Machine annealing-guided navigation of antihypertensive food peptide selectivity between human ACE N- and C-domains in structurally interacting diversity space.

  • Abstract
  • References
  • Citations
  • Similar Papers
Abstract
Translate article icon Translate Article Star icon
Take notes icon Take Notes

Human angiotensin-converting enzyme (ACE) is a well-established druggable target for the treatment of hypertension (HTN), which contains two structurally homologous but functionally distinct N- and C-domains. Selective inhibition of the C-domain primarily contributes to the antihypertensive efficiency and can be exploited as medicinal agents and functional additives for regulating blood pressure with high safety. In this study, we used a machine annealing (MA) strategy to guide the navigation of antihypertensive peptides (AHPs) in structurally interacting diversity space with the two ACE domains based on their crystal/modeled complex structures and an in-house protein-peptide affinity scoring function, aiming to optimize the peptide selectivity for C-domain over N-domain. The strategy generated a panel of theoretically designed AHP hits with a satisfactory C-over-N (C > N) selectivity profile, from which several hits were found to have a good C > N selectivity, which is roughly comparable with or even better than the BPPb, a natural C > N-selective ACE-inhibitory peptide. Structural analysis and comparison of domain-peptide noncovalent interaction patterns revealed that (i) longer peptides (>4 amino aids) generally exhibit stronger selectivity than shorter peptides (<4 amino aids), (ii) peptide sequence can be divided into two, section I (including peptide C-terminal region) and section II (including peptide middle and N-terminal regions); the former contributes to both peptide affinity (primarily) and selectivity (secondarily), while the latter is almost only responsible for peptide selectivity, and (iii) charged/polar amino acids confer to peptide selectivity relative to hydrophobic/nonpolar amino acids (that confer to peptide affinity).

ReferencesShowing 10 of 67 papers
  • Cite Count Icon 87
  • 10.1016/j.foodchem.2006.06.026
Modelling relationship between angiotensin-(I)-converting enzyme inhibition and the bitter taste of peptides
  • Aug 24, 2006
  • Food Chemistry
  • Are Hugo Pripp + 1 more

  • Cite Count Icon 137
  • 10.1021/acs.jcim.6b00673
Targeting Self-Binding Peptides as a Novel Strategy To Regulate Protein Activity and Function: A Case Study on the Proto-oncogene Tyrosine Protein Kinase c-Src.
  • Apr 3, 2017
  • Journal of Chemical Information and Modeling
  • Zhengya Bai + 4 more

  • Open Access Icon
  • Cite Count Icon 2537
  • 10.1016/j.drudis.2014.10.003
Peptide therapeutics: current status and future directions
  • Oct 16, 2014
  • Drug Discovery Today
  • Keld Fosgerau + 1 more

  • Open Access Icon
  • Cite Count Icon 216
  • 10.1146/annurev-biophys-052118-115325
Generalized Born Implicit Solvent Models for Biomolecules.
  • Mar 11, 2019
  • Annual review of biophysics
  • Alexey V Onufriev + 1 more

  • Cite Count Icon 923
  • 10.1002/minf.201100135
Free Energy Calculations by the Molecular Mechanics Poisson-Boltzmann Surface Area Method.
  • Jan 10, 2012
  • Molecular Informatics
  • Nadine Homeyer + 1 more

  • Cite Count Icon 64
  • 10.1002/jcc.21109
2D depiction of nonbonding interactions for protein complexes
  • Oct 22, 2008
  • Journal of Computational Chemistry
  • Peng Zhou + 2 more

  • Cite Count Icon 14
  • 10.1007/bf02138854
Inhibition of angiotensin I converting enzyme and kininase in rabbit plasma by bradykinin potentiating peptide B (Pyr-Gly-Leu-Pro-Arg-Pro-Lys-Ile-Pro-Pro).
  • Sep 1, 1971
  • Experientia
  • E Ueda + 3 more

  • Cite Count Icon 1402
  • 10.1006/jmbi.1998.2401
Asparagine and glutamine: using hydrogen atom contacts in the choice of side-chain amide orientation
  • Jan 1, 1999
  • Journal of Molecular Biology
  • J.Michael Word + 3 more

  • Open Access Icon
  • Cite Count Icon 36577
  • 10.1093/nar/28.1.235
The Protein Data Bank.
  • Jan 1, 2000
  • Nucleic Acids Research
  • H M Berman

  • Cite Count Icon 26
  • 10.1016/j.compbiolchem.2015.01.001
Structure-based grafting and identification of kinase–inhibitors to target mTOR signaling pathway as potential therapeutics for glioblastoma
  • Jan 9, 2015
  • Computational Biology and Chemistry
  • Yu-Hui Cui + 3 more

CitationsShowing 10 of 16 papers
  • Research Article
  • 10.1142/s273741652450087x
Deciphering the Selective Targeting of Noncovalent, Wild Type-sparing, and ATP-competitive Tyrosine Kinase Inhibitors to EGFR T790M Gatekeeper Mutant
  • Jan 22, 2025
  • Journal of Computational Biophysics and Chemistry
  • Shu Meng + 3 more

Various tyrosine kinase inhibitors (TKIs) have been developed to target human epidermal growth factor receptor (EGFR) for cancer therapy. However, many patients treated with first-line TKIs are clinically observed to eventually establish a gatekeeper T790M mutation in the ATP-binding site of the EGFR kinase domain, which is primarily responsible for acquired drug resistance to cancers. Over the past decades, a number of noncovalent, wild-type-sparing and ATP-competitive inhibitors (NWAIs) were reported to selectively target the T790M mutant over wild-type kinase, which are independent of the traditional inhibitor classification system that categorizes EGFR TKIs into four generations. Here, we systematically investigated the intermolecular interaction of wild-type EGFR (EGFRWT) and its T790M mutant (EGFRT790M) with 15 existing NWAI inhibitors, paying attention to the structural and energetic responses of inhibitor ligands to the gatekeeper mutation. It was revealed that the NWAIs can be typed into three classes I, II and III, which can form S[Formula: see text] interactions, hydrophobic (van der Waals) contacts and weak hydrogen (halogen) bonding with the side-chain thioether moiety of the mutant Met790 residue, respectively, thus conferring additional affinity and specificity to inhibitor ligands upon the T790M mutation. In addition, we further performed 2D-chemical similarity search to identify new class I NWAIs, from which two Staurosporine analogs (i.e. UCN01 and ZHD0501) were identified to have a good selectivity for EGFRT790M over EGFRWT. They can be exploited as promising leading chemical scaffolds to further develop potent, selective, wild-type-sparing NWAI inhibitors of EGFRT790M gatekeeper mutant.

  • Research Article
  • Cite Count Icon 2
  • 10.1142/s2737416523500394
Computational Analysis and Rational Design of N-Substituting Perturbation on the Affinity and Specificity of Pediatric Fyn SH3–Peptoid Interaction
  • Jul 26, 2023
  • Journal of Computational Biophysics and Chemistry
  • Qin Liu + 3 more

Human nonreceptor tyrosine kinase (TK) Fyn is implicated in various cellular processes and has been exploited as a sophisticated druggable target of pediatric T-cell lymphoma by blocking its kinase domain with small-molecule inhibitors or disrupting its regulatory SH2 and SH3 domains with peptidic inhibitors. In this study, the proline-heavy 9P1Y-peptide was found as a good binder of Fyn SH3 domain, of which the chemical diversity space was extended by replacing its proline residues with a variety of N-substituted amino acids, since N-substitution can mimic the side-chain location and shape of proline. A systematic single-point N-substituting perturbation profile (SSNPP) for 9P1Y-peptide was created theoretically, from which a systematic combinatorial peptoid library (SCPL) was then generated by introducing favorable N-substituted to different proline residue positions of the peptide. The computational design was further substantiated by fluorescence spectroscopy assays to identify five promising peptoid hits in the SCPL as potential Fyn SH3 binders. In particular, the designed peptoid Ptoid2 exhibited a good affinity to Fyn SH3 domain ([Formula: see text]M) and a moderate selectivity for Fyn over Lyn (3.8-fold), which are improved substantially from the natural 9P1Y-peptide. Molecular mechanism underlying the N-substituting perturbation effect was also examined in detail.

  • Research Article
  • Cite Count Icon 1
  • 10.1016/j.jmgm.2023.108601
N-substituting perturbation on the interaction affinity and recognition specificity between rheumatic immune-related Abl SH3 domain and its peptoid ligands
  • Aug 17, 2023
  • Journal of Molecular Graphics and Modelling
  • Xiaomin Tang + 3 more

N-substituting perturbation on the interaction affinity and recognition specificity between rheumatic immune-related Abl SH3 domain and its peptoid ligands

  • Research Article
  • Cite Count Icon 1
  • 10.1142/s2737416524500236
Computational Design and Experimental Validation of an Orthogonal X-bond/H-bond System to Improve the Binding Affinity and Recognition Specificity between Anesthetic Protein Kinase Cι and its Pseudosubstrate Peptide
  • Jul 23, 2024
  • Journal of Computational Biophysics and Chemistry
  • Rong Kuang + 6 more

Protein kinase C isoform [Formula: see text] (PKC[Formula: see text] is an essential regulator of diverse neurobiological signaling pathways, which has been implicated in general anesthetic action and a variety of nervous neoplasms. Traditional efforts have been primarily addressed on the development of small-molecule inhibitors to target the catalytic or regulatory domain of this kinase. Instead, we herein reported a rational chemical modification of Par3 pseudosubstrate peptide to target the kinase domain with improved affinity and specificity. Structural and energetic analysis suggested that the Par3 peptide can be divided into two binding-independent sections; they are separately anchored to the substrate-binding pocket of PKC[Formula: see text] with two aromatic anchor residues Phe818 and Phe823. The side-chain phenyl group of these two aromatic residues was systematically substituted by different halogen moieties at [Formula: see text]-, [Formula: see text]- and [Formula: see text]-positions of the phenyl group. It is revealed that the [Formula: see text]-bromination of Par3 Phe818 and the [Formula: see text]-iodinization of Par3 Phe823 residue can separately form two geometrically and energetically satisfactory orthogonal halogen (X)-bond/hydrogen (H)-bond [ortX/H] motifs across Par3 complex interface with PKC[Formula: see text]; they co-define a so-called ortX/H system and were demonstrated to improve both the binding affinity and recognition specificity of the kinase–peptide interaction by integrating in silico analysis and in vitro assay. The peptide affinity promotes 23.5-fold and 6.6-fold upon the formation of ortX/H motif-I and motif-II, respectively, which further increases to 43.5-fold by forming the complete ortX/H system, indicating cooperativity between the two motifs in the system. In addition, we also demonstrated that the ortX/H system can considerably enhance the peptide selectivity for its cognate PKC[Formula: see text] and noncogante isoforms PKC[Formula: see text] and PKC[Formula: see text].

  • Research Article
  • Cite Count Icon 2
  • 10.1080/07391102.2023.2272344
Dynamics simulation, energetics calculation and experimental analysis of the intermolecular interaction between human neonatal ABL SH3 domain and its N-substituted peptoid ligands
  • Oct 16, 2023
  • Journal of Biomolecular Structure and Dynamics
  • Shijian Zhao + 3 more

Non-receptor tyrosine kinase of neonatal ABL (nABL) is distributed in the nucleus and cytoplasm of proliferating cells in embryo and neonate, and has been implicated in the pathogenesis of neonatal leukemia and other hematological diseases. The kinase contains a regulatory Src homology 3 (SH3) domain that can specifically recognize proline-rich peptide segments on its partner protein surface. In this study, we systematically investigated the N-substitution effect on the binding of an empirically designed proline-rich peptide p9 to nABL SH3 domain by integrating dynamics simulations, energetics calculations and fluorescence affinity assays. The p9 is an almost all proline-composed decapeptide, with only a sole tyrosine at its residue 4, which has been found to bind nABL SH3 domain at a micromolar level in a class I mode. Here, the non-key residues of p9 peptide were independently replaced by various N-substituted amino acids to create a systematic N-substitution profile, from which we can identify those favorable, neutral and unfavorable substitutions at each peptide residue. On this basis a combinatorial peptoid library was rationally designed by systematically combining the favorable N-substituted amino acids at non-key residues of p9 peptide, thus resulting in a number of its peptoid counterparts. The binding affinity of top peptoid hits was observed to be comparable with or improved moderately relative to p9 peptide, with Kd ranging between 3.1 and 76 μM. Structural analysis revealed that the peptoids can be divided into exposed, polar and hydrophobic regions from N- to C-termini, in which the polar and hydrophobic regions confer specificity and stability to the domain–peptoid interaction, respectively. In addition, a designed peptoid was also observed to exhibit 5.3-fold SH3-selectivity for nABL over cSRC, suggesting that the N-substitution can be used to improve not only binding affinity but also recognition specificity of SH3 binders. Communicated by Ramaswamy H. Sarma

  • Research Article
  • 10.1007/s00249-024-01729-5
Rational design of potent phosphopeptide binders to endocrine Snk PBD domain by integrating machine learning optimization, molecular dynamics simulation, binding energetics rescoring, and in vitro affinity assay.
  • Nov 29, 2024
  • European biophysics journal : EBJ
  • Zhaohui Wang + 4 more

Human Snk is an evolutionarily conserved serine/threonine kinase essential for the maintenance of endocrine stability. The protein consists of a N-terminal catalytic domain and a C-terminal polo-box domain (PBD) that determines subcellular localization and substrate specificity. Here, an integrated strategy is described to explore the vast structural diversity space of Snk PBD-binding phosphopeptides at a molecular level using machine learning modeling, annealing optimization, dynamics simulation, and energetics rescoring, focusing on the recognition specificity and motif preference of the Snk PBD domain. We further performed a systematic rational design of potent phosphopeptide ligands for the domain based on the harvested knowledge, from which a few potent binders were also confirmed by fluorescence-based assays. A phosphopeptide PP17 was designed as a good binder with affinity improvement by 6.7-fold relative to the control PP0, while the other three designed phosphopeptides PP7, PP13, and PP15 exhibit a comparable potency with PP0. In addition, a basic recognition motif that divides potent Snk PBD-binding sequences into four residue blocks was defined, namely [Χ-5Χ-4]block1-[Ω-3Ω-2Ω-1]block2-[pS0/pT0]block3-[Ψ+1]block4, where the X represents any amino acid, Ω indicates polar amino acid, Ψ denotes hydrophobic amino acid, and pS0/pT0 is the anchor phosphoserine/phosphothreonine at reference residue position 0.

  • Research Article
  • 10.1002/jmr.3111
TAZ-hTrap: A Rationally Designed, Disulfide-Stapled Tead Helical Hairpin Trap to Selectively Capture Hippo Signaling Taz With Potent Antigynecological Tumor Activity.
  • Dec 3, 2024
  • Journal of molecular recognition : JMR
  • Bin Tang + 2 more

Transcriptional enhanced associate domain (Tead)-mediated Hippo signaling pathway regulates diverse physiological processes; its dysfunction has been implicated in an increasing number of human gynecological cancers. The transcriptional coactivator with PDZ-binding motif (Taz) binds to and then activates Tead through forming a three-helix bundle (THB) at their complex interface. The THB is defined by a double-helical hairpin from Tead and a single α-helix from Taz, serving as the key interaction hotspot between Tead and Taz. In the present study, the helical hairpin was derived from Tead protein to generate a hairpin segment, which is a 25-mer polypeptide consisting of a longer helical arm-1 and a shorter helical arm-2 as well as a flexible loop linker between them. Dynamics simulation and energetics characterization revealed that the hairpin peptide is intrinsically disordered when splitting from its protein context, thus incurring a large entropy penalty upon binding to Taz α-helix. A disulfide bridge was introduced across the two helical arms of hairpin peptide to obtain a strong binder termed TAZ-hTrap, which can maintain in a considerably structured, native-like conformation in unbound state, and the entropy penalty was minimized by disulfide stapling to effectively improve its affinity toward the α-helix. These computational findings can be further substantiated by circular dichroism and fluorescence polarization at molecular level, and viability assay also observed a potent cytotoxic effect on diverse human gynecological tumors at cellular level. In addition, we further demonstrated that the TAZ-hTrap has a good selectivity for its cognate Taz over other noncognate proteins that share a high conservation with the Taz α-helix.

  • Research Article
  • 10.1002/jmr.3070
Structure-based improvement of the binding affinity and recognition specificity of peptide competitors to target pediatric IL-5R/IL-5 interaction by gluing halogen bonds at their complex interface.
  • Nov 21, 2023
  • Journal of molecular recognition : JMR
  • Peipei Chu + 5 more

Human interleukin-5 (IL-5) cytokine mediates the development of eosinophils and is involved in a variety of immune inflammatory responses that play a major role in the pathogenesis of childhood asthma, leukemia, and other pediatric allergic diseases. The immunomodulatory cytokine functions by binding to its cognate cell surface receptor IL-5R in a sheet-by-sheet manner, which can be conformationally mimicked and competitively disrupted by a double-stranded cyclic AF18748 peptide. In this study, we systematically examined the co-crystallized complex structure of human IL-5R with AF18748 peptide and rationally designed a halogen bond to glue at the protein-peptide complex interface by substituting the indole moiety of AF18748 Trp13 residue with a halogen atom (X = F, Cl, Br, or I). High-level theoretical calculations imparted presence of the halogen bond between the oxygen atom (O) of IL-5R Glu58 backbone and the halogen atom (X) of AF18748 Trp13 side chain. Experimental assays confirmed that the halogen bond can promote peptide binding moderately or considerably. More importantly, the halogen bond not only enhances peptide affinity to IL-5R, but also improves peptide selectivity for its cognate IL-5R over other noncognate IL-R proteins. As might be expected, the affinity and selectivity conferred by halogen bond increase consistently in the order: H < F < Cl < Br < I. Structural modeling revealed that the halogen bond plus its vicinal π-cation-π stacking co-define a ringed noncovalent system at the complex interface, which involves a synergistic effect to effectively improve the peptide binding potency and recognition specificity.

  • Research Article
  • Cite Count Icon 2
  • 10.1016/j.foodchem.2024.139540
Screening and rational identification of a novel angiotensin-converting enzyme C-domain inhibitory peptide from Fabaceae food peptide library
  • May 6, 2024
  • Food chemistry
  • Tin-Yun Ho + 6 more

Screening and rational identification of a novel angiotensin-converting enzyme C-domain inhibitory peptide from Fabaceae food peptide library

  • Research Article
  • 10.1142/s2737416523500576
Computational Design, Combinatorial Screening and Experimental Analysis of Lung Cancer EGFRVIII-Binding Peptides
  • Oct 19, 2023
  • Journal of Computational Biophysics and Chemistry
  • Dongyun Gao + 7 more

Human epidermal growth factor receptor mutation variant III ([Formula: see text] is a cancer-specific cell surface oncogenic marker and has been observed to be widely involved in the formation, progression and metastasis of lung cancer and some other tumors. Previously, a massive quantity of [Formula: see text]-binding peptides were enriched via random phage display (RPD) targeted against the protein. In this study, we reported rational discovery of 12-mer peptides with high affinity to [Formula: see text] and strong selectivity for [Formula: see text] over wild-type EGFR ([Formula: see text]. A combinatorial peptide library was designed to target [Formula: see text] based on over ten thousands of known [Formula: see text] binders enriched from RPD analysis, and a virtual high-throughput screening protocol was then systematically performed against the library to derive those potential candidates, which were further examined rigorously at structural and energetic levels to identify few promising hits. Anisotropy binding assays were carried out to substantiate the computational findings. Consequently, eight 12-mer peptides were designed as effective binders that can target the [Formula: see text] extracellular subdomain 3 (SD3). In particular, two potent peptides (T1: FLHRYEIVTSYF and T3: FLQKYEWNTSYW) were found to have a high affinity to [Formula: see text] and a good selectivity for [Formula: see text] over [Formula: see text]. Structural analysis revealed that the peptide-binding site can be divided into hydrophobic, polar and mixed regions, which correspond to the nonpolar [Formula: see text]-terminal section, polar/charged middle section and hybrid C-terminal section of the peptide. The peptide selectivity originated from the middle section, which can form a different hydrogen bond network between the two proteins upon the mutating perturbation, whereas the N- and C-terminal sections are primarily responsible for the peptide stability but not specificity.

Similar Papers
  • Research Article
  • Cite Count Icon 975
  • 10.1038/sj.emboj.7600640
Receptor and viral determinants of SARS-coronavirus adaptation to human ACE2.
  • Mar 24, 2005
  • The EMBO Journal
  • Wenhui Li + 15 more

Human angiotensin-converting enzyme 2 (ACE2) is a functional receptor for SARS coronavirus (SARS-CoV). Here we identify the SARS-CoV spike (S)-protein-binding site on ACE2. We also compare S proteins of SARS-CoV isolated during the 2002–2003 SARS outbreak and during the much less severe 2003–2004 outbreak, and from palm civets, a possible source of SARS-CoV found in humans. All three S proteins bound to and utilized palm-civet ACE2 efficiently, but the latter two S proteins utilized human ACE2 markedly less efficiently than did the S protein obtained during the earlier human outbreak. The lower affinity of these S proteins could be complemented by altering specific residues within the S-protein-binding site of human ACE2 to those of civet ACE2, or by altering S-protein residues 479 and 487 to residues conserved during the 2002–2003 outbreak. Collectively, these data describe molecular interactions important to the adaptation of SARS-CoV to human cells, and provide insight into the severity of the 2002–2003 SARS epidemic.

  • Research Article
  • Cite Count Icon 109
  • 10.1038/ki.2012.83
Podocyte-specific overexpression of human angiotensin-converting enzyme 2 attenuates diabetic nephropathy in mice
  • Aug 1, 2012
  • Kidney International
  • Renisha Nadarajah + 9 more

Podocyte-specific overexpression of human angiotensin-converting enzyme 2 attenuates diabetic nephropathy in mice

  • Research Article
  • 10.1161/res.111.suppl_1.a169
Abstract 169: Angiotensin-Converting Enzyme 2: Species-Specific Features and Pharmacologic Characterization by RAS Fingerprinting
  • Aug 3, 2012
  • Circulation Research
  • Marko Poglitsch + 4 more

The carboxypeptidase Angiotensin Converting Enzyme 2 (ACE2) is a crucial player of the Renin-Angiotensin-System (RAS) as it balances the ratio between vasoconstrictive and fibrotic Angiotensin II and vasodilative and anti-fibrotic Angiotensin 1-7. This balance is affected by ACE2 either by direct conversion of Angiotensin II to Angiotensin 1-7, or by cleavage of Angiotensin I to Angiotensin 1-9, which is further converted to Ang 1-7 by Angiotensin Converting Enzyme (ACE) or Neutral Endopeptidase (NEP). An increased activity of ACE2 leads to a shift of the RAS towards an Angiotensin 1-7 dominated pattern, which was shown to be protective in various models of cardiovascular and fibrotic diseases. We performed a comparative pharmacologic characterization of recombinant murine and human ACE2 using natural peptide substrates for the evaluation of the substrate specific ACE2 activity in whole blood. Furthermore, we investigated the impact of ACE2 activators and inhibitors on murine and human ACE2 in whole blood using an LC-MS/MS based approach allowing the simultaneous quantification of 10 different angiotensin peptides (RAS-Fingerprinting). The investigation of inhibitors and activators of ACE2 using our experimental setting revealed the presence of species-specific enzymatic features, which was indicated by differences between the human and murine enzyme regarding the susceptibility to pharmacologic manipulation and the specificity for different endogenous peptide substrates. Our studies demonstrate that sequence diversity observed between recombinant human and murine ACE2 significantly affects substrate specificity and pharmacologic enzyme properties. The utilization of Angiotensin I by ACE2 generates Angiotensin 1-7 via Angiotensin 1-9 and represents a pathway of alternative RAS activation, which might have been systematically underestimated in previous studies. The strongly reduced affinity of murine ACE2 for Angiotensin I results in a lack of production of Angiotensin 1-7 via this pathway, which might be of particular importance during anti-hypertensive treatments with ACE inhibitors, where the formation of Angiotensin II is prevented while increased levels of Angiotensin I are observed in circulation.

  • PDF Download Icon
  • Research Article
  • Cite Count Icon 244
  • 10.1074/jbc.m111.325803
Mechanisms of Host Receptor Adaptation by Severe Acute Respiratory Syndrome Coronavirus
  • Mar 1, 2012
  • Journal of Biological Chemistry
  • Kailang Wu + 4 more

The severe acute respiratory syndrome coronavirus (SARS-CoV) from palm civets has twice evolved the capacity to infect humans by gaining binding affinity for human receptor angiotensin-converting enzyme 2 (ACE2). Numerous mutations have been identified in the receptor-binding domain (RBD) of different SARS-CoV strains isolated from humans or civets. Why these mutations were naturally selected or how SARS-CoV evolved to adapt to different host receptors has been poorly understood, presenting evolutionary and epidemic conundrums. In this study, we investigated the impact of these mutations on receptor recognition, an important determinant of SARS-CoV infection and pathogenesis. Using a combination of biochemical, functional, and crystallographic approaches, we elucidated the molecular and structural mechanisms of each of these naturally selected RBD mutations. These mutations either strengthen favorable interactions or reduce unfavorable interactions with two virus-binding hot spots on ACE2, and by doing so, they enhance viral interactions with either human (hACE2) or civet (cACE2) ACE2. Therefore, these mutations were viral adaptations to either hACE2 or cACE2. To corroborate the above analysis, we designed and characterized two optimized RBDs. The human-optimized RBD contains all of the hACE2-adapted residues (Phe-442, Phe-472, Asn-479, Asp-480, and Thr-487) and possesses exceptionally high affinity for hACE2 but relative low affinity for cACE2. The civet-optimized RBD contains all of the cACE2-adapted residues (Tyr-442, Pro-472, Arg-479, Gly-480, and Thr-487) and possesses exceptionally high affinity for cACE2 and also substantial affinity for hACE2. These results not only illustrate the detailed mechanisms of host receptor adaptation by SARS-CoV but also provide a molecular and structural basis for tracking future SARS-CoV evolution in animals.

  • Research Article
  • Cite Count Icon 24
  • 10.4049/jimmunol.157.1.213
Selection and binding of peptides to human transporters associated with antigen processing and rat cim-a and -b.
  • Jul 1, 1996
  • The Journal of Immunology
  • P Wang + 2 more

Cytotoxic T lymphocytes recognize antigenic peptides presented by MHC class I molecules. The peptides are generated in the cytosol by proteasomes, and probably also other proteases, and are then translocated into the endoplasmic reticulum (ER) lumen. The transporters associated with Ag processing (TAP) are key molecules for transporting peptides from the cytosol to the lumen of the ER. Using semipermeabilized cells, TAP-dependent peptide translocation was demonstrated, and the selectivity of peptide translocation was based on the carboxyl-terminal amino acid of peptides. We have examined peptide binding proteins in the ER membrane and the selection of peptides for binding to TAP by using a panel of peptides of different sequences and carboxyl-termini as well as peptides containing D amino acids. Peptides bound to TAP molecules in the absence of ATP. The presence of ATP induced binding of peptides to two additional membrane proteins (58 and 43 kDa). The selection of peptides by TAP molecules was based on peptide sequence and the carboxyl-terminal amino acid. Peptides containing D amino acid did not bind to TAP molecules. Rat cim-a and -b selected peptides differently, and selection was not only dependent on the carboxyl-terminal residue of the peptide, but included an influence of the peptide sequence. The different off-rates after peptide binding to TAP, indicated a dual binding step of peptide to TAP. ATP regulated the off-rate of peptides at a high affinity binding step. Our results demonstrate that the binding of peptides to TAP molecules is specific and most likely involves a multiple step pathway.

  • Peer Review Report
  • 10.7554/elife.61552.sa1
Decision letter: Inhibition of SARS-CoV-2 viral entry upon blocking N- and O-glycan elaboration
  • Sep 3, 2020
  • Morten Thaysen-Andersen + 1 more

Decision letter: Inhibition of SARS-CoV-2 viral entry upon blocking N- and O-glycan elaboration

  • Research Article
  • Cite Count Icon 50
  • 10.1053/j.gastro.2020.09.042
Angiotensin-converting Enzyme 2–containing Small Extracellular Vesicles and Exomeres Bind the Severe Acute Respiratory Syndrome Coronavirus 2 Spike Protein
  • Oct 3, 2020
  • Gastroenterology
  • Qin Zhang + 5 more

Angiotensin-converting Enzyme 2–containing Small Extracellular Vesicles and Exomeres Bind the Severe Acute Respiratory Syndrome Coronavirus 2 Spike Protein

  • Research Article
  • 10.1124/jpet.122.531940
Development of Humanized ACE2 Mouse and Rat Models for COVID-19 Research
  • May 18, 2023
  • The Journal of Pharmacology and Experimental Therapeutics
  • Hongmei Jiang + 11 more

<b>Abstract ID 53194</b> <b>Poster Board 252</b> Over the last 3 years, the COVID-19 pandemic has severely affected human lives and the global economy. The virus causing COVID-19 is called Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). SARS-CoV-2 infects host cells by the binding of its spike protein to the cellular surface protein angiotensin-converting enzyme 2 (ACE2). The predicted 29 amino acid residues of ACE2 that interact with SARS-CoV-2 spike protein receptor binding domain (RBD) vary between human ACE2 and mouse or rat ACE2. Therefore, wild type mice and rats show lower SARS-CoV-2 infection rate and mild symptoms compared to what is seen in humans. Small animal models that recapitulate human COVID-19 disease are urgently needed for better understanding the transmission and therapeutic measurement. Currently, scientists use either mouse-adapted SAS-CoV-2 (SAS-CoV-2 MA) models or random transgenic mouse models that artificially express human ACE2 under the control of cytokeratin 18 promoter or a constitutive promoter. SAS-CoV-2 MA may not completely reflect all aspects of the original human-tropic SAS-CoV-2 and the current transgenic human ACE2 mouse models typically have high mortality rate caused by neuroinvasion and encephalitis due to very high human ACE2 expression. To overcome these limitations, we have developed humanized ACE2 mouse and rat models using CRISPR-Cas9. Specifically, we inserted a ∼3kb human ACE2 cDNA cassette into the mouse and rat Ace2 gene loci to ensure that human ACE2 expression is under the control of rodent Ace2 promoter and regulatory elements, while simultaneously disabling the rodent Ace2 gene. To accomplish this, CRISPR gRNAs targeting close to the translation initiation site of Ace2 were screened in cultured mouse and rat cells. Then CRISPR/Cas9 complex and donor DNA were subsequently microinjected into one-cell stage embryos which were subsequently implanted into pseudo pregnant females. Resulting pups were screened for correct knockin by junction PCR and insert PCR, and the PCR products were Sanger sequenced. Targeted Locus Amplification (TLA) further confirmed the integration sites and transgene sequence. RT-qPCR and Western blot analysis data showed that, in our models, human ACE2 is expressed in tissues expressing endogenous Ace2 (such as lung, kidney, and GI tract), while rodent endogenous Ace2 is absent from these tissues. Further breeding data indicated that both hemizygous and homozygous humanized ACE2 animals appear to be normal and fertile. Most importantly, animals displayed symptoms after infection with SARS-CoV-2. In summary, these data suggest that our novel humanized ACE2 models can be valuable for COVID-19 research.

  • Research Article
  • 10.1096/fasebj.2022.36.s1.r2068
Development of humanized ACE2 mouse and rat models for COVID‐19 research
  • May 1, 2022
  • The FASEB Journal
  • Hongmei Jiang + 9 more

To date (17 November 2021), there has been more than 254 million confirmed cases of COVID‐19, and more than 5 million death globally (World Health Organization. https://covid19.who.int/). The virus causing COVID‐19 is called Severe Acute Respiratory Syndrome Coronavirus 2 (SARS‐CoV‐2). SARS‐CoV‐2 infects host cells by the binding of its spike protein to the cellular surface protein angiotensin‐converting enzyme 2 (ACE2). The predicted 29 amino acid residues of ACE2 that interact with SARS‐CoV‐2 spike protein receptor binding domain (RBD) vary between human ACE2 and mouse or rat ACE2. Therefore, wildtype mice and rats show lower SARS‐CoV‐2 infection rate and mild symptoms compared to what is seen in humans. Small animal models that recapitulate human COVID‐19 disease are urgently needed for better understanding the transmission and therapeutic measurement. Currently, scientists use either mouse‐adapted SAS‐CoV‐2 (SAS‐CoV‐2 MA) models or random transgenic mouse models that artificially express human ACE2 under the control of cytokeratin 18 promoter or a constitutive promoter. SAS‐CoV‐2 MA may not completely reflect all aspects of the original human‐tropic SAS‐CoV‐2 and the current transgenic human ACE2 mouse models typically have high mortality rate caused by neuroinvasion and encephalitis due to very high human ACE2 expression. To overcome these limitations, we have developed humanized ACE2 mouse and rat models using CRISPR‐Cas9. Specifically, we inserted a ~3kb human ACE2 cDNA cassette into the mouse and rat Ace2 gene loci to ensure that human ACE2 expression is under the control of rodent Ace2 promoter and regulatory elements, while simultaneously disabling the rodent Ace2 gene. To accomplish this, CRISPR gRNAs targeting close to the translation initiation site of Ace2 were screened in cultured mouse and rat cells. Then CRISPR/Cas9 complex and donor DNA were subsequently microinjected into one‐cell stage embryos which were subsequently implanted into pseudo pregnant females. Resulting pups were screened for correct knockin by junction PCR and insert PCR, and the PCR products were Sanger sequenced. Targeted Locus Amplification (TLA) further confirmed the integration sites and transgene sequence. RT‐qPCR and Western blot analysis data showed that, in our models, human ACE2 is expressed in tissues expressing endogenous Ace2 (such as lung, kidney, and GI tract), while rodent endogenous Ace2 is absent from these tissues. Further breeding data indicated that both hemizygous and homozygous humanized ACE2 animals appear to be normal and fertile. Most importantly, animals displayed symptoms after infection with SARS‐CoV‐2. In summary, these data suggest that our humanized ACE2 models can be valuable for COVID‐19 research.

  • Research Article
  • Cite Count Icon 69
  • 10.1152/ajpregu.00339.2011
Species-specific inhibitor sensitivity of angiotensin-converting enzyme 2 (ACE2) and its implication for ACE2 activity assays
  • Aug 31, 2011
  • American Journal of Physiology-Regulatory, Integrative and Comparative Physiology
  • Kim Brint Pedersen + 4 more

Angiotensin-converting enzyme 2 (ACE2) is a component of the renin-angiotensin system, and its expression and activity have been shown to be reduced in cardiovascular diseases. Enzymatic activity of ACE2 is commonly measured by hydrolysis of quenched fluorescent substrates in the absence or presence of an ACE2-specific inhibitor, such as the commercially available inhibitor DX600. Whereas recombinant human ACE2 is readily detected in mouse tissues using 1 μM DX600 at pH 7.5, the endogenous ACE2 activity in mouse tissues is barely detectable. We compared human, mouse, and rat ACE2 overexpressed in cell lines for their sensitivity to inhibition by DX600. ACE2 from all three species could be inhibited by DX600, but the half maximal inhibitory concentration (IC(50)) for human ACE2 was much lower (78-fold) than for rodent ACE2. Following optimization of pH, substrate concentration, and antagonist concentration, rat and mouse ACE2 expressed in a cell line could be accurately quantified with 10 μM DX600 (>95% inhibition) but not with 1 μM DX600 (<75% inhibition). Validation that the optimized method robustly quantifies ACE2 in mouse tissues (kidney, brain, heart, and plasma) was performed using wild-type and ACE2 knockout mice. This study provides a reliable method for measuring human, as well as endogenous ACE2 activity in rodents. Our data underscore the importance of validating the effect of DX600 on ACE2 from each particular species at the experimental conditions employed.

  • Research Article
  • Cite Count Icon 23
  • 10.1007/s40203-020-00055-w
Peptide modelling and screening against human ACE2 and spike glycoprotein RBD of SARS-CoV-2
  • Nov 9, 2020
  • In Silico Pharmacology
  • Shravan B Rathod + 5 more

Outbreak of Coronavirus Disease 2019 (COVID-19) has become a great challenge for scientific community globally. Virus enters cell through spike glycoprotein fusion with ACE2 (Angiotensin-Converting Enzyme 2) human receptor. Hence, spike glycoprotein of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a potential target for diagnostics, vaccines, and antibodies. Also, virus entry can be prevented by blocking ACE2 thus, ACE2 can be considered potential target for therapeutics. As being highly specific, safe and efficacious, peptides hold their place in therapeutics. In present study, we retrieved sequence of 70 peptides from Antiviral Peptide Database (AVPdb), modelled them using 3D structure predicting web tool and docked them with receptor binding domain (RBD) of spike protein and human host receptor ACE2 using peptide-protein docking. It was observed that peptides have more affinity towards ACE2 in comparison with spike RBD. Interestingly it was noticed that most of the peptides bind to RBM (residue binding motif) which is responsible for ACE2 binding at the interface of RBD while, for ACE2, peptides prefer to bind the core cavity rather than RBD binding interface. To further investigate how peptides at the interface of RBD or ACE2 alter the binding between RBD and ACE2, protein–protein docking of RBD and ACE2 with and without peptides was performed. Peptides, AVP0671 at RBD and AVP1244 at ACE2 interfaces significantly reduce the binding affinity and change the orientation of RBD and ACE2 binding. This finding suggests that peptides can be used as a drug to inhibit virus entry in cells to stop COVID-19 pandemic in the future after experimental evidences.Electronic supplementary materialThe online version of this article (10.1007/s40203-020-00055-w) contains supplementary material, which is available to authorized users.

  • Discussion
  • Cite Count Icon 24
  • 10.1016/j.cell.2022.05.004
Evidence in favor of the essentiality of human cell membrane-bound ACE2 and against soluble ACE2 for SARS-CoV-2 infectivity
  • May 1, 2022
  • Cell
  • Daniel Batlle + 10 more

Evidence in favor of the essentiality of human cell membrane-bound ACE2 and against soluble ACE2 for SARS-CoV-2 infectivity

  • PDF Download Icon
  • Research Article
  • Cite Count Icon 208
  • 10.1038/s41598-020-74715-4
Molecular docking study of potential phytochemicals and their effects on the complex of SARS-CoV2 spike protein and human ACE2
  • Oct 19, 2020
  • Scientific Reports
  • Anamika Basu + 2 more

Angiotensin converting enzyme 2 (ACE2) (EC:3.4.17.23) is a transmembrane protein which is considered as a receptor for spike protein binding of novel coronavirus (SARS-CoV2). Since no specific medication is available to treat COVID-19, designing of new drug is important and essential. In this regard, in silico method plays an important role, as it is rapid and cost effective compared to the trial and error methods using experimental studies. Natural products are safe and easily available to treat coronavirus affected patients, in the present alarming situation. In this paper five phytochemicals, which belong to flavonoid and anthraquinone subclass, have been selected as small molecules in molecular docking study of spike protein of SARS-CoV2 with its human receptor ACE2 molecule. Their molecular binding sites on spike protein bound structure with its receptor have been analyzed. From this analysis, hesperidin, emodin and chrysin are selected as competent natural products from both Indian and Chinese medicinal plants, to treat COVID-19. Among them, the phytochemical hesperidin can bind with ACE2 protein and bound structure of ACE2 protein and spike protein of SARS-CoV2 noncompetitively. The binding sites of ACE2 protein for spike protein and hesperidin, are located in different parts of ACE2 protein. Ligand spike protein causes conformational change in three-dimensional structure of protein ACE2, which is confirmed by molecular docking and molecular dynamics studies. This compound modulates the binding energy of bound structure of ACE2 and spike protein. This result indicates that due to presence of hesperidin, the bound structure of ACE2 and spike protein fragment becomes unstable. As a result, this natural product can impart antiviral activity in SARS CoV2 infection. The antiviral activity of these five natural compounds are further experimentally validated with QSAR study.

  • Research Article
  • Cite Count Icon 16
  • 10.31635/ccschem.020.202000322
New Insights from Chemical Biology: Molecular Basis of Transmission, Diagnosis, and Therapy of SARS-CoV-2
  • Jul 10, 2020
  • CCS Chemistry
  • Zilong Zhao + 10 more

Coronavirus disease 2019 (COVID-19) is caused by a novel strain of coronavirus, designated as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). It has caused a global pandemic rapidly s...

  • Research Article
  • Cite Count Icon 43
  • 10.1073/pnas.1807656116
Successive crystal structure snapshots suggest the basis for MHC class I peptide loading and editing by tapasin
  • Feb 26, 2019
  • Proceedings of the National Academy of Sciences
  • Ida Hafstrand + 16 more

MHC-I epitope presentation to CD8+ T cells is directly dependent on peptide loading and selection during antigen processing. However, the exact molecular bases underlying peptide selection and binding by MHC-I remain largely unknown. Within the peptide-loading complex, the peptide editor tapasin is key to the selection of MHC-I-bound peptides. Here, we have determined an ensemble of crystal structures of MHC-I in complex with the peptide exchange-associated dipeptide GL, as well as the tapasin-associated scoop loop, alone or in combination with candidate epitopes. These results combined with mutation analyses allow us to propose a molecular model underlying MHC-I peptide selection by tapasin. The N termini of bound peptides most probably bind first in the N-terminal and middle region of the MHC-I peptide binding cleft, upon which the peptide C termini are tested for their capacity to dislodge the tapasin scoop loop from the F pocket of the MHC-I cleft. Our results also indicate important differences in peptide selection between different MHC-I alleles.

More from: Journal of Molecular Recognition
  • Journal Issue
  • 10.1002/jmr.v38.5
  • Sep 1, 2025
  • Journal of Molecular Recognition

  • Research Article
  • 10.1002/jmr.3096
Issue Information
  • Jul 30, 2025
  • Journal of Molecular Recognition

  • Research Article
  • 10.1002/jmr.70009
Computational Cross‐Aggregation Study of Dengue Virus NS1, Capsid Anchor and 2k Peptides With Human Amylin, Aβ42 and α‐Synuclein Peptides
  • Jul 30, 2025
  • Journal of Molecular Recognition
  • Dwaipayan Chaudhuri + 2 more

  • Journal Issue
  • 10.1002/jmr.v38.4
  • Jul 1, 2025
  • Journal of Molecular Recognition

  • Research Article
  • 10.1002/jmr.3095
Issue Information
  • Jul 1, 2025
  • Journal of Molecular Recognition

  • Journal Issue
  • 10.1002/jmr.v38.3
  • May 1, 2025
  • Journal of Molecular Recognition

  • Research Article
  • 10.1002/jmr.3094
Issue Information
  • May 1, 2025
  • Journal of Molecular Recognition

  • Journal Issue
  • 10.1002/jmr.v38.2
  • Mar 1, 2025
  • Journal of Molecular Recognition

  • Research Article
  • 10.1002/jmr.3093
Issue Information
  • Jan 30, 2025
  • Journal of Molecular Recognition

  • Journal Issue
  • 10.1002/jmr.v38.1
  • Jan 1, 2025
  • Journal of Molecular Recognition

Save Icon
Up Arrow
Open/Close
  • Ask R Discovery Star icon
  • Chat PDF Star icon

AI summaries and top papers from 250M+ research sources.

Search IconWhat is the difference between bacteria and viruses?
Open In New Tab Icon
Search IconWhat is the function of the immune system?
Open In New Tab Icon
Search IconCan diabetes be passed down from one generation to the next?
Open In New Tab Icon