. Hoelzle, Donatus Nohr, Markus Burkard, Luigi Marongiu, Sascha Venturelli. Vitamin C Deficiency in Blood Samples of COVID-19 Patients. Antioxidants 2022, 11
(8)
, 1580. https://doi.org/10.3390/antiox11081580
- Yutaro Yamaoka, Sundararaj Stanleyraj Jeremiah, Rikako Funabashi, Kei Miyakawa, Takeshi Morita, Yusaku Mihana, Hideaki Kato, Akihide Ryo. Characterization and Utilization of Disulfide-Bonded SARS-CoV-2 Receptor Binding Domain of Spike Protein Synthesized by Wheat Germ Cell-Free Production System. Viruses 2022, 14
(7)
, 1461. https://doi.org/10.3390/v14071461
- Jonathan Foo, Gregory Bellot, Shazib Pervaiz, Sylvie Alonso. Mitochondria-mediated oxidative stress during viral infection. Trends in Microbiology 2022, 30
(7)
, 679-692. https://doi.org/10.1016/j.tim.2021.12.011
- Francesco Galli, Giada Marcantonini, Daniela Giustarini, Maria Cristina Albertini, Anna Migni, Linda Zatini, Antimo Gioiello, Ranieri Rossi, Desirée Bartolini. How Aging and Oxidative Stress Influence the Cytopathic and Inflammatory Effects of SARS-CoV-2 Infection: The Role of Cellular Glutathione and Cysteine Metabolism. Antioxidants 2022, 11
(7)
, 1366. https://doi.org/10.3390/antiox11071366
- Igor Belenichev, Lyudmyla Kucherenko, Sergii Pavlov, Nina Bukhtiyarova, Olena Popazova, Natalia Derevianko, Ganna Nimenko. Therapy of post-COVID-19 syndrome: improving the efficiency and safety of basic metabolic drug treatment with tiazotic acid (thiotriazoline). Pharmacia 2022, 69
(2)
, 509-516. https://doi.org/10.3897/pharmacia.69.e82596
- Rajalakshmi Rajendran, Anjana Chathambath, Abdullah G. Al-Sehemi, Mehboobali Pannipara, Mazhuvancherry Kesavan Unnikrishnan, Lotfi Aleya, Roshni Pushpa Raghavan, Bijo Mathew. Critical role of nitric oxide in impeding COVID-19 transmission and prevention: a promising possibility. Environmental Science and Pollution Research 2022, 29
(26)
, 38657-38672. https://doi.org/10.1007/s11356-022-19148-4
- Magnus S. Bauer, Sophia Gruber, Adina Hausch, Priscila S. F. C. Gomes, Lukas F. Milles, Thomas Nicolaus, Leonard C. Schendel, Pilar López Navajas, Erik Procko, Daniel Lietha, Marcelo C. R. Melo, Rafael C. Bernardi, Hermann E. Gaub, Jan Lipfert. A tethered ligand assay to probe SARS-CoV-2:ACE2 interactions. Proceedings of the National Academy of Sciences 2022, 119
(14)
https://doi.org/10.1073/pnas.2114397119
- Tania Bakshi, David Pham, Raminderjeet Kaur, Bingyun Sun. Hidden Relationships between N-Glycosylation and Disulfide Bonds in Individual Proteins. International Journal of Molecular Sciences 2022, 23
(7)
, 3742. https://doi.org/10.3390/ijms23073742
- Joanna Perła-Kaján, Hieronim Jakubowski. COVID-19 and One-Carbon Metabolism. International Journal of Molecular Sciences 2022, 23
(8)
, 4181. https://doi.org/10.3390/ijms23084181
- Rauf Melekoğlu, Şeyma Yaşar, Nesibe Zeyveli Çelik, Halis Özdemir. Evaluation of dyslipidemia in preeclamptic pregnant women and determination of the predictive value of the hemato-lipid profile: A prospective, cross-sectional, case-control study. Journal of Turkish Society of Obstetric and Gynecology 2022, 19
(1)
, 7-20. https://doi.org/10.4274/tjod.galenos.2022.36744
- Tatjana Djukic, Goran Stevanovic, Vesna Coric, Zoran Bukumiric, Marija Pljesa-Ercegovac, Marija Matic, Djurdja Jerotic, Nevena Todorovic, Milika Asanin, Marko Ercegovac, Jovan Ranin, Ivana Milosevic, Ana Savic-Radojevic, Tatjana Simic. GSTO1, GSTO2 and ACE2 Polymorphisms Modify Susceptibility to Developing COVID-19. Journal of Personalized Medicine 2022, 12
(3)
, 458. https://doi.org/10.3390/jpm12030458
- Alexandros Nikolaidis, Ron Kramer, Sergej Ostojic. Nitric Oxide: The Missing Factor in COVID-19 Severity?. Medical Sciences 2022, 10
(1)
, 3. https://doi.org/10.3390/medsci10010003
- Marcos Martinez-Banaclocha. Interfering with the Reactive Cysteine Proteome in COVID-19. Current Medicinal Chemistry 2022, 29
(10)
, 1657-1663. https://doi.org/10.2174/0929867328666210623142811
- Mana Murae, Yoshimi Shimizu, Yuichiro Yamamoto, Asuka Kobayashi, Masumi Houri, Tetsuya Inoue, Takuya Irie, Ryutaro Gemba, Yosuke Kondo, Yoshio Nakano, Satoru Miyazaki, Daisuke Yamada, Akiyoshi Saitoh, Isao Ishii, Taishi Onodera, Yoshimasa Takahashi, Takaji Wakita, Masayoshi Fukasawa, Kohji Noguchi. The function of SARS-CoV-2 spike protein is impaired by disulfide-bond disruption with mutation at cysteine-488 and by thiol-reactive N-acetyl-cysteine and glutathione. Biochemical and Biophysical Research Communications 2022, 597 , 30-36. https://doi.org/10.1016/j.bbrc.2022.01.106
- George J. Dugbartey, Karl K. Alornyo, Bright O. Ohene, Vincent Boima, Sampson Antwi, Alp Sener. Renal consequences of the novel coronavirus disease 2019 (COVID-19) and hydrogen sulfide as a potential therapy. Nitric Oxide 2022, 120 , 16-25. https://doi.org/10.1016/j.niox.2022.01.002
- Chiu Ling Ong, Thorsten Heidelberg, Joon Ching Juan, Nader Ghaffari Khaligh. Metal-free and green synthesis of a series of new bis(2-alkylsulfanyl-[1,3,4]thiadiazolyl)-5,5′-disulfides and 2,2′-Dibenzothiazyl disulfide via oxidative self-coupling using hydrogen peroxide. Polyhedron 2022, 213 , 115610. https://doi.org/10.1016/j.poly.2021.115610
- Andrey M. Grishin, Nataliya V. Dolgova, Shelby Landreth, Olivier Fisette, Ingrid J. Pickering, Graham N. George, Darryl Falzarano, Miroslaw Cygler. Disulfide Bonds Play a Critical Role in the Structure and Function of the Receptor-binding Domain of the SARS-CoV-2 Spike Antigen. Journal of Molecular Biology 2022, 434
(2)
, 167357. https://doi.org/10.1016/j.jmb.2021.167357
- Mahnaz D. Gholami, Kristyan Guppy-Coles, Serena Nihal, Daman Langguth, Prashant Sonar, Godwin A. Ayoko, Chamindie Punyadeera, Emad Louis Izake. A Paper-Based Optical Sensor for the Screening of Viruses Through the Cysteine Residues of Their Surface Proteins: The Detection of Coronavirus Infection. SSRN Electronic Journal 2022, 13 https://doi.org/10.2139/ssrn.4057071
- Benjamin L. Springstein, Padraig Deighan, Grzegorz J. Grabe, Ann Hochschild, . A Bacterial Cell-Based Assay To Study SARS-CoV-2 Protein-Protein Interactions. mBio 2021, 12
(6)
https://doi.org/10.1128/mBio.02936-21
- Kamyar Khoshnevisan, Hassan Maleki, Hadi Baharifar. Nanobiocide Based-Silver Nanomaterials Upon Coronaviruses: Approaches for Preventing Viral Infections. Nanoscale Research Letters 2021, 16
(1)
https://doi.org/10.1186/s11671-021-03558-3
- Jorge R. Miranda-Massari, Alondra P. Toro, Doris Loh, Jose R. Rodriguez, Raul Morales Borges, Victor Marcial-Vega, Jose Olalde, Miguel J. Berdiel, Neil H. Riordan, Juan Manuel Martinez, Armando Gil, Michael J. Gonzalez. The Effects of Vitamin C on the Multiple Pathophysiological Stages of COVID-19. Life 2021, 11
(12)
, 1341. https://doi.org/10.3390/life11121341
- Pawel Hikisz, Joanna Bernasinska-Slomczewska. Beneficial Properties of Bromelain. Nutrients 2021, 13
(12)
, 4313. https://doi.org/10.3390/nu13124313
- Andrew F. Cumpstey, Anna D. Clark, Jérôme Santolini, Alan A. Jackson, Martin Feelisch. COVID-19: A Redox Disease—What a Stress Pandemic Can Teach Us About Resilience and What We May Learn from the Reactive Species Interactome About Its Treatment. Antioxidants & Redox Signaling 2021, 35
(14)
, 1226-1268. https://doi.org/10.1089/ars.2021.0017
- Mobeen Abdrabbo, Cole M. Birch, Michael Brandt, Kelsey A. Cicigoi, Stephen J. Coffey, Connor C. Dolan, Hannah Dvorak, Ava C. Gehrke, Audrey E. L. Gerzema, Abby Hansen, Ethan J. Henseler, Alyssa C. Huelsbeck, Ben LaBerge, Caterra M. Leavens, Christine N. Le, Allison C. Lindquist, Rickaela K. Ludwig, Jacob H. Reynolds, Nathaniel J. Severson, Brandon A. Sherman, Hunter W. Sillman, Michael A. Smith, Macey A. Smith, Marissa J. Snortheim, Levi M. Svaren, Emily C. Vanderpas, Miles J. Wackett, Alec J. Wozney, Sudeep Bhattacharyya, Sanchita Hati. Vitamin D and
COVID
‐19: A review on the role of vitamin D in preventing and reducing the severity of
COVID
‐19 infection. Protein Science 2021, 30
(11)
, 2206-2220. https://doi.org/10.1002/pro.4190
- Yudum Tepeli Büyüksünetçi, Burak Ekrem Çitil, Utku Tapan, Ülkü Anık. Development and application of a SARS-CoV-2 colorimetric biosensor based on the peroxidase-mimic activity of γ-Fe2O3 nanoparticles. Microchimica Acta 2021, 188
(10)
https://doi.org/10.1007/s00604-021-04989-6
- Evgeny Vladimirovich Kryukov, Alexander Vladimirovich Ivanov, Vladimir Olegovich Karpov, Valery Vasil’evich Alexandrin, Alexander Mikhaylovich Dygai, Maria Petrovna Kruglova, Gennady Ivanovich Kostiuchenko, Sergei Petrovich Kazakov, Aslan Amirkhanovich Kubatiev, . Association of Low Molecular Weight Plasma Aminothiols with the Severity of Coronavirus Disease 2019. Oxidative Medicine and Cellular Longevity 2021, 2021 , 1-10. https://doi.org/10.1155/2021/9221693
- Paolo Bellavite. Reappraisal of Dietary Phytochemicals for Coronavirus Infection: Focus on Hesperidin and Quercetin. 2021https://doi.org/10.5772/intechopen.95529
- Jing Dai, Xu Teng, Sheng Jin, Yuming Wu, . The Antiviral Roles of Hydrogen Sulfide by Blocking the Interaction between SARS-CoV-2 and Its Potential Cell Surface Receptors. Oxidative Medicine and Cellular Longevity 2021, 2021 , 1-11. https://doi.org/10.1155/2021/7866992
- Ayşe Özlem Mete, Kübra Koçak, Ahmet Saracaloglu, Seniz Demiryürek, Özgür Altınbaş, Abdullah T. Demiryürek. Effects of antiviral drug therapy on dynamic thiol/disulphide homeostasis and nitric oxide levels in COVID-19 patients. European Journal of Pharmacology 2021, 907 , 174306. https://doi.org/10.1016/j.ejphar.2021.174306
- Günnur Güler, Helin Özdemir, Dilara Omar, Gül Akdoğan. Coronavirus disease 2019 (COVID-19): Biophysical and biochemical aspects of SARS-CoV-2 and general characteristics. Progress in Biophysics and Molecular Biology 2021, 164 , 3-18. https://doi.org/10.1016/j.pbiomolbio.2021.05.007
- Kubra Aykac, Yasemin Ozsurekci, Burcu Ceylan Cura Yayla, Sibel Lacinel Gurlevik, Pembe Derin Oygar, Nuriye Boduc Bolu, Medine Aysin Tasar, Fatma Sebnem Erdinc, Gulay Tuncer Ertem, Salim Neselioglu, Ozcan Erel, Ali Bülent Cengiz, Mehmet Ceyhan. Oxidant and antioxidant balance in patients with COVID‐19. Pediatric Pulmonology 2021, 56
(9)
, 2803-2810. https://doi.org/10.1002/ppul.25549
- D. T. Rejepov, A. A. Vodyashkin, A. V. Sergorodceva, Ya. M. Stanishevskiy. Biomedical Applications of Silver Nanoparticles (Review). Drug development & registration 2021, 10
(3)
, 176-187. https://doi.org/10.33380/2305-2066-2021-10-3-176-187
- Bindu D. Paul, Marian D. Lemle, Anthony L. Komaroff, Solomon H. Snyder. Redox imbalance links COVID-19 and myalgic encephalomyelitis/chronic fatigue syndrome. Proceedings of the National Academy of Sciences 2021, 118
(34)
https://doi.org/10.1073/pnas.2024358118
- Adriana Fodor, Brandusa Tiperciuc, Cezar Login, Olga H. Orasan, Andrada L. Lazar, Cristina Buchman, Patricia Hanghicel, Adela Sitar-Taut, Ramona Suharoschi, Romana Vulturar, Angela Cozma, . Endothelial Dysfunction, Inflammation, and Oxidative Stress in COVID-19—Mechanisms and Therapeutic Targets. Oxidative Medicine and Cellular Longevity 2021, 2021 , 1-15. https://doi.org/10.1155/2021/8671713
- A. I. Zinchenko, L. L. Birichevskaya, I. S. Kazlouski, A. B. Bulatovski. Possible response of molecular biotechnology to call SARS-CoV-2. Proceedings of the National Academy of Sciences of Belarus, Biological Series 2021, 66
(3)
, 357-369. https://doi.org/10.29235/1029-8940-2021-66-3-357-369
- Mario Cazzola, Paola Rogliani, Sundeep Santosh Salvi, Josuel Ora, Maria Gabriella Matera. Use of Thiols in the Treatment of COVID-19: Current Evidence. Lung 2021, 199
(4)
, 335-343. https://doi.org/10.1007/s00408-021-00465-3
- Luc Rochette, Steliana Ghibu. Mechanics Insights of Alpha-Lipoic Acid against Cardiovascular Diseases during COVID-19 Infection. International Journal of Molecular Sciences 2021, 22
(15)
, 7979. https://doi.org/10.3390/ijms22157979
- Zilan Zhu, Ziyi Zheng, Jian Liu. Comparison of COVID-19 and Lung Cancer via Reactive Oxygen Species Signaling. Frontiers in Oncology 2021, 11 https://doi.org/10.3389/fonc.2021.708263
- Leonardo Warzea Lima, Serenella Nardi, Veronica Santoro, Michela Schiavon. The Relevance of Plant-Derived Se Compounds to Human Health in the SARS-CoV-2 (COVID-19) Pandemic Era. Antioxidants 2021, 10
(7)
, 1031. https://doi.org/10.3390/antiox10071031
- Tijana Kosanovic, Dragan Sagic, Vladimir Djukic, Marija Pljesa-Ercegovac, Ana Savic-Radojevic, Zoran Bukumiric, Miodrag Lalosevic, Marjana Djordjevic, Vesna Coric, Tatjana Simic. Time Course of Redox Biomarkers in COVID-19 Pneumonia: Relation with Inflammatory, Multiorgan Impairment Biomarkers and CT Findings. Antioxidants 2021, 10
(7)
, 1126. https://doi.org/10.3390/antiox10071126
- Morgane Ducastel, Camille Chenevier-Gobeaux, Yassine Ballaa, Jean-François Meritet, Michel Brack, Nicolas Chapuis, Frédéric Pene, Nicolas Carlier, Tali-Anne Szwebel, Nicolas Roche, Benjamin Terrier, Didier Borderie. Oxidative Stress and Inflammatory Biomarkers for the Prediction of Severity and ICU Admission in Unselected Patients Hospitalized with COVID-19. International Journal of Molecular Sciences 2021, 22
(14)
, 7462. https://doi.org/10.3390/ijms22147462
- Saroj Khatiwada, Astha Subedi. A Mechanistic Link Between Selenium and Coronavirus Disease 2019 (COVID-19). Current Nutrition Reports 2021, 10
(2)
, 125-136. https://doi.org/10.1007/s13668-021-00354-4
- Mateja Manček‐Keber, Iva Hafner‐Bratkovič, Duško Lainšček, Mojca Benčina, Tea Govednik, Sara Orehek, Tjaša Plaper, Vid Jazbec, Valter Bergant, Vincent Grass, Andreas Pichlmair, Roman Jerala. Disruption of disulfides within RBD of SARS‐CoV‐2 spike protein prevents fusion and represents a target for viral entry inhibition by registered drugs. The FASEB Journal 2021, 35
(6)
https://doi.org/10.1096/fj.202100560R
- Manoj Kolel-Veetil, Ayusman Sen, Markus J. Buehler. Surface adhesion of viruses and bacteria: Defend only and/or vibrationally extinguish also?! A perspective. MRS Advances 2021, 6
(13)
, 355-361. https://doi.org/10.1557/s43580-021-00079-0
- Noemí Cárdenas-Rodríguez, Cindy Bandala, América Vanoye-Carlo, Iván Ignacio-Mejía, Saúl Gómez-Manzo, Estefani Yaquelin Hernández-Cruz, José Pedraza-Chaverri, Liliana Carmona-Aparicio, Beatriz Hernández-Ochoa. Use of Antioxidants for the Neuro-Therapeutic Management of COVID-19. Antioxidants 2021, 10
(6)
, 971. https://doi.org/10.3390/antiox10060971
- Emily M. Nakada, Rui Sun, Utako Fujii, James G. Martin. The Impact of Endoplasmic Reticulum-Associated Protein Modifications, Folding and Degradation on Lung Structure and Function. Frontiers in Physiology 2021, 12 https://doi.org/10.3389/fphys.2021.665622
- Daniela Giustarini, Annalisa Santucci, Desirée Bartolini, Francesco Galli, Ranieri Rossi. The age-dependent decline of the extracellular thiol-disulfide balance and its role in SARS-CoV-2 infection. Redox Biology 2021, 41 , 101902. https://doi.org/10.1016/j.redox.2021.101902
- Daniela Giustarini, Isabella Dalle-Donne, Graziano Colombo, Aldo Milzani, Annalisa Santucci, Ranieri Rossi. Protein thiolation index in microvolumes of plasma. Analytical Biochemistry 2021, 618 , 114125. https://doi.org/10.1016/j.ab.2021.114125
- Javed Akhter, Grégory Quéromès, Krishna Pillai, Vahan Kepenekian, Samina Badar, Ahmed H. Mekkawy, Emilie Frobert, Sarah J. Valle, David L. Morris. The Combination of Bromelain and Acetylcysteine (BromAc) Synergistically Inactivates SARS-CoV-2. Viruses 2021, 13
(3)
, 425. https://doi.org/10.3390/v13030425
- Jean Bousquet, Josep M. Anto, Wienczyslawa Czarlewski, Tari Haahtela, Susana C. Fonseca, Guido Iaccarino, Hubert Blain, Alain Vidal, Aziz Sheikh, Cezmi A. Akdis, Torsten Zuberbier, . Cabbage and fermented vegetables: From death rate heterogeneity in countries to candidates for mitigation strategies of severe COVID‐19. Allergy 2021, 76
(3)
, 735-750. https://doi.org/10.1111/all.14549
- Anna M. Fratta Pasini, Chiara Stranieri, Luciano Cominacini, Chiara Mozzini. Potential Role of Antioxidant and Anti-Inflammatory Therapies to Prevent Severe SARS-Cov-2 Complications. Antioxidants 2021, 10
(2)
, 272. https://doi.org/10.3390/antiox10020272
Abstract
Figure 1
Figure 1. Sequence alignment (generated by Clustal Omega (35)) between the receptor-binding domain of SARS-CoV and SARS-CoV-2 proteins. “*” represents the identical residues, “:” and “.” represent strongly and weakly similar residues, respectively, and gap represents dissimilar residues. The cysteine residues are highlighted in yellow.
Figure 2
Figure 2. Structures of protein complexes: (a) SARS-CoV···ACE2 and (b) SARS-CoV-2···ACE2. All of the disulfide bridges between cysteine residues are shown in green van der Waals (vdW) spheres and thiol groups in cyan licorice.
Figure 3
Figure 3. RMSDs, averaged over 1 ns MD simulation, of the protein complexes of ACE2 and SARS-CoV or SARS-CoV-2. Disulfide-containing proteins are referred to as oxidized with a shorthand notation of “ox”, while thiol variants are denoted with “red” notation.
Figure 4
Figure 4. Change in residue-level RMSFs of Cα atoms in the oxidized and reduced protein complexes: (a) SARS-CoV···ACE2 and (b) SARS-CoV-2···ACE2. The cysteine residues, for each protein system (labeled in Figure 2), are highlighted as vdW spheres. The backbone as well as the cysteine residues are color-coded, large fluctuations are shown in red, and small fluctuations are in blue. The medium-scale fluctuations are shown in green.
Figure 5
Figure 5. Comparison of the conformational change at the protein–protein interface in CoVox···ACE2ox (left) and CoVred···ACE2red (right). CoV and ACE2 subunits in the complex are shown in blue and red colors, respectively, at the center. The structural motif containing the two helices of ACE2 and a β sheet of the CoV (or CoV-2) was monitored before (top) and after (bottom) 20 ns MD simulation. The left and right panels show the difference in conformational changes in the oxidized form (CoVox···ACE2ox) and reduced form (CoVred···ACE2red), respectively.
Scheme 1
References
ARTICLE SECTIONSThis article references 35 other publications.
- 1Schoeman, D.; Fielding, B. C. Coronavirus envelope protein: current knowledge. Virol. J. 2019, 16, 69 DOI: 10.1186/s12985-019-1182-0Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3M3gtFOrsQ%253D%253D&md5=c3c0865f45fb51cfe809c55d62207d59Coronavirus envelope protein: current knowledgeSchoeman Dewald; Fielding Burtram CVirology journal (2019), 16 (1), 69 ISSN:.BACKGROUND: Coronaviruses (CoVs) primarily cause enzootic infections in birds and mammals but, in the last few decades, have shown to be capable of infecting humans as well. The outbreak of severe acute respiratory syndrome (SARS) in 2003 and, more recently, Middle-East respiratory syndrome (MERS) has demonstrated the lethality of CoVs when they cross the species barrier and infect humans. A renewed interest in coronaviral research has led to the discovery of several novel human CoVs and since then much progress has been made in understanding the CoV life cycle. The CoV envelope (E) protein is a small, integral membrane protein involved in several aspects of the virus' life cycle, such as assembly, budding, envelope formation, and pathogenesis. Recent studies have expanded on its structural motifs and topology, its functions as an ion-channelling viroporin, and its interactions with both other CoV proteins and host cell proteins. MAIN BODY: This review aims to establish the current knowledge on CoV E by highlighting the recent progress that has been made and comparing it to previous knowledge. It also compares E to other viral proteins of a similar nature to speculate the relevance of these new findings. Good progress has been made but much still remains unknown and this review has identified some gaps in the current knowledge and made suggestions for consideration in future research. CONCLUSIONS: The most progress has been made on SARS-CoV E, highlighting specific structural requirements for its functions in the CoV life cycle as well as mechanisms behind its pathogenesis. Data shows that E is involved in critical aspects of the viral life cycle and that CoVs lacking E make promising vaccine candidates. The high mortality rate of certain CoVs, along with their ease of transmission, underpins the need for more research into CoV molecular biology which can aid in the production of effective anti-coronaviral agents for both human CoVs and enzootic CoVs.
- 2Fehr, A. R.; Perlman, S. Coronaviruses: an overview of their replication and pathogenesis. Methods Mol. Biol. 2015, 1282, 1– 23, DOI: 10.1007/978-1-4939-2438-7_1Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xls12ksL8%253D&md5=2e0fc18cd6b7312f0704f933a3614913Coronaviruses: an overview of their replication and pathogenesisFehr, Anthony R.; Perlman, StanleyMethods in Molecular Biology (New York, NY, United States) (2015), 1282 (Coronaviruses), 1-23CODEN: MMBIED; ISSN:1940-6029. (Springer)A review. Coronaviruses (CoVs), enveloped pos.-sense RNA viruses, are characterized by club-like spikes that project from their surface, an unusually large RNA genome, and a unique replication strategy. Coronaviruses cause a variety of diseases in mammals and birds ranging from enteritis in cows and pigs and upper respiratory disease in chickens to potentially lethal human respiratory infections. Here we provide a brief introduction to coronaviruses discussing their replication and pathogenicity, and current prevention and treatment strategies. We also discuss the outbreaks of the highly pathogenic Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) and the recently identified Middle Eastern Respiratory Syndrome Coronavirus (MERS-CoV).
- 3Chen, Y.; Guo, Y.; Pan, Y.; Zhao, Z. J. Structure analysis of the receptor binding of 2019-nCoV. Biochem. Biophys. Res. Commun. 2020, 525, 135– 140, DOI: 10.1016/j.bbrc.2020.02.071Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXjsFygtbs%253D&md5=53f249f15e3558acaf8eec89574bdf18Structure analysis of the receptor binding of 2019-nCoVChen, Yun; Guo, Yao; Pan, Yihang; Zhao, Zhizhuang JoeBiochemical and Biophysical Research Communications (2020), 525 (1), 135-140CODEN: BBRCA9; ISSN:0006-291X. (Elsevier B.V.)2019-nCoV is a newly identified coronavirus with high similarity to SARS-CoV. We performed a structural anal. of the receptor binding domain (RBD) of spike glycoprotein responsible for entry of coronaviruses into host cells. The RBDs from the two viruses share 72% identity in amino acid sequences, and mol. simulation reveals highly similar ternary structures. However, 2019-nCoV has a distinct loop with flexible glycyl residues replacing rigid prolyl residues in SARS-CoV. Mol. modeling revealed that 2019-nCoV RBD has a stronger interaction with angiotensin converting enzyme 2 (ACE2). A unique phenylalanine F486 in the flexible loop likely plays a major role because its penetration into a deep hydrophobic pocket in ACE2. ACE2 is widely expressed with conserved primary structures throughout the animal kingdom from fish, amphibians, reptiles, birds, to mammals. Structural anal. suggests that ACE2 from these animals can potentially bind RBD of 2019-nCoV, making them all possible natural hosts for the virus. 2019-nCoV is thought to be transmitted through respiratory droplets. However, since ACE2 is predominantly expressed in intestines, testis, and kidney, fecal-oral and other routes of transmission are also possible. Finally, antibodies and small mol. inhibitors that can block the interaction of ACE2 with RBD should be developed to combat the virus.
- 4Xu, H.; Zhong, L.; Deng, J.; Peng, J.; Dan, H.; Zeng, X.; Li, T.; Chen, Q. High expression of ACE2 receptor of 2019-nCoV on the epithelial cells of oral mucosa. Int. J. Oral Sci. 2020, 12, 8 DOI: 10.1038/s41368-020-0074-xGoogle Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXksVWnsL4%253D&md5=98de43264b244daee736cfdc99e87aadHigh expression of ACE2 receptor of 2019-nCoV on the epithelial cells of oral mucosaXu, Hao; Zhong, Liang; Deng, Jiaxin; Peng, Jiakuan; Dan, Hongxia; Zeng, Xin; Li, Taiwen; Chen, QianmingInternational Journal of Oral Science (2020), 12 (1), 8CODEN: IJOSFA; ISSN:2049-3169. (Nature Research)It has been reported that ACE2 is the main host cell receptor of 2019-nCoV and plays a crucial role in the entry of virus into the cell to cause the final infection. To investigate the potential route of 2019-nCov infection on the mucosa of oral cavity, bulk RNA-seq profiles from two public databases including The Cancer Genome Atlas (TCGA) and Functional Annotation of The Mammalian Genome Cap Anal. of Gene Expression (FANTOM5 CAGE) dataset were collected. RNA-seq profiling data of 13 organ types with para-carcinoma normal tissues from TCGA and 14 organ types with normal tissues from FANTOM5 CAGE were analyzed in order to explore and validate the expression of ACE2 on the mucosa of oral cavity. Further, single-cell transcriptomes from an independent data generated inhouse were used to identify and confirm the ACE2-expressing cell compn. and proportion in oral cavity. The results demonstrated that the ACE2 expressed on the mucosa of oral cavity. Interestingly, this receptor was highly enriched in epithelial cells of tongue. Preliminarily, those findings have explained the basic mechanism that the oral cavity is a potentially high risk for 2019-nCoV infectious susceptibility and provided a piece of evidence for the future prevention strategy in dental clin. practice as well as daily life.
- 5Ryser, H. J.; Levy, E. M.; Mandel, R.; DiSciullo, G. J. Inhibition of human immunodeficiency virus infection by agents that interfere with thiol-disulfide interchange upon virus-receptor interaction. Proc. Natl. Acad. Sci. U.S.A. 1994, 91, 4559– 4563, DOI: 10.1073/pnas.91.10.4559Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXktVSgs70%253D&md5=772426ff00253d510d64026f556da4f5Inhibition of human immunodeficiency virus infection by agents that interfere with thiol-disulfide interchange upon virus-receptor interactionRyser, Hugues J.-P.; Levy, Elinor M.; Mandel, Richard; DiSciullo, Gino J.Proceedings of the National Academy of Sciences of the United States of America (1994), 91 (10), 4559-63CODEN: PNASA6; ISSN:0027-8424.The cell surface of mammalian cells is capable of reductively cleaving disulfide bonds of exogenous membrane-bound macromols. (for instance, the interchain disulfide of diphtheria toxin), and inhibiting this process with membrane-impermeant sulfhydryl reagents prevents diphtheria toxin cytotoxicity. More recently it was found that the same membrane function can be inhibited by bacitracin, an inhibitor of protein disulfide-isomerase (PDI), and by monoclonal antibodies against PDI, suggesting that PDI catalyzes a thiol-disulfide interchange between its thiols and the disulfides of membrane-bound macromols. The authors provide evidence that the same reductive process plays a role in the penetration of membrane-bound human immunodeficiency virus (HIV) and show that HIV infection of human lymphoid cells is markedly inhibited by the membrane-impermeant sulfhydryl blocker 5,5'-dithiobis(2-nitrobenzoic acid), by bacitracin, and by anti-PDI antibodies. The results imply that HIV and its target cell engage in a thiol-disulfide interchange mediated by PDI and that the redn. of crit. disulfides in viral envelope glycoproteins may be the initial event that triggers conformational changes required for HIV entry and cell infection. These findings suggest addnl. approaches to impede cell infection by HIV.
- 6Gallina, A.; Hanley, T. M.; Mandel, R.; Trahey, M.; Broder, C. C.; Viglianti, G. A.; Ryser, H. J. Inhibitors of protein-disulfide isomerase prevent cleavage of disulfide bonds in receptor-bound glycoprotein 120 and prevent HIV-1 entry. J. Biol. Chem. 2002, 277, 50579– 50588, DOI: 10.1074/jbc.M204547200Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38Xps12ntrw%253D&md5=23bbd63c77a16fb983cd993886822676Inhibitors of Protein-Disulfide Isomerase Prevent Cleavage of Disulfide Bonds in Receptor-bound Glycoprotein 120 and Prevent HIV-1 EntryGallina, Angelo; Hanley, Timothy M.; Mandel, Richard; Trahey, Meg; Broder, Christopher C.; Viglianti, Gregory A.; Ryser, Hugues J.-P.Journal of Biological Chemistry (2002), 277 (52), 50579-50588CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)We previously reported that monoclonal antibodies to protein-disulfide isomerase (PDI) and other membrane-impermeant PDI inhibitors prevented HIV-1 infection. PDI is present at the surface of HIV-1 target cells and reduces disulfide bonds in a model peptide attached to the cell membrane. Here we show that sol. PDI cleaves disulfide bonds in recombinant envelope glycoprotein gp120 and that gp120 bound to the surface receptor CD4 undergoes a disulfide redn. that is prevented by PDI inhibitors. Concns. of inhibitors that prevent this redn. and inhibit the cleavage of surface-bound disulfide conjugate prevent infection at the level of HIV-1 entry. The entry of HIV-1 strains differing in their coreceptor specificities is similarly inhibited, and so is the redn. of gp120 bound to CD4 of coreceptor-neg. cells. PDI inhibitors also prevent HIV envelope-mediated cell-cell fusion but have no effect on the entry of HIV-1 pseudo-typed with murine leukemia virus envelope. Importantly, PDI coppts. with both sol. and cellular CD4. We propose that a PDI·CD4 assocn. at the cell surface enables PDI to reach CD4-bound virus and to reduce disulfide bonds present in the domain of gp120 that binds to CD4. Conformational changes resulting from the opening of gp120-disulfide loops may drive the processes of virus-cell and cell-cell fusion. The biochem. events described identify new potential targets for anti-HIV agents.
- 7Lavillette, D.; Barbouche, R.; Yao, Y.; Boson, B.; Cosset, F. L.; Jones, I. M.; Fenouillet, E. Significant redox insensitivity of the functions of the SARS-CoV spike glycoprotein: comparison with HIV envelope. J. Biol. Chem. 2006, 281, 9200– 9204, DOI: 10.1074/jbc.M512529200Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XjtVSrsrs%253D&md5=2b4152b59dc1dbe5d4a2f1e1b69bdafeSignificant Redox Insensitivity of the Functions of the SARS-CoV Spike Glycoprotein: Comparison with HIV envelopeLavillette, Dimitri; Barbouche, Rym; Yao, Yongxiu; Boson, Bertrand; Cosset, Francois-Loic; Jones, Ian M.; Fenouillet, EmmanuelJournal of Biological Chemistry (2006), 281 (14), 9200-9204CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)The capacity of the surface glycoproteins of enveloped viruses to mediate virus/cell binding and membrane fusion requires a proper thiol/disulfide balance. Chem. manipulation of their redox state using reducing agents or free sulfhydryl reagents affects virus/cell interaction. Conversely, natural thiol/disulfide rearrangements often occur during the cell interaction to trigger fusogenicity, hence the virus entry. We examd. the relationship between the redox state of the 20 cysteine residues of the SARS-CoV (severe acute respiratory syndrome coronavirus) Spike glycoprotein S1 subdomain and its functional properties. Mature S1 exhibited ∼4 unpaired cysteines, and chem. reduced S1 displaying up to ∼6 addnl. unpaired cysteines still bound ACE2 and enabled fusion. In addn., virus/cell membrane fusion occurred in the presence of sulfhydryl-blocking reagents and oxidoreductase inhibitors. Thus, in contrast to various viruses including HIV (human immunodeficiency virus) examd. in parallel, the functions of the SARS-CoV Spike glycoprotein exhibit a significant and surprising independence of redox state, which may contribute to the wide host range of the virus. These data suggest clues for molecularly engineering vaccine immunogens.
- 8Pan, S.; Chen, H. H.; Correia, C.; Dai, H.; Witt, T. A.; Kleppe, L. S.; Burnett, J. C., Jr.; Simari, R. D. Cell surface protein disulfide isomerase regulates natriuretic peptide generation of cyclic guanosine monophosphate. PLoS One 2014, 9, e112986 DOI: 10.1371/journal.pone.0112986Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitV2htb7L&md5=2b7f451941a888a4e0b170bf8f510138Cell surface protein disulfide isomerase regulates natriuretic peptide generation of cyclic guanosine monophosphatePan, Shuchong; Chen, Horng H.; Correia, Cristina; Dai, Haiming; Witt, Tyra A.; Kleppe, Laurel S.; Burnett, John C., Jr.; Simari, Robert D.PLoS One (2014), 9 (11), e112986/1-e112986/14, 14 pp.CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)Rationale: The family of natriuretic peptides (NPs), including atrial natriuretic peptide (ANP), B-type natriuretic peptide (BNP), and C-type natriuretic peptide (CNP), exert important and diverse actions for cardiovascular and renal homeostasis. The autocrine and paracrine functions of the NPs are primarily mediated through the cellular membrane bound guanylyl cyclase-linked receptors GC-A (NPR-A) and GC-B (NPR-B). As the ligands and receptors each contain disulfide bonds, a regulatory role for the cell surface protein disulfide isomerase (PDI) was investigated. Objective: We utilized complementary in vitro and in vivo models to det. the potential role of PDI in regulating the ability of the NPs to generate its second messenger, cyclic guanosine monophosphate. Methods and Results: Inhibition of PDI attenuated the ability of ANP, BNP and CNP to generate cGMP in human mesangial cells (HMCs), human umbilical vein endothelial cells (HUVECs), and human aortic smooth muscle cells (HASMCs), each of which were shown to express PDI. In LLC-PK1 cells, where PDI expression was undetectable by immunoblotting, PDI inhibition had a minimal effect on cGMP generation. Addn. of PDI to cultured LLC-PK1 cells increased intracellular cGMP generation mediated by ANP. Inhibition of PDI in vivo attenuated NP-mediated generation of cGMP by ANP. Surface Plasmon Resonance demonstrated modest and differential binding of the natriuretic peptides with immobilized PDI in a cell free system. However, PDI was shown to co-localize on the surface of cells with GC-A and GC-B by co-immunoprecpitation and immunohistochem. Conclusion: These data demonstrate for the first time that cell surface PDI expression and function regulate the capacity of natriuretic peptides to generate cGMP through interaction with their receptors.
- 9Mathys, L.; Balzarini, J. The role of cellular oxidoreductases in viral entry and virus infection-associated oxidative stress: potential therapeutic applications. Expert Opin. Ther. Targets 2016, 20, 123– 143, DOI: 10.1517/14728222.2015.1068760Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslKltbs%253D&md5=392ee62019de0363448d2b5fe86ac408The role of cellular oxidoreductases in viral entry and virus infection-associated oxidative stress: potential therapeutic applicationsMathys, Leen; Balzarini, JanExpert Opinion on Therapeutic Targets (2016), 20 (1), 123-143CODEN: EOTTAO; ISSN:1472-8222. (Taylor & Francis Ltd.)Cellular oxidoreductases catalyze thiol/disulfide exchange reactions in susceptible proteins and contribute to the cellular defense against oxidative stress. Oxidoreductases and oxidative stress are also involved in viral infections. In this overview, different aspects of the role of cellular oxidoreductases and oxidative stress during viral infections are discussed from a chemotherapeutic viewpoint. Entry of enveloped viruses into their target cells is triggered by the interaction of viral envelope glycoproteins with cellular (co)receptor(s) and depends on obligatory conformational changes in these viral envelope glycoproteins and/or cellular receptors. For some viruses, these conformational changes are mediated by cell surface-assocd. cellular oxidoreductases, which mediate disulfide bridge redns. in viral envelope glycoprotein(s). Therefore, targeting these oxidoreductases using oxidoreductase inhibitors might yield an interesting strategy to block viral entry of these viruses. Furthermore, since viral infections are often assocd. with systemic oxidative stress, contributing to disease progression, the enhancement of the cellular antioxidant defense systems might have potential as an adjuvant antiviral strategy, slowing down disease progression. Promising antiviral data were obtained for both strategies. However, potential pitfalls have also been identified for these strategies, indicating that it is important to carefully assess the benefits vs. risks of these antiviral strategies.
- 10Camini, F. C.; da Silva Caetano, C. C.; Almeida, L. T.; de Brito Magalhães, C. L. Implications of oxidative stress on viral pathogenesis. Arch. Virol. 2017, 162, 907– 917, DOI: 10.1007/s00705-016-3187-yGoogle Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtVOntg%253D%253D&md5=8acc801377bd621b2ea2fc12b5178fd7Implications of oxidative stress on viral pathogenesisCamini, Fernanda Caetano; Caetano, Camila Carla da Silva; Almeida, Leticia Trindade; Magalhaes, Cintia Lopes de BritoArchives of Virology (2017), 162 (4), 907-917CODEN: ARVIDF; ISSN:0304-8608. (Springer-Verlag GmbH)Reactive species are frequently formed after viral infections. Antioxidant defences, including enzymic and non-enzymic components, protect against reactive species, but sometimes these defences are not completely adequate. An imbalance in the prodn. of reactive species and the body's inability to detoxify these reactive species is referred to as oxidative stress. The aim of this review is to analyze the role of oxidative stress in the pathogenesis of viral infections and highlight some major therapeutic approaches that have gained importance, with regards to controlling virus-induced oxidative injury. Attention will be focused on DNA viruses (papillomaviruses, hepadnaviruses), RNA viruses (flaviviruses, orthomyxoviruses, paramyxoviruses, togaviruses) and retroviruses (human immunodeficiency virus). In general, viruses cause an imbalance in the cellular redox environment, which depending on the virus and the cell can result in different responses, e.g. cell signaling, antioxidant defences, reactive species, and other processes. Therefore, the modulation of reactive species prodn. and oxidative stress potentially represents a novel pharmacol. approach for reducing the consequences of viral pathogenesis.
- 11Fenouillet, E.; Barbouche, R.; Jones, I. M. Cell entry by enveloped viruses: redox considerations for HIV and SARS-coronavirus. Antioxid. Redox Signaling 2007, 9, 1009– 1034, DOI: 10.1089/ars.2007.1639Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXot1Wktrk%253D&md5=14c6275868393216f703e406135af1a1Cell entry by enveloped viruses: redox considerations for HIV and SARS-coronavirusFenouillet, Emmanuel; Barbouche, Rym; Jones, Ian M.Antioxidants & Redox Signaling (2007), 9 (8), 1009-1034CODEN: ARSIF2; ISSN:1523-0864. (Mary Ann Liebert, Inc.)A review. For enveloped viruses, genome entry into the target cell involves two major steps: virion binding to the cell-surface receptor and fusion of the virion and cell membranes. Virus-cell membrane fusion is mediated by the virus envelope complex, and its fusogenicity is the result of an active virus-cell interaction process that induces conformation changes within the envelope. For some viruses, such as influenza, exposure to an acidic milieu within the cell during the early steps of infection triggers the necessary structural changes. However, for other pathogens which are not exposed to such environmental stress, activation of fusogenicity can result from precise thiol/disulfide rearrangements mediated by either an endogenous redox autocatalytic isomerase or a cell-assocd. oxidoreductase. Study of the activation of HIV envelope fusogenicity has revealed new knowledge about how redox changes within a viral envelope trigger fusion. We discuss these findings and their implication for anti-HIV therapy. In addn., to compare and contrast the situation outlined for HIV with an enveloped virus that can fuse with the cell plasma membrane independent of the redox status of its envelope protein, we review parallel data obtained on SARS coronavirus entry.
- 12Yi, M. C.; Khosla, C. Thiol-Disulfide Exchange Reactions in the Mammalian Extracellular Environment. Annu. Rev. Chem. Biomol. Eng. 2016, 7, 197– 222, DOI: 10.1146/annurev-chembioeng-080615-033553Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XltFGrsLs%253D&md5=4c2bb579953388a3de9ab8c86c606a22Thiol-Disulfide Exchange Reactions in the Mammalian Extracellular EnvironmentYi, Michael C.; Khosla, ChaitanAnnual Review of Chemical and Biomolecular Engineering (2016), 7 (), 197-222CODEN: ARCBCY; ISSN:1947-5438. (Annual Reviews)Disulfide bonds represent versatile posttranslational modifications whose roles encompass the structure, catalysis, and regulation of protein function. Due to the oxidizing nature of the extracellular environment, disulfide bonds found in secreted proteins were once believed to be inert. This notion has been challenged by the discovery of redox-sensitive disulfides that, once cleaved, can lead to changes in protein activity. These functional disulfides are twisted into unique configurations, leading to high strain and potential energy. In some cases, cleavage of these disulfides can lead to a gain of function in protein activity. Thus, these motifs can be referred to as switches. We describe the couples that control redox in the extracellular environment, examine several examples of proteins with switchable disulfides, and discuss the potential applications of disulfides in mol. biol.
- 13Li, F. Structural analysis of major species barriers between humans and palm civets for severe acute respiratory syndrome coronavirus infections. J. Virol. 2008, 82, 6984– 6991, DOI: 10.1128/JVI.00442-08Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXosFWmsro%253D&md5=5df8566921a86428b63d3e3b261896b3Structural analysis of major species barriers between humans and palm civets for severe acute respiratory syndrome coronavirus infectionsLi, FangJournal of Virology (2008), 82 (14), 6984-6991CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)It is believed that a novel coronavirus, severe acute respiratory syndrome coronavirus (SARS-CoV), was passed from palm civets to humans and caused the epidemic of SARS in 2002 to 2003. The major species barriers between humans and civets for SARS-CoV infections are the specific interactions between a defined receptor-binding domain (RBD) on a viral spike protein and its host receptor, angiotensin-converting enzyme 2 (ACE2). In this study a chimeric ACE2 bearing the crit. N-terminal helix from civet and the remaining peptidase domain from human was constructed, and it was shown that this construct has the same receptor activity as civet ACE2. In addn., crystal structures of the chimeric ACE2 complexed with RBDs from various human and civet SARS-CoV strains were detd. These structures, combined with a previously detd. structure of human ACE2 complexed with the RBD from a human SARS-CoV strain, have revealed a structural basis for understanding the major species barriers between humans and civets for SARS-CoV infections. They show that the major species barriers are detd. by interactions between four ACE2 residues (residues 31, 35, 38, and 353) and two RBD residues (residues 479 and 487), that early civet SARS-CoV isolates were prevented from infecting human cells due to imbalanced salt bridges at the hydrophobic virus/receptor interface, and that SARS-CoV has evolved to gain sustained infectivity for human cells by eliminating unfavorable free charges at the interface through stepwise mutations at positions 479 and 487. These results enhance our understanding of host adaptations and cross-species infections of SARS-CoV and other emerging animal viruses.
- 14Wrapp, D.; Wang, N.; Corbett, K. S.; Goldsmith, J. A.; Hsieh, C. L.; Abiona, O.; Graham, B. S.; McLellan, J. S. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science 2020, 367, 1260– 1263, DOI: 10.1126/science.abb2507Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXkvFemt70%253D&md5=27d08cbb9a43d1da051a8a92a9f68aa5Cryo-EM structure of the 2019-nCoV spike in the prefusion conformationWrapp, Daniel; Wang, Nianshuang; Corbett, Kizzmekia S.; Goldsmith, Jory A.; Hsieh, Ching-Lin; Abiona, Olubukola; Graham, Barney S.; McLellan, Jason S.Science (Washington, DC, United States) (2020), 367 (6483), 1260-1263CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)The outbreak of a novel coronavirus (2019-nCoV) represents a pandemic threat that has been declared a public health emergency of international concern. The CoV spike (S) glycoprotein is a key target for vaccines, therapeutic antibodies, and diagnostics. To facilitate medical countermeasure development, we detd. a 3.5-angstrom-resoln. cryo-electron microscopy structure of the 2019-nCoV S trimer in the prefusion conformation. The predominant state of the trimer has one of the three receptor-binding domains (RBDs) rotated up in a receptor-accessible conformation. We also provide biophys. and structural evidence that the 2019-nCoV S protein binds angiotensin-converting enzyme 2 (ACE2) with higher affinity than does severe acute respiratory syndrome (SARS)-CoV S. Addnl., we tested several published SARS-CoV RBD-specific monoclonal antibodies and found that they do not have appreciable binding to 2019-nCoV S, suggesting that antibody cross-reactivity may be limited between the two RBDs. The structure of 2019-nCoV S should enable the rapid development and evaluation of medical countermeasures to address the ongoing public health crisis.
- 15Wang, Q.; Zhang, Y.; Wu, L.; Niu, S.; Song, C.; Zhang, Z.; Lu, G.; Qiao, C.; Hu, Y.; Yuen, K. Y.; Wang, Q.; Zhou, H.; Yan, J.; Qi, J. Structural and Functional Basis of SARS-CoV-2 Entry by Using Human ACE2. Cell 2020, 181, 894– 904.e9, DOI: 10.1016/j.cell.2020.03.045Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXmvFGrur4%253D&md5=188108e44f104860d4a2b83707ce7230Structural and Functional Basis of SARS-CoV-2 Entry by Using Human ACE2Wang, Qihui; Zhang, Yanfang; Wu, Lili; Niu, Sheng; Song, Chunli; Zhang, Zengyuan; Lu, Guangwen; Qiao, Chengpeng; Hu, Yu; Yuen, Kwok-Yung; Wang, Qisheng; Zhou, Huan; Yan, Jinghua; Qi, JianxunCell (Cambridge, MA, United States) (2020), 181 (4), 894-904.e9CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The recent emergence of a novel coronavirus (SARS-CoV-2) in China has caused significant public health concerns. Recently, ACE2 was reported as an entry receptor for SARS-CoV-2. In this study, we present the crystal structure of the C-terminal domain of SARS-CoV-2 (SARS-CoV-2-CTD) spike (S) protein in complex with human ACE2 (hACE2), which reveals a hACE2-binding mode similar overall to that obsd. for SARS-CoV. However, at. details at the binding interface demonstrate that key residue substitutions in SARS-CoV-2-CTD slightly strengthen the interaction and lead to higher affinity for receptor binding than SARS-RBD. Addnl., a panel of murine monoclonal antibodies (mAbs) and polyclonal antibodies (pAbs) against SARS-CoV-S1/receptor-binding domain (RBD) were unable to interact with the SARS-CoV-2 S protein, indicating notable differences in antigenicity between SARS-CoV and SARS-CoV-2. These findings shed light on the viral pathogenesis and provide important structural information regarding development of therapeutic countermeasures against the emerging virus.
- 16Yan, R.; Zhang, Y.; Li, Y.; Xia, L.; Guo, Y.; Zhou, Q. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science 2020, 367, 1444– 1448, DOI: 10.1126/science.abb2762Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXlslymsLo%253D&md5=ff4dfdfc646ea878cfb325019160e94aStructural basis for the recognition of SARS-CoV-2 by full-length human ACE2Yan, Renhong; Zhang, Yuanyuan; Li, Yaning; Xia, Lu; Guo, Yingying; Zhou, QiangScience (Washington, DC, United States) (2020), 367 (6485), 1444-1448CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)Angiotensin-converting enzyme 2 (ACE2) is the cellular receptor for severe acute respiratory syndrome coronavirus (SARS-CoV) and the new coronavirus (SARS-CoV-2) that is causing the serious coronavirus disease 2019 (COVID-19) epidemic. Here, we present cryo-electron microscopy structures of full-length human ACE2 in the presence of the neutral amino acid transporter B0AT1 with or without the receptor binding domain (RBD) of the surface spike glycoprotein (S protein) of SARS-CoV-2, both at an overall resoln. of 2.9 angstroms, with a local resoln. of 3.5 angstroms at the ACE2-RBD interface. The ACE2-B0AT1 complex is assembled as a dimer of heterodimers, with the collectrin-like domain of ACE2 mediating homodimerization. The RBD is recognized by the extracellular peptidase domain of ACE2 mainly through polar residues. These findings provide important insights into the mol. basis for coronavirus recognition and infection.
- 17Adams, L. M.; Andrews, R. J.; Hu, Q. H.; Schmit, H. L.; Hati, S.; Bhattacharyya, S. Crowder-Induced Conformational Ensemble Shift in Escherichia coli Prolyl-tRNA Synthetase. Biophys. J. 2019, 117, 1269– 1284, DOI: 10.1016/j.bpj.2019.08.033Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhslyisLzJ&md5=c2c9242b3ced38f0814a96f577208347Crowder-Induced Conformational Ensemble Shift in Escherichia coli Prolyl-tRNA SynthetaseAdams, Lauren M.; Andrews, Ryan J.; Hu, Quin H.; Schmit, Heidi L.; Hati, Sanchita; Bhattacharyya, SudeepBiophysical Journal (2019), 117 (7), 1269-1284CODEN: BIOJAU; ISSN:0006-3495. (Cell Press)The effect of mol. crowding on the structure and function of Escherichia coli prolyl-tRNA synthetase (Ec ProRS), a member of the aminoacyl-tRNA synthetase family, has been investigated using a combined exptl. and theor. method. Ec ProRS is a multidomain enzyme; coupled-domain dynamics are essential for efficient catalysis. To gain insight into the mechanistic detail of the crowding effect, kinetic studies were conducted with varying concns. and sizes of crowders. In parallel, spectroscopic and quantum chem. studies were employed to probe the "soft interactions" between crowders and protein side chains. Finally, the dynamics of the dimeric protein was examd. in the presence of crowders using a long-duration (70 ns) classical mol. dynamic simulations. The results of the simulations revealed a shift in the conformational ensemble, which is consistent with the preferential exclusion of cosolutes. The "soft interactions" model of the crowding effect also explained the alteration in kinetic parameters. In summary, the study found that the effects of mol. crowding on both conformational dynamics and catalytic function are correlated in the multidomain Ec ProRS, an enzyme that is central to protein synthesis in all living cells. This study affirmed that large and small cosolutes have considerable impacts on the structure, dynamics, and function of modular proteins and therefore must be considered for stabilizing protein-based pharmaceuticals and industrial enzymes.
- 18Bechtel, T. J.; Weerapana, E. From structure to redox: The diverse functional roles of disulfides and implications in disease. Proteomics 2017, 17, 1600391 DOI: 10.1002/pmic.201600391Google ScholarThere is no corresponding record for this reference.
- 19Ballatori, N.; Krance, S. M.; Marchan, R.; Hammond, C. L. Plasma membrane glutathione transporters and their roles in cell physiology and pathophysiology. Mol. Aspects Med. 2009, 30, 13– 28, DOI: 10.1016/j.mam.2008.08.004Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXltFelurc%253D&md5=43e9fd45d618eecfd3b2c73780b0f833Plasma membrane glutathione transporters and their roles in cell physiology and pathophysiologyBallatori, Nazzareno; Krance, Suzanne M.; Marchan, Rosemarie; Hammond, Christine L.Molecular Aspects of Medicine (2009), 30 (1-2), 13-28CODEN: MAMED5; ISSN:0098-2997. (Elsevier B.V.)A review. Reduced glutathione (GSH) is crit. for many cellular processes, and both its intracellular and extracellular concns. are tightly regulated. Intracellular GSH levels are regulated by two main mechanisms: by adjusting the rates of synthesis and of export from cells. Some of the proteins responsible for GSH export from mammalian cells have recently been identified, and there is increasing evidence that these GSH exporters are multispecific and multifunctional, regulating a no. of key biol. processes. In particular, some of the multidrug resistance-assocd. proteins (Mrp/Abcc) appear to mediate GSH export and homeostasis. The Mrp proteins mediate not only GSH efflux, but they also export oxidized glutathione derivs. (e.g., glutathione disulfide (GSSG), S-nitrosoglutathione (GS-NO), and glutathione-metal complexes), as well as other glutathione S-conjugates. The ability to export both GSH and oxidized derivs. of GSH, endows these transporters with the capacity to directly regulate the cellular thiol-redox status, and therefore the ability to influence many key signaling and biochem. pathways. Among the many processes that are influenced by the GSH transporters are apoptosis, cell proliferation, and cell differentiation. This report summarizes the evidence that Mrps contribute to the regulation of cellular GSH levels and the thiol-redox state, and thus to the many biochem. processes that are influenced by this tripeptide.
- 20Xia, H.; Suda, S.; Bindom, S.; Feng, Y.; Gurley, S. B.; Seth, D.; Navar, L. G.; Lazartigues, E. ACE2-mediated reduction of oxidative stress in the central nervous system is associated with improvement of autonomic function. PLoS One 2011, 6, e22682 DOI: 10.1371/journal.pone.0022682Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtVOisr%252FP&md5=8a10eafb719a132285886913f5c245c0ACE2-mediated reduction of oxidative stress in the central nervous system is associated with improvement of autonomic functionXia, Huijing; Suda, Sonia; Bindom, Sharell; Feng, Yumei; Gurley, Susan B.; Seth, Dale; Navar, L. Gabriel; Lazartigues, EricPLoS One (2011), 6 (7), e22682CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)Oxidative stress in the central nervous system mediates the increase in sympathetic tone that precedes the development of hypertension. We hypothesized that by transforming Angiotensin-II (AngII) into Ang-(1-7), ACE2 might reduce AngII-mediated oxidative stress in the brain and prevent autonomic dysfunction. To test this hypothesis, a relationship between ACE2 and oxidative stress was first confirmed in a mouse neuroblastoma cell line (Neuro2A cells) treated with AngII and infected with Ad-hACE2. ACE2 overexpression resulted in a redn. of reactive oxygen species (ROS) formation. In vivo, ACE2 knockout (ACE2-/y) mice and non-transgenic (NT) littermates were infused with AngII (10 days) and infected with Ad-hACE2 in the paraventricular nucleus (PVN). Baseline blood pressure (BP), AngII and brain ROS levels were not different between young mice (12 wk). However, cardiac sympathetic tone, brain NADPH oxidase and SOD activities were significantly increased in ACE2-/y. Post infusion, plasma and brain AngII levels were also significantly higher in ACE2-/y, although BP was similarly increased in both genotypes. ROS formation in the PVN and RVLM was significantly higher in ACE2-/y mice following AngII infusion. Similar phenotypes, i.e. increased oxidative stress, exacerbated dysautonomia and hypertension, were also obsd. on baseline in mature ACE2-/y mice (48 wk). ACE2 gene therapy to the PVN reduced AngII-mediated increase in NADPH oxidase activity and normalized cardiac dysautonomia in ACE2-/y mice. Altogether, these data indicate that ACE2 gene deletion promotes age-dependent oxidative stress, autonomic dysfunction and hypertension, while PVN-targeted ACE2 gene therapy decreases ROS formation via NADPH oxidase inhibition and improves autonomic function. Accordingly, ACE2 could represent a new target for the treatment of hypertension-assocd. dysautonomia and oxidative stress.
- 21Humphrey, W.; Dalke, A.; Schulten, K. VMD: visual molecular dynamics. J. Mol. Graphics 1996, 14, 33– 38, DOI: 10.1016/0263-7855(96)00018-5Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28Xis12nsrg%253D&md5=1e3094ec3151fb85c5ff05f8505c78d5VDM: visual molecular dynamicsHumphrey, William; Dalke, Andrew; Schulten, KlausJournal of Molecular Graphics (1996), 14 (1), 33-8, plates, 27-28CODEN: JMGRDV; ISSN:0263-7855. (Elsevier)VMD is a mol. graphics program designed for the display and anal. of mol. assemblies, in particular, biopolymers such as proteins and nucleic acids. VMD can simultaneously display any no. of structures using a wide variety of rendering styles and coloring methods. Mols. are displayed as one or more "representations," in which each representation embodies a particular rendering method and coloring scheme for a selected subset of atoms. The atoms displayed in each representation are chosen using an extensive atom selection syntax, which includes Boolean operators and regular expressions. VMD provides a complete graphical user interface for program control, as well as a text interface using the Tcl embeddable parser to allow for complex scripts with variable substitution, control loops, and function calls. Full session logging is supported, which produces a VMD command script for later playback. High-resoln. raster images of displayed mols. may be produced by generating input scripts for use by a no. of photorealistic image-rendering applications. VMD has also been expressly designed with the ability to animate mol. dynamics (MD) simulation trajectories, imported either from files or from a direct connection to a running MD simulation. VMD is the visualization component of MDScope, a set of tools for interactive problem solving in structural biol., which also includes the parallel MD program NAMD, and the MDCOMM software used to connect the visualization and simulation programs, VMD is written in C++, using an object-oriented design; the program, including source code and extensive documentation, is freely available via anonymous ftp and through the World Wide Web.
- 22Guvench, O.; Hatcher, E.; Venable, R. M.; Pastor, R. W.; MacKerell, A. D. CHARMM Additive All-Atom Force Field for Glycosidic Linkages between Hexopyranoses. J. Chem. Theory Comput. 2009, 5, 2353– 2370, DOI: 10.1021/ct900242eGoogle Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtVartbrM&md5=575b6daab16fbafc4d00ee445f16328dCHARMM Additive All-Atom Force Field for Glycosidic Linkages between HexopyranosesGuvench, Olgun; Hatcher, Elizabeth; Venable, Richard M.; Pastor, Richard W.; MacKerell, Alexander D., Jr.Journal of Chemical Theory and Computation (2009), 5 (9), 2353-2370CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)We present an extension of the CHARMM hexopyranose monosaccharide additive all-atom force field to enable modeling of glycosidic-linked hexopyranose polysaccharides. The new force field parameters encompass 1-1, 1-2, 1-3, 1-4, and 1-6 hexopyranose glycosidic linkages, as well as O-methylation at the C1 anomeric carbon, and are developed to be consistent with the CHARMM all-atom biomol. force fields for proteins, nucleic acids, and lipids. The parameters are developed in a hierarchical fashion using model compds. contg. the key atoms in the full carbohydrates, in particular O-methyl-tetrahydropyran and glycosidic-linked dimers consisting of two mols. of tetrahyropyran or one mol. of tetrahydropyran and one of cyclohexane. Target data for parameter optimization include full two-dimensional energy surfaces defined by the Φ/Ψ glycosidic dihedral angles in the disaccharide analogs, as detd. by quantum mech. MP2/cc-pVTZ single point energies on MP2/6-31G(d) optimized structures (MP2/cc-pVTZ//MP2/6-31G(d)). In order to achieve balanced, transferable dihedral parameters for the Φ/Ψ glycosidic dihedral angles, surfaces for all possible chiralities at the ring carbon atoms involved in the glycosidic linkages are considered, resulting in over 5000 MP2/cc-pVTZ//MP2/6-31G(d) conformational energies. Also included as target data are vibrational frequencies, pair interaction energies and distances with water mols., and intramol. geometries including distortion of the glycosidic valence angle as a function of the glycosidic dihedral angles. The model compd. optimized force field parameters are validated on full disaccharides through the comparison of mol. dynamics results to available exptl. data. Good agreement is achieved with expt. for a variety of properties including crystal cell parameters and intramol. geometries, aq. densities, and aq. NMR coupling consts. assocd. with the glycosidic linkage. The newly developed parameters allow for the modeling of linear, branched, and cyclic hexopyranose glycosides both alone and in heterogeneous systems including proteins, nucleic acids, and/or lipids when combined with existing CHARMM biomol. force fields.
- 23Raman, E. P.; Guvench, O.; MacKerell, A. D., Jr. CHARMM additive all-atom force field for glycosidic linkages in carbohydrates involving furanoses. J. Phys. Chem. B 2010, 114, 12981– 12994, DOI: 10.1021/jp105758hGoogle Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtFKlu73F&md5=a4619fcb09cafebb3f84a3d7f59ea999CHARMM Additive All-Atom Force Field for Glycosidic Linkages in Carbohydrates Involving FuranosesRaman, E. Prabhu; Guvench, Olgun; MacKerell, Alexander D., Jr.Journal of Physical Chemistry B (2010), 114 (40), 12981-12994CODEN: JPCBFK; ISSN:1520-6106. (American Chemical Society)Presented is an extension of the CHARMM additive carbohydrate all-atom force field to enable modeling of polysaccharides contg. furanose sugars. The new force field parameters encompass 1 ↔ 2, 1 → 3, 1 → 4, and 1 → 6 pyranose-furanose linkages and 2 → 1 and 2 → 6 furanose-furanose linkages, building on existing hexopyranose and furanose monosaccharide parameters. The model compds. were chosen to be monomers or glycosidic-linked dimers of tetrahydropyran (THP) and THF as to contain the key atoms in full carbohydrates. Target data for optimization included two-dimensional quantum mech. (QM) potential energy scans of the Φ/Ψ glycosidic dihedral angles, with geometry optimization at the MP2/6-31G(d) level followed by MP2/cc-pVTZ single-point energies. All possible chiralities of the model compds. at the linkage carbons were considered, and for each geometry, the THF ring was constrained to the favorable south or north conformations. Target data also included QM vibrational frequencies and pair interaction energies and distances with water mols. Force field validation included comparison of computed crystal properties, aq. soln. densities, and NMR J-coupling consts. to exptl. ref. values. Simulations of infinite crystals showed good agreement with exptl. values for intramol. geometries as well as for crystal unit cell parameters. Addnl., aq. soln. densities and available NMR data were reproduced to a high degree of accuracy, thus validating the hierarchically optimized parameters in both cryst. and aq. condensed phases. The newly developed parameters allow for the modeling of linear, branched, and cyclic pyranose/furanose polysaccharides both alone and in heterogeneous systems including proteins, nucleic acids, and/or lipids when combined with existing additive CHARMM biomol. force fields.
- 24Guvench, O.; Mallajosyula, S. S.; Raman, E. P.; Hatcher, E.; Vanommeslaeghe, K.; Foster, T. J.; Jamison, F. W., II; Mackerell, A. D., Jr. CHARMM additive all-atom force field for carbohydrate derivatives and its utility in polysaccharide and carbohydrate-protein modeling. J. Chem. Theory Comput. 2011, 7, 3162– 3180, DOI: 10.1021/ct200328pGoogle Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtFSru77P&md5=fc3c05ef4c95895ee3e7b08472ef3554CHARMM Additive All-Atom Force Field for Carbohydrate Derivatives and Its Utility in Polysaccharide and Carbohydrate-Protein ModelingGuvench, Olgun; Mallajosyula, Sairam S.; Raman, E. Prabhu; Hatcher, Elizabeth; Vanommeslaeghe, Kenno; Foster, Theresa J.; Jamison, Francis W.; MacKerell, Alexander D.Journal of Chemical Theory and Computation (2011), 7 (10), 3162-3180CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Monosaccharide derivs. such as xylose, fucose, N-acetylglucosamine (GlcNAc), N-acetylgalactosamine (GlaNAc), glucuronic acid, iduronic acid, and N-acetylneuraminic acid (Neu5Ac) are important components of eukaryotic glycans. The present work details the development of force-field parameters for these monosaccharides and their covalent connections to proteins via O linkages to serine or threonine side chains and via N linkages to asparagine side chains. The force field development protocol was designed to explicitly yield parameters that are compatible with the existing CHARMM additive force field for proteins, nucleic acids, lipids, carbohydrates, and small mols. Therefore, when combined with previously developed parameters for pyranose and furanose monosaccharides, for glycosidic linkages between monosaccharides, and for proteins, the present set of parameters enables the mol. simulation of a wide variety of biol. important mols. such as complex carbohydrates and glycoproteins. Parametrization included fitting to quantum mech. (QM) geometries and conformational energies of model compds., as well as to QM pair interaction energies and distances of model compds. with water. Parameters were validated in the context of crystals of relevant monosaccharides, as well NMR and/or x-ray crystallog. data on larger systems including oligomeric hyaluronan, sialyl Lewis X, O- and N-linked glycopeptides, and a lectin:sucrose complex. As the validated parameters are an extension of the CHARMM all-atom additive biomol. force field, they further broaden the types of heterogeneous systems accessible with a consistently developed force-field model.
- 25Best, R. B.; Zhu, X.; Shim, J.; Lopes, P. E. M.; Mittal, J.; Feig, M.; MacKerell, A. D. Optimization of the additive CHARMM all-atom protein force field targeting improved sampling of the backbone φ, ψ and side-chain χ(1) and χ(2) dihedral angles. J. Chem. Theory Comput. 2012, 8, 3257– 3273, DOI: 10.1021/ct300400xGoogle Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtVKqurfP&md5=9a48a0c5770fb1e887c3bb34d45b1354Optimization of the Additive CHARMM All-Atom Protein Force Field Targeting Improved Sampling of the Backbone .vphi., ψ and Side-Chain χ1 and χ2 Dihedral AnglesBest, Robert B.; Zhu, Xiao; Shim, Jihyun; Lopes, Pedro E. M.; Mittal, Jeetain; Feig, Michael; MacKerell, Alexander D.Journal of Chemical Theory and Computation (2012), 8 (9), 3257-3273CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)While the quality of the current CHARMM22/CMAP additive force field for proteins has been demonstrated in a large no. of applications, limitations in the model with respect to the equil. between the sampling of helical and extended conformations in folding simulations have been noted. To overcome this, as well as make other improvements in the model, we present a combination of refinements that should result in enhanced accuracy in simulations of proteins. The common (non-Gly, -Pro) backbone CMAP potential has been refined against exptl. soln. NMR data for weakly structured peptides, resulting in a rebalancing of the energies of the α-helix and extended regions of the Ramachandran map, correcting the α-helical bias of CHARMM22/CMAP. The Gly and Pro CMAPs have been refitted to more accurate quantum-mech. energy surfaces. Side-chain torsion parameters have been optimized by fitting to backbone-dependent quantum-mech. energy surfaces, followed by addnl. empirical optimization targeting NMR scalar couplings for unfolded proteins. A comprehensive validation of the revised force field was then performed against a collection of exptl. data: (i) comparison of simulations of eight proteins in their crystal environments with crystal structures; (ii) comparison with backbone scalar couplings for weakly structured peptides; (iii) comparison with NMR residual dipolar couplings and scalar couplings for both backbone and side-chains in folded proteins; (iv) equil. folding of mini-proteins. The results indicate that the revised CHARMM 36 parameters represent an improved model for modeling and simulation studies of proteins, including studies of protein folding, assembly, and functionally relevant conformational changes.
- 26Mallajosyula, S. S.; Guvench, O.; Hatcher, E.; Mackerell, A. D., Jr. CHARMM Additive All-Atom Force Field for Phosphate and Sulfate Linked to Carbohydrates. J. Chem. Theory Comput. 2012, 8, 759– 776, DOI: 10.1021/ct200792vGoogle Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs1Oju7zP&md5=8350bcdedba6a5e80675f9cae77dfc7eCHARMM Additive All-Atom Force Field for Phosphate and Sulfate Linked to CarbohydratesMallajosyula, Sairam S.; Guvench, Olgun; Hatcher, Elizabeth; MacKerell, Alexander D.Journal of Chemical Theory and Computation (2012), 8 (2), 759-776CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Presented is an extension of the CHARMM additive all-atom carbohydrate force field to enable the modeling of phosphate and sulfate linked to carbohydrates. The parameters are developed in a hierarchical fashion using model compds. contg. the key atoms in the full carbohydrates. Target data for parameter optimization included full two-dimensional energy surfaces defined by the glycosidic dihedral angle pairs in the phosphate/sulfate model compd. analogs of hexopyranose monosaccharide phosphates and sulfates, as detd. by quantum mech. (QM) MP2/cc-pVTZ single point energies on MP2/6-31+G(d) optimized structures. To achieve balanced, transferable dihedral parameters for the dihedral angles, surfaces for all possible anomeric and conformational states were included during the parametrization process. To model physiol. relevant systems, both the mono- and dianionic charged states were studied for the phosphates. This resulted in over 7000 MP2/cc-pVTZ//MP2/6-31G+(d) model compd. conformational energies which, supplemented with QM geometries, were the main target data for the parametrization. Parameters were validated against crystals of relevant monosaccharide derivs. obtained from the Cambridge Structural Database (CSD) and larger systems, inositol-(tri/tetra/penta) phosphates noncovalently bound to the pleckstrin homol. (PH) domain and oligomeric chondroitin sulfate in soln. and in complex with cathepsin K protein.
- 27Essmann, U.; Perera, L.; Berkowitz, M. L. A smooth particle mesh Ewald method. J. Chem. Phys. 1995, 103, 8577, DOI: 10.1063/1.470117Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXptlehtrw%253D&md5=092a679dd3bee08da28df41e302383a7A smooth particle mesh Ewald methodEssmann, Ulrich; Perera, Lalith; Berkowitz, Max L.; Darden, Tom; Lee, Hsing; Pedersen, Lee G.Journal of Chemical Physics (1995), 103 (19), 8577-93CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)The previously developed particle mesh Ewald method is reformulated in terms of efficient B-spline interpolation of the structure factors. This reformulation allows a natural extension of the method to potentials of the form 1/rp with p ≥ 1. Furthermore, efficient calcn. of the virial tensor follows. Use of B-splines in the place of Lagrange interpolation leads to analytic gradients as well as a significant improvement in the accuracy. The authors demonstrate that arbitrary accuracy can be achieved, independent of system size N, at a cost that scales as N log(N). For biomol. systems with many thousands of atoms and this method permits the use of Ewald summation at a computational cost comparable to that of a simple truncation method of 10 Å or less.
- 28Jurrus, E.; Engel, D.; Star, K.; Monson, K.; Brandi, J.; Felberg, L. E.; Brookes, D. H.; Wilson, L.; Chen, J.; Liles, K.; Chun, M.; Li, P.; Gohara, D. W.; Dolinsky, T.; Konecny, R.; Koes, D. R.; Nielsen, J. E.; Head-Gordon, T.; Geng, W.; Krasny, R.; Wei, G. W.; Holst, M. J.; McCammon, J. A.; Baker, N. A. Improvements to the APBS biomolecular solvation software suite. Protein Sci. 2018, 27, 112– 128, DOI: 10.1002/pro.3280Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhslSkt7vI&md5=99651d125e38f4a85d453fecf0f71652Improvements to the APBS biomolecular solvation software suiteJurrus, Elizabeth; Engel, Dave; Star, Keith; Monson, Kyle; Brandi, Juan; Felberg, Lisa E.; Brookes, David H.; Wilson, Leighton; Chen, Jiahui; Liles, Karina; Chun, Minju; Li, Peter; Gohara, David W.; Dolinsky, Todd; Konecny, Robert; Koes, David R.; Nielsen, Jens Erik; Head-Gordon, Teresa; Geng, Weihua; Krasny, Robert; Wei, Guo-Wei; Holst, Michael J.; McCammon, J. Andrew; Baker, Nathan A.Protein Science (2018), 27 (1), 112-128CODEN: PRCIEI; ISSN:1469-896X. (Wiley-Blackwell)The Adaptive Poisson-Boltzmann Solver (APBS) software was developed to solve the equations of continuum electrostatics for large biomol. assemblages that have provided impact in the study of a broad range of chem., biol., and biomedical applications. APBS addresses the three key technol. challenges for understanding solvation and electrostatics in biomedical applications: accurate and efficient models for biomol. solvation and electrostatics, robust and scalable software for applying those theories to biomol. systems, and mechanisms for sharing and analyzing biomol. electrostatics data in the scientific community. To address new research applications and advancing computational capabilities, we have continually updated APBS and its suite of accompanying software since its release in 2001. In this article, we discuss the models and capabilities that have recently been implemented within the APBS software package including a Poisson-Boltzmann anal. and a semi-anal. solver, an optimized boundary element solver, a geometry-based geometric flow solvation model, a graph theory-based algorithm for detg. pKa values, and an improved web-based visualization tool for viewing electrostatics.
- 29Dolinsky, T. J.; Czodrowski, P.; Li, H.; Nielsen, J. E.; Jensen, J. H.; Klebe, G.; Baker, N. A. PDB2PQR: expanding and upgrading automated preparation of biomolecular structures for molecular simulations. Nucleic Acids Res. 2007, 35, W522– W525, DOI: 10.1093/nar/gkm276Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2svhsVGltQ%253D%253D&md5=18883d2f9a6f30dba143a4a2ca80b0d8PDB2PQR: expanding and upgrading automated preparation of biomolecular structures for molecular simulationsDolinsky Todd J; Czodrowski Paul; Li Hui; Nielsen Jens E; Jensen Jan H; Klebe Gerhard; Baker Nathan ANucleic acids research (2007), 35 (Web Server issue), W522-5 ISSN:.Real-world observable physical and chemical characteristics are increasingly being calculated from the 3D structures of biomolecules. Methods for calculating pK(a) values, binding constants of ligands, and changes in protein stability are readily available, but often the limiting step in computational biology is the conversion of PDB structures into formats ready for use with biomolecular simulation software. The continued sophistication and integration of biomolecular simulation methods for systems- and genome-wide studies requires a fast, robust, physically realistic and standardized protocol for preparing macromolecular structures for biophysical algorithms. As described previously, the PDB2PQR web server addresses this need for electrostatic field calculations (Dolinsky et al., Nucleic Acids Research, 32, W665-W667, 2004). Here we report the significantly expanded PDB2PQR that includes the following features: robust standalone command line support, improved pK(a) estimation via the PROPKA framework, ligand parameterization via PEOE_PB charge methodology, expanded set of force fields and easily incorporated user-defined parameters via XML input files, and improvement of atom addition and optimization code. These features are available through a new web interface (http://pdb2pqr.sourceforge.net/), which offers users a wide range of options for PDB file conversion, modification and parameterization.
- 30Johnson, J. M.; Sanford, B. L.; Strom, A. M.; Tadayon, S. N.; Lehman, B. P.; Zirbes, A. M.; Bhattacharyya, S.; Musier-Forsyth, K.; Hati, S. Multiple pathways promote dynamical coupling between catalytic domains in Escherichia coli prolyl-tRNA synthetase. Biochemistry 2013, 52, 4399– 4412, DOI: 10.1021/bi400079hGoogle Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXosFygsL8%253D&md5=e4130c42e5a6c34a1f9f15135222f6ceMultiple Pathways Promote Dynamical Coupling between Catalytic Domains in Escherichia coli Prolyl-tRNA SynthetaseJohnson, James M.; Sanford, Brianne L.; Strom, Alexander M.; Tadayon, Stephanie N.; Lehman, Brent P.; Zirbes, Arrianna M.; Bhattacharyya, Sudeep; Musier-Forsyth, Karin; Hati, SanchitaBiochemistry (2013), 52 (25), 4399-4412CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)Aminoacyl-tRNA synthetases are multidomain enzymes that catalyze covalent attachment of amino acids to their cognate tRNA. Cross-talk between functional domains is a prerequisite for this process. In this study, we investigate the mol. mechanism of site-to-site communication in Escherichia coli prolyl-tRNA synthetase (Ec ProRS). Earlier studies have demonstrated that evolutionarily conserved and/or co-evolved residues that are engaged in correlated motion are crit. for the propagation of functional conformational changes from one site to another in modular proteins. Here, mol. simulation and bioinformatics-based anal. were performed to identify dynamically coupled and evolutionarily constrained residues that form contiguous pathways of residue-residue interactions between the aminoacylation and editing domains of Ec ProRS. The results of this study suggest that multiple pathways exist between these two domains to maintain the dynamic coupling essential for enzyme function. Moreover, residues in these interaction networks are generally highly conserved. Site-directed changes of on-pathway residues have a significant impact on enzyme function and dynamics, suggesting that any perturbation along these pathways disrupts the native residue-residue interactions that are required for effective communication between the two functional domains. Free energy anal. revealed that communication between residues within a pathway and cross-talk between pathways are important for coordinating functions of different domains of Ec ProRS for efficient catalysis.
- 31Verlet, L. Computer “experiments” on classic fluids. I. Thermodynamical properties of Lennard-Jones molecules. Phys. Rev. 1967, 159, 98– 103, DOI: 10.1103/PhysRev.159.98Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaF2sXks1Orsrc%253D&md5=4dbb891023a4169feaa95513ce8075a2Computer "experiments" on classical fluids. I. Thermodynamical properties of Lennard-Jones moleculesVerlet, LoupPhysical Review (1967), 159 (1), 98-103CODEN: PHRVAO; ISSN:0031-899X.The equation of motion of a system of 864 particles interacting through a Lennard-Jones potential was integrated for various values of the temp. and d., relative, generally, to a fluid state. The equil. properties agree with the corresponding properties of Ar. The equil. state of Ar can be described through a 2-body potential.
- 32Phillips, J. C.; Braun, R.; Wang, W.; Gumbart, J.; Tajkhorshid, E.; Villa, E.; Chipot, C.; Skeel, R. D.; Kalé, L.; Schulten, K. Scalable molecular dynamics with NAMD. J. Comput. Chem. 2005, 26, 1781– 1802, DOI: 10.1002/jcc.20289Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXht1SlsbbJ&md5=189051128443b547f4300a1b8fb0e034Scalable molecular dynamics with NAMDPhillips, James C.; Braun, Rosemary; Wang, Wei; Gumbart, James; Tajkhorshid, Emad; Villa, Elizabeth; Chipot, Christophe; Skeel, Robert D.; Kale, Laxmikant; Schulten, KlausJournal of Computational Chemistry (2005), 26 (16), 1781-1802CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)NAMD is a parallel mol. dynamics code designed for high-performance simulation of large biomol. systems. NAMD scales to hundreds of processors on high-end parallel platforms, as well as tens of processors on low-cost commodity clusters, and also runs on individual desktop and laptop computers. NAMD works with AMBER and CHARMM potential functions, parameters, and file formats. This article, directed to novices as well as experts, first introduces concepts and methods used in the NAMD program, describing the classical mol. dynamics force field, equations of motion, and integration methods along with the efficient electrostatics evaluation algorithms employed and temp. and pressure controls used. Features for steering the simulation across barriers and for calcg. both alchem. and conformational free energy differences are presented. The motivations for and a roadmap to the internal design of NAMD, implemented in C++ and based on Charm++ parallel objects, are outlined. The factors affecting the serial and parallel performance of a simulation are discussed. Finally, typical NAMD use is illustrated with representative applications to a small, a medium, and a large biomol. system, highlighting particular features of NAMD, for example, the Tcl scripting language. The article also provides a list of the key features of NAMD and discusses the benefits of combining NAMD with the mol. graphics/sequence anal. software VMD and the grid computing/collab. software BioCoRE. NAMD is distributed free of charge with source code at www.ks.uiuc.edu.
- 33Geng, W.; Krasny, R. A treecode-accelerated boundary integral Poisson–Boltzmann solver for electrostatics of solvated biomolecules. J. Comput. Phys. 2013, 247, 62– 78, DOI: 10.1016/j.jcp.2013.03.056Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXosFyltbY%253D&md5=4959c5c5c74974ac8167c78a00042842A treecode-accelerated boundary integral Poisson-Boltzmann solver for electrostatics of solvated biomoleculesGeng, Weihua; Krasny, RobertJournal of Computational Physics (2013), 247 (), 62-78CODEN: JCTPAH; ISSN:0021-9991. (Elsevier Inc.)We present a treecode-accelerated boundary integral (TABI) solver for electrostatics of solvated biomols. described by the linear Poisson-Boltzmann equation. The method employs a well-conditioned boundary integral formulation for the electrostatic potential and its normal deriv. on the mol. surface. The surface is triangulated and the integral equations are discretized by centroid collocation. The linear system is solved by GMRES iteration and the matrix-vector product is carried out by a Cartesian treecode which reduces the cost from O(N2) to O(NlogN), where N is the no. of faces in the triangulation. The TABI solver is applied to compute the electrostatic solvation energy in two cases, the Kirkwood sphere and a solvated protein. We present the error, CPU time, and memory usage, and compare results for the Poisson-Boltzmann and Poisson equations. We show that the treecode approxn. error can be made smaller than the discretization error, and we compare two versions of the treecode, one with uniform clusters and one with non-uniform clusters adapted to the mol. surface. For the protein test case, we compare TABI results with those obtained using the grid-based APBS code, and we also present parallel TABI simulations using up to eight processors. We find that the TABI solver exhibits good serial and parallel performance combined with relatively simple implementation, efficient memory usage, and geometric adaptability.
- 34Lan, J.; Ge, J.; Yu, J.; Shan, S.; Zhou, H.; Fan, S.; Zhang, Q.; Shi, X.; Wang, Q.; Zhang, L.; Wang, X. Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature 2020, 581, 215– 220, DOI: 10.1038/s41586-020-2180-5Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXoslOqtL8%253D&md5=279c60143e8e5eb505457e0778baa8efStructure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptorLan, Jun; Ge, Jiwan; Yu, Jinfang; Shan, Sisi; Zhou, Huan; Fan, Shilong; Zhang, Qi; Shi, Xuanling; Wang, Qisheng; Zhang, Linqi; Wang, XinquanNature (London, United Kingdom) (2020), 581 (7807), 215-220CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Abstr.: A new and highly pathogenic coronavirus (severe acute respiratory syndrome coronavirus-2, SARS-CoV-2) caused an outbreak in Wuhan city, Hubei province, China, starting from Dec. 2019 that quickly spread nationwide and to other countries around the world1-3. Here, to better understand the initial step of infection at an at. level, we detd. the crystal structure of the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 bound to the cell receptor ACE2. The overall ACE2-binding mode of the SARS-CoV-2 RBD is nearly identical to that of the SARS-CoV RBD, which also uses ACE2 as the cell receptor4. Structural anal. identified residues in the SARS-CoV-2 RBD that are essential for ACE2 binding, the majority of which either are highly conserved or share similar side chain properties with those in the SARS-CoV RBD. Such similarity in structure and sequence strongly indicate convergent evolution between the SARS-CoV-2 and SARS-CoV RBDs for improved binding to ACE2, although SARS-CoV-2 does not cluster within SARS and SARS-related coronaviruses1-3,5. The epitopes of two SARS-CoV antibodies that target the RBD are also analyzed for binding to the SARS-CoV-2 RBD, providing insights into the future identification of cross-reactive antibodies.
- 35Madeira, F.; Park, Y. M.; Lee, J.; Buso, N.; Gur, T.; Madhusoodanan, N.; Basutkar, P.; Tivey, A. R. N.; Potter, S. C.; Finn, R. D.; Lopez, R. The EMBL-EBI search and sequence analysis tools APIs in 2019. Nucleic Acids Res. 2019, 47, W636– W641, DOI: 10.1093/nar/gkz268Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXktVyitbY%253D&md5=ece6310cea25d80c352cbff7ec5d0661The EMBL-EBI search and sequence analysis tools APIs in 2019Madeira, Fabio; Park, Young Mi; Lee, Joon; Buso, Nicola; Gur, Tamer; Madhusoodanan, Nandana; Basutkar, Prasad; Tivey, Adrian R. N.; Potter, Simon C.; Finn, Robert D.; Lopez, RodrigoNucleic Acids Research (2019), 47 (W1), W636-W641CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)The EMBL-EBI provides free access to popular bioinformatics sequence anal. applications as well as to a full-featured text search engine with powerful cross-referencing and data retrieval capabilities. Access to these services is provided via user-friendly web interfaces and via established RESTful and SOAP Web Services APIs (https://www.ebi.ac.uk/seqdb/confluence/display/JDSAT/EMBL-EBI+Web+Services+APIs+-+Data+Retrieval). Both systems have been developed with the same core principles that allow them to integrate an ever-increasing vol. of biol. data, making them an integral part of many popular data resources provided at the EMBL-EBI. Here, we describe the latest improvements made to the frameworks which enhance the interconnectivity between public EMBL-EBI resources and ultimately enhance biol. data discoverability, accessibility, interoperability and reusability.