1
|
Iaculli D, Ballet S. Discovery of Bioactive Peptides Through Peptide Scanning. J Pept Sci 2025; 31:e70029. [PMID: 40347116 DOI: 10.1002/psc.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/25/2025] [Accepted: 04/30/2025] [Indexed: 05/12/2025]
Abstract
Therapeutic peptides targeted at various diseases are becoming increasingly relevant for the pharmaceutical industry. Several of these drugs were originally designed by mimicking a segment of a protein of interest. As such, protein mimicry represents a promising strategy both in immunology, for the identification of B- and T-cell epitopes, as well as for the modulation of protein activity, including the disruption of protein-protein interactions (PPIs) and the interference with biological or pathological cellular functions. Several methods have been developed to pinpoint the (binding) epitopes of a protein or the regions responsible for biological activity. One of such strategies is the scanning of the protein or selected domains with synthetic overlapping peptides. As the mechanism of action of a mimetic peptide can be similar to that of the whole protein, this method offers a powerful tool for the investigation of protein function, along with providing a solid basis for the development of therapeutic candidates. This review gives a general overview of different applications of the peptide scanning methodology, describing a comparison of the preparation and use of solid-phase libraries (peptide arrays) with isolated peptide libraries and highlighting their strengths and most common applications.
Collapse
Affiliation(s)
- Debora Iaculli
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
2
|
Tanwar N, Ojha R, Aggarwal S, Prajapati VK, Munde M. Design of inhibitor peptide sequences based on the interfacial knowledge of the protein G-IgG crystallographic complex and their binding studies with IgG. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2024; 53:159-170. [PMID: 38493432 DOI: 10.1007/s00249-024-01704-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/18/2024] [Accepted: 03/06/2024] [Indexed: 03/19/2024]
Abstract
Protein-protein interactions (PPI) have emerged as valuable targets in medicinal chemistry due to their key roles in important biological processes. The modulation of PPI by small peptides offers an excellent opportunity to develop drugs against human diseases. Here, we exploited the knowledge of the binding interface of the IgG-protein G complex (PDB:1FCC) for designing peptides that can inhibit these complexes. Herein, we have designed several closely related peptides, and the comparison of results from experiments and computational studies indicated that all the peptides bind close to the expected binding site on IgG and the complexes are stable. A minimal sequence consisting of 11 amino acids (P5) with binding constants in the range of 100 nM was identified. We propose that the main affinity differences across the series of peptides arose from the presence of polar amino acid residues. Further, the molecular dynamic studies helped to understand the dynamic properties of complexes in terms of flexibility of residues and structural stability at the interface. The ability of P5 to compete with the protein G in recognizing IgG can help in the detection and purification of antibodies. Further, it can serve as a versatile tool for a better understanding of protein-protein interactions.
Collapse
Affiliation(s)
- Neetu Tanwar
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Rupal Ojha
- Department of Nephrology, Washington University School of Medicine, St. Louis, MO, USA
| | - Soumya Aggarwal
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | | | - Manoj Munde
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
3
|
Zhang W, Forester NT, Chettri P, Heilijgers M, Mace WJ, Maes E, Morozova Y, Applegate ER, Johnson RD, Johnson LJ. Characterization of the Biosynthetic Gene Cluster for the Ribosomally Synthesized Cyclic Peptide Epichloëcyclins in Epichloë festucae. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:13965-13978. [PMID: 37704203 PMCID: PMC10540207 DOI: 10.1021/acs.jafc.3c03073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 09/15/2023]
Abstract
The various grass-induced epichloëcyclins of the Epichloë spp. are ribosomally synthesized and post-translationally modified peptides (RiPPs), produced as small, secreted cyclopeptides from a single gene, gigA. Here, four clustered and coregulated genes (gigA, gigB, gigC, and kexB) with predicted roles in epichloëcyclin production in Epichloë festucae were evaluated through gene disruption. Subsequent chemical analysis indicates that GigB is a DUF3328 domain-containing protein associated with cyclization of epichloëcyclins; GigC is a methyltransferase enzyme responsible for N-methylation of desmethylepichloëcyclins; and KexB is a subtilisin-like enzyme, partly responsible for the propeptide cleavage of epichloëcyclin intermediates. Symbiotic effects on the host phenotype were not observed for gigA, gigC, or kexB mutants, although ΔgigB infection correlated with increased host tiller height and biomass, while only ΔkexB exhibited an effect on endophyte morphology. Disrupting epichloëcyclin biosynthesis showed negligible influence on the biosynthesis of E. festucae-associated alkaloids. Epichloëcyclins may perform other secondary metabolism functions in Epichloë and other fungi.
Collapse
Affiliation(s)
- Wei Zhang
- Grasslands
Research Centre, AgResearch Limited, Palmerston North 4442, New Zealand
| | - Natasha T. Forester
- Grasslands
Research Centre, AgResearch Limited, Palmerston North 4442, New Zealand
| | - Pranav Chettri
- Grasslands
Research Centre, AgResearch Limited, Palmerston North 4442, New Zealand
| | - Maurice Heilijgers
- Grasslands
Research Centre, AgResearch Limited, Palmerston North 4442, New Zealand
| | - Wade J. Mace
- Grasslands
Research Centre, AgResearch Limited, Palmerston North 4442, New Zealand
| | - Evelyne Maes
- Lincoln
Research Centre, AgResearch Limited, Lincoln 7608, New Zealand
| | - Yulia Morozova
- Grasslands
Research Centre, AgResearch Limited, Palmerston North 4442, New Zealand
| | - Emma R. Applegate
- Grasslands
Research Centre, AgResearch Limited, Palmerston North 4442, New Zealand
| | - Richard D. Johnson
- Grasslands
Research Centre, AgResearch Limited, Palmerston North 4442, New Zealand
| | - Linda J. Johnson
- Grasslands
Research Centre, AgResearch Limited, Palmerston North 4442, New Zealand
| |
Collapse
|
4
|
Prediction and Modeling of Protein–Protein Interactions Using “Spotted” Peptides with a Template-Based Approach. Biomolecules 2022; 12:biom12020201. [PMID: 35204702 PMCID: PMC8961654 DOI: 10.3390/biom12020201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/20/2022] [Accepted: 01/22/2022] [Indexed: 12/10/2022] Open
Abstract
Protein–peptide interactions (PpIs) are a subset of the overall protein–protein interaction (PPI) network in the living cell and are pivotal for the majority of cell processes and functions. High-throughput methods to detect PpIs and PPIs usually require time and costs that are not always affordable. Therefore, reliable in silico predictions represent a valid and effective alternative. In this work, a new algorithm is described, implemented in a freely available tool, i.e., “PepThreader”, to carry out PPIs and PpIs prediction and analysis. PepThreader threads multiple fragments derived from a full-length protein sequence (or from a peptide library) onto a second template peptide, in complex with a protein target, “spotting” the potential binding peptides and ranking them according to a sequence-based and structure-based threading score. The threading algorithm first makes use of a scoring function that is based on peptides sequence similarity. Then, a rerank of the initial hits is performed, according to structure-based scoring functions. PepThreader has been benchmarked on a dataset of 292 protein–peptide complexes that were collected from existing databases of experimentally determined protein–peptide interactions. An accuracy of 80%, when considering the top predicted 25 hits, was achieved, which performs in a comparable way with the other state-of-art tools in PPIs and PpIs modeling. Nonetheless, PepThreader is unique in that it is able at the same time to spot a binding peptide within a full-length sequence involved in PPI and model its structure within the receptor. Therefore, PepThreader adds to the already-available tools supporting the experimental PPIs and PpIs identification and characterization.
Collapse
|
5
|
Betlem P, Maas MN, Middelburg J, Pieters BJGE, Mecinović J. Recognition of stapled histone H3K4me3 peptides by epigenetic reader proteins. Chem Commun (Camb) 2022; 58:12196-12199. [DOI: 10.1039/d2cc04294k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Epigenetic reader proteins can display stronger or weaker binding affinities for cyclic histone peptides relative to linear histones, indicating that selectivity of biomolecular recognition can be achieved.
Collapse
Affiliation(s)
- Peter Betlem
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Marijn N. Maas
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - Jim Middelburg
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Bas J. G. E. Pieters
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Jasmin Mecinović
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| |
Collapse
|
6
|
Liu J, Chen S, Chai XY, Gao F, Wang C, Tang H, Li X, Liu Y, Hu HG. Design, synthesis, and biological evaluation of stapled ascaphin-8 peptides. Bioorg Med Chem 2021; 40:116158. [PMID: 33932712 DOI: 10.1016/j.bmc.2021.116158] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 03/31/2021] [Accepted: 04/06/2021] [Indexed: 01/07/2023]
Abstract
Ascaphin-8 is an α-helical anti-tumor and antimicrobial peptide containing 19 residues, which was isolated from norepinephrine-stimulated skin secretions of the North American tailed frog Ascaphus truei. To improve both its stability and biological activities, a series of hydrocarbon-stapled analogs of Ascaphin-8 were synthesized and investigated for their potential antiproliferative activities. The activity studies were evaluated using the CCK-8 method and colony formation assay on human cancer cell lines. Ascaphin-8-3, as the most active peptide, showed a stronger inhibition effect when compared with the parent peptide for the tested cell lines. In addition, the effect of Ascaphin-8-3 on inhibiting the metastatic capabilities of A549 cells was more powerful than that of the parent peptide. This peptide derivative showed potentiality for further optimization in antitumor drugs.
Collapse
Affiliation(s)
- Jing Liu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China.
| | - Si Chen
- School of Medicine, Shanghai University, Shanghai 200444, China.
| | - Xiao-Yun Chai
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| | - Fei Gao
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China.
| | - Chen Wang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China.
| | - Hua Tang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China.
| | - Xiang Li
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| | - Ying Liu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China.
| | - Hong-Gang Hu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
7
|
Abstract
The implication of several TRP ion channels (e.g., TRPV1) in diverse physiological and pathological processes has signaled them as pivotal drug targets. Consequently, the identification of selective and potent ligands for these channels is of great interest in pharmacology and biomedicine. However, a major challenge in the design of modulators is ensuring the specificity for their intended targets. In recent years, the emergence of high-resolution structures of ion channels facilitates the computer-assisted drug design at molecular levels. Here we describe some computational methods and general protocols to discover channel modulators, including homology modelling, docking and virtual screening, and structure-based peptide design.
Collapse
Affiliation(s)
- Magdalena Nikolaeva Koleva
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche, Universitas Miguel Hernández, Elche, Spain
- AntalGenics SL. Ed. Quorum III, University Scientific Park, Universitas Miguel Hernández, Elche, Spain
| | - Gregorio Fernandez-Ballester
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche, Universitas Miguel Hernández, Elche, Spain.
| |
Collapse
|
8
|
Rubin SJS, Tal-Gan Y, Gilon C, Qvit N. Conversion of Protein Active Regions into Peptidomimetic Therapeutic Leads Using Backbone Cyclization and Cycloscan - How to Do it Yourself! Curr Top Med Chem 2018; 18:556-565. [PMID: 29773063 DOI: 10.2174/1568026618666180518094322] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/24/2018] [Accepted: 04/26/2018] [Indexed: 12/21/2022]
Abstract
Protein-protein Interactions (PPIs) are particularly important for controlling both physiologic and pathologic biological processes but are difficult to target due to their large and/or shallow interaction surfaces unsuitable for small molecules. Linear peptides found in nature interact with some PPIs, and protein active regions can be used to design synthetic peptide compounds for inhibition of PPIs. However, linear peptides are limited therapeutically by poor metabolic and conformational stability, which can compromise their bioactivity and half-life. Cyclic peptidomimetics (modified peptides) can be used to overcome these challenges because they are more resistant to metabolic degradation and can be engineered to adopt desired conformations. Backbone cyclization is a strategy that we developed to improve drug-like properties of linear peptide leads without jeopardizing the integrity of functionally relevant side-chains. Here, we provide the first description of an entire approach for developing backbone cyclized peptide compounds, based upon two straightforward 'ABC' and 'DEF' processes. We present practical examples throughout our discussion of revealing active regions important for PPIs and identifying critical pharmacophores, as well as developing backbone cyclized peptide libraries and screening them using cycloscan. Finally, we review the impact of these advances and provide a summary of current ongoing work in the field.
Collapse
Affiliation(s)
- Samuel J S Rubin
- Stanford Immunology Program, School of Medicine, Stanford University, 269 Campus drive, Stanford CA 94305-5174, United States
| | - Yftah Tal-Gan
- Department of Chemistry, University of Nevada, Reno, 1664 North Virginia Street, NV 89557, United States
| | - Chaim Gilon
- The Institute of Chemistry, The Hebrew University of Jerusalem, 91904, Jerusalem, Israel
| | - Nir Qvit
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, POB 1589, Safed, Israel
| |
Collapse
|
9
|
Villavicencio B, Ligabue-Braun R, Verli H. All-Hydrocarbon Staples and Their Effect over Peptide Conformation under Different Force Fields. J Chem Inf Model 2018; 58:2015-2023. [DOI: 10.1021/acs.jcim.8b00404] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Bianca Villavicencio
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), 91500-970 Porto Alegre-RS, Brazil
| | - Rodrigo Ligabue-Braun
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), 91500-970 Porto Alegre-RS, Brazil
| | - Hugo Verli
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), 91500-970 Porto Alegre-RS, Brazil
| |
Collapse
|
10
|
Mapping the interaction site and effect of the Siglec-9 inflammatory biomarker on human primary amine oxidase. Sci Rep 2018; 8:2086. [PMID: 29391504 PMCID: PMC5794975 DOI: 10.1038/s41598-018-20618-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 01/22/2018] [Indexed: 12/21/2022] Open
Abstract
Human primary amine oxidase (hAOC3), also known as vascular adhesion protein 1, mediates leukocyte rolling and trafficking to sites of inflammation by a multistep adhesion cascade. hAOC3 is absent on the endothelium of normal tissues and is kept upregulated during inflammatory conditions, which is an applicable advantage for imaging inflammatory diseases. Sialic acid binding immunoglobulin like-lectin 9 (Siglec-9) is a leukocyte ligand for hAOC3. The peptide (CARLSLSWRGLTLCPSK) based on the region of Siglec-9 that interacts with hAOC3, can be used as a specific tracer for hAOC3-targeted imaging of inflammation using Positron Emission Tomography (PET). In the present study, we show that the Siglec-9 peptide binds to hAOC3 and triggers its amine oxidase activity towards benzylamine. Furthermore, the hAOC3 inhibitors semicarbazide and imidazole reduce the binding of wild type and Arg/Ala mutated Siglec-9 peptides to hAOC3. Molecular docking of the Siglec-9 peptide is in accordance with the experimental results and predicts that the R3 residue in the peptide interacts in the catalytic site of hAOC3 when the topaquinone cofactor is in the non-catalytic on-copper conformation. The predicted binding mode of Siglec-9 peptide to hAOC3 is supported by the PET studies using rodent, rabbit and pig AOC3 proteins.
Collapse
|
11
|
Rossmann M, J Greive S, Moschetti T, Dinan M, Hyvönen M. Development of a multipurpose scaffold for the display of peptide loops. Protein Eng Des Sel 2017; 30:419-430. [PMID: 28444399 PMCID: PMC5897841 DOI: 10.1093/protein/gzx017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 02/26/2017] [Accepted: 03/28/2017] [Indexed: 12/20/2022] Open
Abstract
Protein-protein interactions (PPIs) determine a wide range of biological processes and analysis of these dynamic networks is increasingly becoming a mandatory tool for studying protein function. Using the globular ATPase domain of recombinase RadA as a scaffold, we have developed a peptide display system (RAD display), which allows for the presentation of target peptides, protein domains or full-length proteins and their rapid recombinant production in bacteria. The design of the RAD display system includes differently tagged versions of the scaffold, which allows for flexibility in the protein purification method, and chemical coupling for small molecule labeling or surface immobilization. When combined with the significant thermal stability of the RadA protein, these features create a versatile multipurpose scaffold system. Using various orthogonal biophysical techniques, we show that peptides displayed on the scaffold bind to their natural targets in a fashion similar to linear parent peptides. We use the examples of CK2β/CK2α kinase and TPX2/Aurora A kinase protein complexes to demonstrate that the peptide displayed by the RAD scaffold can be used in PPI studies with the same binding efficacy but at lower costs compared with their linear synthetic counterparts.
Collapse
Affiliation(s)
- Maxim Rossmann
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Sandra J Greive
- York Structural Biology Laboratory, Department of Chemistry, University of York, Heslington, York YO10 5DD, UK
| | - Tommaso Moschetti
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Michael Dinan
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Marko Hyvönen
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK. Correspondence:
| |
Collapse
|
12
|
Chabert V, Hologne M, Sénèque O, Crochet A, Walker O, Fromm KM. Model peptide studies of Ag+ binding sites from the silver resistance protein SilE. Chem Commun (Camb) 2017; 53:6105-6108. [DOI: 10.1039/c7cc02630g] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A model peptide study characterizes several Ag+-binding sites of the bacterial silver resistant protein SilE, providing new insights into its physiological role.
Collapse
Affiliation(s)
- V. Chabert
- Univ. Fribourg
- Department of Chemistry
- 1700 Fribourg
- Switzerland
| | - M. Hologne
- Univ. Lyon
- CNRS
- UCB Lyon 1
- ENS-Lyon
- Institut des Sciences Analytiques
| | - O. Sénèque
- Univ. Grenoble Alpes
- CNRS
- CEA, LCBM (UMR 5249)
- F-38000 Grenoble
- France
| | - A. Crochet
- Univ. Fribourg
- Fribourg Center for Nanomaterials
- FriMat
- 1700 Fribourg
- Switzerland
| | - O. Walker
- Univ. Lyon
- CNRS
- UCB Lyon 1
- ENS-Lyon
- Institut des Sciences Analytiques
| | - K. M. Fromm
- Univ. Fribourg
- Department of Chemistry
- 1700 Fribourg
- Switzerland
| |
Collapse
|
13
|
Hiew SH, Guerette PA, Zvarec OJ, Phillips M, Zhou F, Su H, Pervushin K, Orner BP, Miserez A. Modular peptides from the thermoplastic squid sucker ring teeth form amyloid-like cross-β supramolecular networks. Acta Biomater 2016; 46:41-54. [PMID: 27693688 DOI: 10.1016/j.actbio.2016.09.040] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 09/23/2016] [Accepted: 09/28/2016] [Indexed: 12/22/2022]
Abstract
The hard sucker ring teeth (SRT) from decapodiforme cephalopods, which are located inside the sucker cups lining the arms and tentacles of these species, have recently emerged as a unique model structure for biomimetic structural biopolymers. SRT are entirely composed of modular, block co-polymer-like proteins that self-assemble into a large supramolecular network. In order to unveil the molecular principles behind SRT's self-assembly and robustness, we describe a combinatorial screening assay that maps the molecular-scale interactions between the most abundant modular peptide blocks of suckerin proteins. By selecting prominent interaction hotspots from this assay, we identified four peptides that exhibited the strongest homo-peptidic interactions, and conducted further in-depth biophysical characterizations complemented by molecular dynamic (MD) simulations to investigate the nature of these interactions. Circular Dichroism (CD) revealed conformations that transitioned from semi-extended poly-proline II (PII) towards β-sheet structure. The peptides spontaneously self-assembled into microfibers enriched with cross β-structures, as evidenced by Fourier-Transform Infrared Spectroscopy (FTIR) and Congo red staining. Nuclear Magnetic Resonance (NMR) experiments identified the residues involved in the hydrogen-bonded network and demonstrated that these self-assembled β-sheet-based fibers exhibit high protection factors that bear resemblance to amyloids. The high stability of the β-sheet network and an amyloid-like model of fibril assembly were supported by MD simulations. The work sheds light on how Nature has evolved modular sequence design for the self-assembly of mechanically robust functional materials, and expands our biomolecular toolkit to prepare load-bearing biomaterials from protein-based block co-polymers and self-assembled peptides. STATEMENT OF SIGNIFICANCE The sucker ring teeth (SRT) located on the arms and tentacles of cephalopods represent as a very promising protein-based biopolymer with the potential to rival silk in biomedical and engineering applications. SRT are made of modular, block co-polymer like proteins (suckerins), which assemble into a semicrystalline polymer reinforced by nano-confined β-sheets, resulting in a supramolecular network with mechanical properties that match those of the strongest engineering polymers. In this study, we aimed to understand the molecular mechanisms behind SRT's self-assembly and robustness. The most abundant modular peptidic blocks of suckerin proteins were studied by various spectroscopic methods, which demonstrate that SRT peptides form amyloid-like cross-β structures.
Collapse
|
14
|
Gao M, Cheng K, Yin H. Targeting protein-protein interfaces using macrocyclic peptides. Biopolymers 2016; 104:310-6. [PMID: 25664609 DOI: 10.1002/bip.22625] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 01/29/2015] [Accepted: 02/01/2015] [Indexed: 01/10/2023]
Abstract
Protein-protein interactions (PPIs) are critical in numerous biological processes including signaling transduction, function regulations, and disease development. To regulate PPIs has been thought to be challenging due to their highly dynamic and expansive interfacial areas. Nonetheless, successful examples have been reported of targeting PPIs using small molecules, peptides, and proteins. Peptides, especially macrocyclic peptides have proven to be a particularly useful tool to inhibit PPIs for their exquisite potency, stability and selectivity. Herein we review the recent developments of this area of research, focusing on the macrocyclic peptides isolated from natural products, identified from library screening, and rationally designed based on structures, as PPI regulators.
Collapse
Affiliation(s)
- Meng Gao
- Department of Chemistry, Center of Basic Molecular Science, Tsinghua University, Beijing, China , 100082
| | - Kui Cheng
- Department of Chemistry, Center of Basic Molecular Science, Tsinghua University, Beijing, China , 100082
| | - Hang Yin
- Department of Chemistry, Center of Basic Molecular Science, Tsinghua University, Beijing, China , 100082.,Department of Chemistry and Biochemistry, the BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80309-0596
| |
Collapse
|
15
|
Unexpected involvement of staple leads to redesign of selective bicyclic peptide inhibitor of Grb7. Sci Rep 2016; 6:27060. [PMID: 27257138 PMCID: PMC4891710 DOI: 10.1038/srep27060] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 05/12/2016] [Indexed: 01/11/2023] Open
Abstract
The design of potent and specific peptide inhibitors to therapeutic targets is of enormous utility for both proof-of-concept studies and for the development of potential new therapeutics. Grb7 is a key signaling molecule in the progression of HER2 positive and triple negative breast cancers. Here we report the crystal structure of a stapled bicyclic peptide inhibitor G7-B1 in complex with the Grb7-SH2 domain. This revealed an unexpected binding mode of the peptide, in which the staple forms an alternative contact with the surface of the target protein. Based on this structural information, we designed a new series of bicyclic G7 peptides that progressively constrain the starting peptide, to arrive at the G7-B4 peptide that binds with an approximately 2-fold enhanced affinity to the Grb7-SH2 domain (KD = 0.83 μM) compared to G7-B1 and shows low affinity binding to Grb2-, Grb10- and Grb14-SH2 domains (KD > 100 μM). Furthermore, we determined the structure of the G7-B4 bicyclic peptide in complex with the Grb7-SH2 domain, both before and after ring closing metathesis to show that the closed staple is essential to the target interaction. The G7-B4 peptide represents an advance in the development of Grb7 inhibitors and is a classical example of structure aided inhibitor development.
Collapse
|
16
|
Kanthala S, Gauthier T, Satyanarayanajois S. Structure-activity relationships of peptidomimetics that inhibit PPI of HER2-HER3. Biopolymers 2016; 101:693-702. [PMID: 24222531 DOI: 10.1002/bip.22441] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 11/01/2013] [Accepted: 11/05/2013] [Indexed: 12/25/2022]
Abstract
Human epidermal growth factor receptor-2 (HER2) is a tyrosine kinase family protein receptor that is known to undergo heterodimerization with other members of the family of epidermal growth factor receptors (EGFR) for cell signaling. Overexpression of HER2 and deregulation of signaling has implications in breast, ovarian, and lung cancers. We have designed several peptidomimetics to block the HER2-mediated dimerization, resulting in antiproliferative activity for cancer cells. In this work, we have investigated the structure-activity relationships of peptidomimetic analogs of Compound 5. Compound 5 was conformationally constrained by N- and C-terminal modification and cyclization as well as by substitution with d-amino acids at the N-and C-termini. Among the compounds studied in this work, a peptidomimetic Compound 21 with d-amino acid substitution and its N- and C-termini capped with acetyl and amide functional groups and a reversed sequence compared to that of Compound 5 exhibited better antiproliferative activity in HER2-overexpressed breast, ovarian, and lung cancer cell lines. Compound 21 was further evaluated for its protein-protein interaction (PPI) inhibition ability using enzyme fragment complementation assay, proximity ligation assay, and Western blot analysis. Results suggested that Compound 21 is able to block HER2:HER3 interaction and inhibit phosphorylation of the kinase domain of HER2. The mode of binding of Compound 21 to HER2 protein was modeled using a docking method. Compound 21 seems to bind to domain IV of HER2 near the PPI site of EGFR:HER2, and HER:HER3 and inhibit PPI.
Collapse
Affiliation(s)
- Shanthi Kanthala
- Department of Basic Pharmaceutical Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201
| | | | | |
Collapse
|
17
|
Fouché M, Schäfer M, Berghausen J, Desrayaud S, Blatter M, Piéchon P, Dix I, Martin Garcia A, Roth HJ. Design and Development of a Cyclic Decapeptide Scaffold with Suitable Properties for Bioavailability and Oral Exposure. ChemMedChem 2016; 11:1048-59. [DOI: 10.1002/cmdc.201600082] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 03/09/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Marianne Fouché
- Global Discovery Chemistry/Macrocycles; Novartis Institute for BioMedical Research; Basel 4002 Switzerland
| | - Michael Schäfer
- Global Discovery Chemistry/CADD; Novartis Institute for BioMedical Research; Basel 4002 Switzerland
| | - Jörg Berghausen
- Metabolism and Pharmacokinetics; Novartis Institute for BioMedical Research; Basel 4002 Switzerland
| | - Sandrine Desrayaud
- Metabolism and Pharmacokinetics; Novartis Institute for BioMedical Research; Basel 4002 Switzerland
| | - Markus Blatter
- Global Discovery Chemistry/Analytics (NMR); Novartis Institute for BioMedical Research; Basel 4002 Switzerland
| | - Philippe Piéchon
- Global Discovery Chemistry/Analytics (Crystallography); Novartis Institute for BioMedical Research; Basel 4002 Switzerland
| | - Ina Dix
- Global Discovery Chemistry/Analytics (Crystallography); Novartis Institute for BioMedical Research; Basel 4002 Switzerland
| | - Aimar Martin Garcia
- The University of the Basque Country-Euskal Herriko Unibertsitatea; Campus de Leioa 48949 Leioa Spain
| | - Hans-Jörg Roth
- Global Discovery Chemistry/Macrocycles; Novartis Institute for BioMedical Research; Basel 4002 Switzerland
| |
Collapse
|
18
|
Mokrushin AA. Mystixin-7 mini-peptide protects ionotropic glutamatergic mechanisms against oxygen-glucose deprivation in vitro. Neuropeptides 2016; 56:51-7. [PMID: 26526227 DOI: 10.1016/j.npep.2015.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 10/22/2015] [Accepted: 10/22/2015] [Indexed: 11/22/2022]
Abstract
The aim of the present study was to explore the neuroprotective effects of the mystixin-7 mini-peptide (MTX, a synthetic corticotropin-releasing-factor-like, 7-amino-acid peptide) on an in vitro oxygen glucose deprivation model (OGD, 10min). The study used a technique of on-line monitoring of changes in α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic receptor (AMPAR) and N-methyl-d-aspartic acid receptor (NMDAR)-mediated field excitatory postsynaptic potentials (fEPSPs) in the olfactory cortex slices in the OGD model. OGD resulted in an irreversible blockade of both AMPAR and NMDAR activity. Pretreatment of slices by MTX and their subsequent exposure to OGD resulted in decreased activity of these postsynaptic mechanisms (AMPARs, 71%; NMDARs, 68% as compared to baseline), but they were not blocked altogether. The degree protection of activity of both AMPARs and NMDARs had dose-dependent manner, with a maximal effect at 100mg/mL. These protective effects were retained after the removal of MTX from the bathing medium. To evaluate the protective efficacy of MTX on NMDARs, the slices were pretreated by MTX and exposed to OGD and then treated with l-glutamate (1mM). NMDARs' response to application of l-glutamate was minimal at higher concentrations of MTX and maximal at lower concentrations. These findings indicate that the molecules of MTX interact with a certain amount of NMDARs, and thereby protect them from the OGD. Pretreatment of slices with MTX contributed to the protection of network activity against OGD and promoted the development of the learning process in the form of long-term potentiation. To specify the protective effects of MTX, it was denatured by trypsin. The proteolytic cleavage of MTX resulted to a significant decrease in the activity of both AMPARs and NMDARs against OGD as compared with that of the native peptide. Together, these findings provide further insight into the protective potential of the MTX mini-peptide. We believe that the data presented can be the basis for the development of therapeutics MTX-based medications for the treatment of the ischemic stroke.
Collapse
Affiliation(s)
- Anatoly A Mokrushin
- I. P. Pavlov Institute of Physiology, Russian Academy of Science, 199034, nab. Makarova, 6, Saint-Petersburg, Russia.
| |
Collapse
|
19
|
Lu MC, Yuan ZW, Jiang YL, Chen ZY, You QD, Jiang ZY. A systematic molecular dynamics approach to the study of peptide Keap1–Nrf2 protein–protein interaction inhibitors and its application to p62 peptides. MOLECULAR BIOSYSTEMS 2016; 12:1378-87. [DOI: 10.1039/c6mb00030d] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Protein–protein interactions (PPIs) as drug targets have been gaining growing interest, though developing drug-like small molecule PPI inhibitors remains challenging.
Collapse
Affiliation(s)
- Meng-Chen Lu
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- China
- Jiangsu Key Laboratory of Drug Design and Optimization
| | - Zhen-Wei Yuan
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- China
- Jiangsu Key Laboratory of Drug Design and Optimization
| | - Yong-Lin Jiang
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- China
- Jiangsu Key Laboratory of Drug Design and Optimization
| | - Zhi-Yun Chen
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- China
- Jiangsu Key Laboratory of Drug Design and Optimization
| | - Qi-Dong You
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- China
- Jiangsu Key Laboratory of Drug Design and Optimization
| | - Zheng-Yu Jiang
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- China
- Jiangsu Key Laboratory of Drug Design and Optimization
| |
Collapse
|
20
|
Review cyclic peptides on a merry-go-round; towards drug design. Biopolymers 2015; 104:453-61. [DOI: 10.1002/bip.22669] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 04/14/2015] [Accepted: 05/04/2015] [Indexed: 01/15/2023]
|
21
|
Aihara K, Inokuma T, Komiya C, Shigenaga A, Otaka A. Synthesis of lactam-bridged cyclic peptides using sequential olefin metathesis and diimide reduction reactions. Tetrahedron 2015. [DOI: 10.1016/j.tet.2015.04.093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
22
|
Interactions of HIV-1 proteins as targets for developing anti-HIV-1 peptides. Future Med Chem 2015; 7:1055-77. [DOI: 10.4155/fmc.15.46] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Protein–protein interactions (PPI) are essential in every step of the HIV replication cycle. Mapping the interactions between viral and host proteins is a fundamental target for the design and development of new therapeutics. In this review, we focus on rational development of anti-HIV-1 peptides based on mapping viral–host and viral–viral protein interactions all across the HIV-1 replication cycle. We also discuss the mechanism of action, specificity and stability of these peptides, which are designed to inhibit PPI. Some of these peptides are excellent tools to study the mechanisms of PPI in HIV-1 replication cycle and for the development of anti-HIV-1 drug leads that modulate PPI.
Collapse
|
23
|
Gokhale AS, Satyanarayanajois S. Peptides and peptidomimetics as immunomodulators. Immunotherapy 2015; 6:755-74. [PMID: 25186605 DOI: 10.2217/imt.14.37] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Peptides and peptidomimetics can function as immunomodulating agents by either blocking the immune response or stimulating the immune response to generate tolerance. Knowledge of B- or T-cell epitopes along with conformational constraints is important in the design of peptide-based immunomodulating agents. Work on the conformational aspects of peptides, synthesis and modified amino acid side chains have contributed to the development of a new generation of therapeutic agents for autoimmune diseases and cancer. The design of peptides/peptidomimetics for immunomodulation in autoimmune diseases such as multiple sclerosis, rheumatoid arthritis, systemic lupus and HIV infection is reviewed. In cancer therapy, peptide epitopes are used in such a way that the body is trained to recognize and fight the cancer cells locally as well as systemically.
Collapse
Affiliation(s)
- Ameya S Gokhale
- Basic Pharmaceutical Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | | |
Collapse
|
24
|
Visconti A, Ermondi G, Caron G, Esposito R. Prediction and interpretation of the lipophilicity of small peptides. J Comput Aided Mol Des 2015; 29:361-70. [PMID: 25577035 DOI: 10.1007/s10822-015-9829-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 01/02/2015] [Indexed: 01/28/2023]
Abstract
Peptide-based drug discovery has considerably expanded and solid in silico tools for the prediction of physico-chemical properties of peptides are urgently needed. In this work we tested some combinations of descriptors/algorithms to find the best model to predict [Formula: see text] of a series of peptides. To do that we evaluate the models statistical performances but also their skills in providing a reliable deconvolution of the balance of intermolecular forces governing the partitioning phenomenon. Results prove that a PLS model based on VolSurf+ descriptors is the best tool to predict [Formula: see text] of neutral and ionised peptides. The mechanistic interpretation also reveals that the inclusion in the chemical structure of a HBD group is more efficient in decreasing lipophilicity than the inclusion of a HBA group.
Collapse
Affiliation(s)
- Alessia Visconti
- Department of Genomics of Common Disease, Imperial College London, Du Cane Road, W12 ONN, London, UK,
| | | | | | | |
Collapse
|
25
|
Stability of structured Kaposi's sarcoma-associated herpesvirus ORF57 protein is regulated by protein phosphorylation and homodimerization. J Virol 2015; 89:3256-74. [PMID: 25568207 DOI: 10.1128/jvi.03721-14] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
UNLABELLED Kaposi's sarcoma-associated herpesvirus (KSHV) ORF57 plays an essential role in KSHV lytic infection by promoting viral gene expression at the posttranscriptional level. Using bioinformatic and biochemical approaches, we determined that ORF57 contains two structurally and functionally distinct domains: a disordered nonstructural N-terminal domain (amino acids [aa] 1 to 152) and a structured α-helix-rich C-terminal domain (aa 153 to 455). The N-terminal domain mediates ORF57 interaction with several RNA-protein complexes essential for ORF57 to function. The N-terminal phosphorylation by cellular casein kinase II (CKII) at S21, T32, and S43, and other cellular kinases at S95 and S97 residues in proximity of the caspase-7 cleavage site, 30-DETD-33, inhibits caspase-7 digestion of ORF57. The structured C-terminal domain mediates homodimerization of ORF57, and the critical region for this function was mapped carefully to α-helices 7 to 9. Introduction of point mutations into α-helix 7 at ORF57 aa 280 to 299, a region highly conserved among ORF57 homologues from other herpesviruses, inhibited ORF57 homodimerization and led to proteasome-mediated degradation of ORF57 protein. Thus, homodimerization of ORF57 via its C terminus prevents ORF57 from degrading and allows two structure-free N termini of the dimerized ORF57 to work coordinately for host factor interactions, leading to productive KSHV lytic infection and pathogenesis. IMPORTANCE KSHV is a human oncogenic virus linked to the development of several malignancies. KSHV-mediated oncogenesis requires both latent and lytic infection. The KSHV ORF57 protein is essential for KSHV lytic replication, as it regulates the expression of viral lytic genes at the posttranscriptional level. This report provides evidence that the structural conformation of the ORF57 protein plays a critical role in regulation of ORF57 stability. Phosphorylation by CKII on the identified serine/threonine residues at the N-terminal unstructured domain of ORF57 prevents its digestion by caspase-7. The C-terminal domain of ORF57, which is rich in α-helices, contributes to homodimerization of ORF57 to prevent proteasome-mediated protein degradation. Elucidation of the ORF57 structure not only enables us to better understand ORF57 stability and functions but also provides an important tool for us to modulate ORF57's activity with the aim to inhibit KSHV lytic replication.
Collapse
|
26
|
Amartely H, Iosub-Amir A, Friedler A. Identifying protein-protein interaction sites using peptide arrays. J Vis Exp 2014:e52097. [PMID: 25490271 DOI: 10.3791/52097] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Protein-protein interactions mediate most of the processes in the living cell and control homeostasis of the organism. Impaired protein interactions may result in disease, making protein interactions important drug targets. It is thus highly important to understand these interactions at the molecular level. Protein interactions are studied using a variety of techniques ranging from cellular and biochemical assays to quantitative biophysical assays, and these may be performed either with full-length proteins, with protein domains or with peptides. Peptides serve as excellent tools to study protein interactions since peptides can be easily synthesized and allow the focusing on specific interaction sites. Peptide arrays enable the identification of the interaction sites between two proteins as well as screening for peptides that bind the target protein for therapeutic purposes. They also allow high throughput SAR studies. For identification of binding sites, a typical peptide array usually contains partly overlapping 10-20 residues peptides derived from the full sequences of one or more partner proteins of the desired target protein. Screening the array for binding the target protein reveals the binding peptides, corresponding to the binding sites in the partner proteins, in an easy and fast method using only small amount of protein. In this article we describe a protocol for screening peptide arrays for mapping the interaction sites between a target protein and its partners. The peptide array is designed based on the sequences of the partner proteins taking into account their secondary structures. The arrays used in this protocol were Celluspots arrays prepared by INTAVIS Bioanalytical Instruments. The array is blocked to prevent unspecific binding and then incubated with the studied protein. Detection using an antibody reveals the binding peptides corresponding to the specific interaction sites between the proteins.
Collapse
Affiliation(s)
- Hadar Amartely
- Institute of Chemistry, The Hebrew University of Jerusalem
| | | | - Assaf Friedler
- Institute of Chemistry, The Hebrew University of Jerusalem;
| |
Collapse
|
27
|
Mercurio FA, Scognamiglio PL, Di Natale C, Marasco D, Pellecchia M, Leone M. CD and NMR conformational studies of a peptide encompassing the Mid Loop interface of Ship2-Sam. Biopolymers 2014; 101:1088-98. [DOI: 10.1002/bip.22512] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 05/23/2014] [Accepted: 05/23/2014] [Indexed: 11/07/2022]
Affiliation(s)
| | - Pasqualina L. Scognamiglio
- Department of Pharmacy; University "Federico II"; Naples Italy
- Centro Interuniversitario di Ricerca sui Peptidi Bioattivi (CIRPEB); Naples Italy
- IIT Italian Institute of Technology; Naples Italy
| | - Concetta Di Natale
- Department of Pharmacy; University "Federico II"; Naples Italy
- IIT Italian Institute of Technology; Naples Italy
| | - Daniela Marasco
- Institute of Biostructures and Bioimaging (CNR); Naples Italy
- Department of Pharmacy; University "Federico II"; Naples Italy
- Centro Interuniversitario di Ricerca sui Peptidi Bioattivi (CIRPEB); Naples Italy
| | | | - Marilisa Leone
- Institute of Biostructures and Bioimaging (CNR); Naples Italy
- Centro Interuniversitario di Ricerca sui Peptidi Bioattivi (CIRPEB); Naples Italy
| |
Collapse
|
28
|
Focused chemical libraries--design and enrichment: an example of protein-protein interaction chemical space. Future Med Chem 2014; 6:1291-307. [PMID: 24773599 DOI: 10.4155/fmc.14.57] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
One of the many obstacles in the development of new drugs lies in the limited number of therapeutic targets and in the quality of screening collections of compounds. In this review, we present general strategies for building target-focused chemical libraries with a particular emphasis on protein-protein interactions (PPIs). We describe the chemical spaces spanned by nine commercially available PPI-focused libraries and compare them to our 2P2I3D academic library, dedicated to orthosteric PPI modulators. We show that although PPI-focused libraries have been designed using different strategies, they share common subspaces. PPI inhibitors are larger and more hydrophobic than standard drugs; however, an effort has been made to improve the drug-likeness of focused chemical libraries dedicated to this challenging class of targets.
Collapse
|
29
|
Holub JM, Larochelle JR, Appelbaum JS, Schepartz A. Improved assays for determining the cytosolic access of peptides, proteins, and their mimetics. Biochemistry 2013; 52:9036-46. [PMID: 24256505 DOI: 10.1021/bi401069g] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Proteins and other macromolecules that cross biological membranes have great potential as tools for research and next-generation therapeutics. Here, we describe two assays that effectively quantify the cytosolic localization of a number of previously reported peptides and protein domains. One assay, which we call GIGI (glucocorticoid-induced eGFP induction), is an amplified assay that informs on relative cytosolic access without the need for sophisticated imaging equipment or adherent cells. The second, GIGT (glucocorticoid-induced eGFP translocation), is a nonamplified assay that informs on relative cytosolic access and exploits sophisticated imaging equipment to facilitate high-content screens in live cells. Each assay was employed to quantify the cytosolic delivery of several canonical "cell permeable peptides," as well as more recently reported minimally cationic miniature proteins and zinc finger nuclease domains. Our results show definitively that both overall charge as well as charge distribution influence cytosolic access and that small protein domains containing a discrete, helical, penta-Arg motif can dramatically improve the cytosolic delivery of small folded proteins such as zinc finger domains. We anticipate that the assays described herein will prove useful to explore and discover the fundamental physicochemical and genetic properties that influence both the uptake and endosomal release of peptidic molecules and their mimetics.
Collapse
Affiliation(s)
- Justin M Holub
- Department of Chemistry, Yale University , P.O. Box 208107, New Haven, Connecticut 06520-8107, United States
| | | | | | | |
Collapse
|
30
|
Góngora-Benítez M, Tulla-Puche J, Albericio F. Multifaceted Roles of Disulfide Bonds. Peptides as Therapeutics. Chem Rev 2013; 114:901-26. [DOI: 10.1021/cr400031z] [Citation(s) in RCA: 388] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Miriam Góngora-Benítez
- Institute
for Research in Biomedicine (IRB Barcelona), Barcelona, 08028 Spain
- CIBER-BBN, Barcelona Science
Park, Barcelona, 08028 Spain
| | - Judit Tulla-Puche
- Institute
for Research in Biomedicine (IRB Barcelona), Barcelona, 08028 Spain
- CIBER-BBN, Barcelona Science
Park, Barcelona, 08028 Spain
| | - Fernando Albericio
- Institute
for Research in Biomedicine (IRB Barcelona), Barcelona, 08028 Spain
- CIBER-BBN, Barcelona Science
Park, Barcelona, 08028 Spain
- Department
of Organic Chemistry, University of Barcelona, Barcelona, 08028 Spain
- School of Chemistry & Physics, University of KwaZulu-Natal, 4001 Durban, South Africa
| |
Collapse
|
31
|
Abstract
The human Mediator complex is a central integrator for transcription and represents a primary interface that allows DNA-binding transcription factors to communicate their regulatory signals to the RNA polymerase II enzyme. Because Mediator is dynamic both in terms of subunit composition and structure, it presents challenges as a target for small molecule probes. Moreover, little high-resolution structural information exists for Mediator. Its global requirement for transcription, as well as its distinct, transcription factor specific interaction surfaces, however, suggest that development of probes that bind specific Mediator subunits might enable gene- and pathway-specific modulation of transcription. Here we provide a brief overview of the Mediator complex, highlighting biological and structural features that make it an attractive target for molecular probes. We then outline several chemical strategies that might be effective for targeting the complex.
Collapse
Affiliation(s)
| | - Dylan J Taatjes
- Dept. of Chemistry and Biochemistry, University of Colorado, Boulder, CO 80303 USA
| |
Collapse
|
32
|
Wawrzyńska A, Kurzyk A, Mierzwińska M, Płochocka D, Wieczorek G, Sirko A. Direct targeting of Arabidopsis cysteine synthase complexes with synthetic polypeptides to selectively deregulate cysteine synthesis. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2013; 207:148-157. [PMID: 23602110 DOI: 10.1016/j.plantsci.2013.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 02/22/2013] [Accepted: 02/24/2013] [Indexed: 06/02/2023]
Abstract
Biosynthesis of cysteine is one of the fundamental processes in plants providing the reduced sulfur for cell metabolism. It is accomplished by the sequential action of two enzymes, serine acetyltransferase (SAT) and O-acetylserine (thiol) lyase (OAS-TL). Together they constitute the hetero-oligomeric cysteine synthase (CS) complex through specific protein-protein interactions influencing the rate of cysteine production. The aim of our studies was to deregulate the CS complex formation in order to investigate its function in the control of sulfur homeostasis and optimize cysteine synthesis. Computational modeling was used to build a model of the Arabidopsis thaliana mitochondrial CS complex. Several polypeptides based on OAS-TL C amino-acid sequence found at SAT-OASTL interaction sites were designed as probable competitors for SAT3 binding. After verification of the binding in a yeast two-hybrid assay, the most strongly interacting polypeptide was introduced to different cellular compartments of Arabidopsis cell via genetic transformation. Moderate increase in total SAT and OAS-TL activities, but not thiols content, was observed dependent on the transgenic line and sulfur availability in the hydroponic medium. Though our studies demonstrate the proof of principle, they also suggest more complex interaction of both enzymes underlying the mechanism of their reciprocal regulation.
Collapse
Affiliation(s)
- Anna Wawrzyńska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5A St, 02-106 Warsaw, Poland.
| | | | | | | | | | | |
Collapse
|
33
|
Hurevich M, Ratner-Hurevich M, Tal-Gan Y, Shalev DE, Ben-Sasson SZ, Gilon C. Backbone cyclic helix mimetic of chemokine (C-C motif) receptor 2: a rational approach for inhibiting dimerization of G protein-coupled receptors. Bioorg Med Chem 2013; 21:3958-66. [PMID: 23706536 DOI: 10.1016/j.bmc.2013.03.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 02/28/2013] [Accepted: 03/01/2013] [Indexed: 10/27/2022]
Abstract
The transmembrane helical bundle of G protein-coupled receptors (GPCRs) dimerize through helix-helix interactions in response to inflammatory stimulation. A strategy was developed to target the helical dimerization site of GPCRs by peptidomimetics with drug like properties. The concept was demonstrated by selecting a potent backbone cyclic helix mimetic from a library that derived from the dimerization region of chemokine (C-C motif) receptor 2 (CCR2) that is a key player in Multiple Sclerosis. We showed that CCR2 based backbone cyclic peptide having a stable helix structure inhibits specific CCR2-mediated chemotactic migration.
Collapse
Affiliation(s)
- Mattan Hurevich
- Institute of Chemistry, The Hebrew University of Jerusalem, Safra Campus, Givat Ram, Jerusalem 91904, Israel
| | | | | | | | | | | |
Collapse
|
34
|
Hogan M, Bahta M, Cherry S, Lountos GT, Tropea JE, Zhao BM, Burke TR, Waugh DS, Ulrich RG. Biomolecular Interactions of small-molecule inhibitors affecting the YopH protein tyrosine phosphatase. Chem Biol Drug Des 2013; 81:323-33. [PMID: 23241354 PMCID: PMC3573263 DOI: 10.1111/cbdd.12097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We have developed competitive and direct binding methods to examine small-molecule inhibitors of protein tyrosine phosphatase activity. Focusing on the Yersinia pestis outer protein H, a potent bacterial protein tyrosine phosphatase, we describe how an understanding of the kinetic interactions involving Yersinia pestis outer protein H, peptide substrates, and small-molecule inhibitors of protein tyrosine phosphatase activity can be beneficial for inhibitor screening, and we further translate these results into a microarray assay for high-throughput screening.
Collapse
Affiliation(s)
- Megan Hogan
- United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702, United States
| | - Medhanit Bahta
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, United States
| | - Scott Cherry
- Macromolecular Crystallography Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick National Lab, Frederick, Maryland 21702, United States
| | - George T. Lountos
- Macromolecular Crystallography Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick National Lab, Frederick, Maryland 21702, United States
- Basic Science Program, SAIC-Frederick, Inc., Frederick National Lab, Frederick, Maryland 21702, United States
| | - Joseph E. Tropea
- Macromolecular Crystallography Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick National Lab, Frederick, Maryland 21702, United States
| | - Bryan M. Zhao
- United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702, United States
| | - Terrence R. Burke
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, United States
| | - David S. Waugh
- Macromolecular Crystallography Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick National Lab, Frederick, Maryland 21702, United States
| | - Robert G. Ulrich
- United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702, United States
| |
Collapse
|
35
|
Kastritis PL, Bonvin AMJJ. On the binding affinity of macromolecular interactions: daring to ask why proteins interact. J R Soc Interface 2012; 10:20120835. [PMID: 23235262 PMCID: PMC3565702 DOI: 10.1098/rsif.2012.0835] [Citation(s) in RCA: 306] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Interactions between proteins are orchestrated in a precise and time-dependent manner, underlying cellular function. The binding affinity, defined as the strength of these interactions, is translated into physico-chemical terms in the dissociation constant (Kd), the latter being an experimental measure that determines whether an interaction will be formed in solution or not. Predicting binding affinity from structural models has been a matter of active research for more than 40 years because of its fundamental role in drug development. However, all available approaches are incapable of predicting the binding affinity of protein–protein complexes from coordinates alone. Here, we examine both theoretical and experimental limitations that complicate the derivation of structure–affinity relationships. Most work so far has concentrated on binary interactions. Systems of increased complexity are far from being understood. The main physico-chemical measure that relates to binding affinity is the buried surface area, but it does not hold for flexible complexes. For the latter, there must be a significant entropic contribution that will have to be approximated in the future. We foresee that any theoretical modelling of these interactions will have to follow an integrative approach considering the biology, chemistry and physics that underlie protein–protein recognition.
Collapse
Affiliation(s)
- Panagiotis L Kastritis
- Bijvoet Center for Biomolecular Research, Faculty of Science, Chemistry, Utrecht University, , Padualaan 8, Utrecht, The Netherlands
| | | |
Collapse
|
36
|
Maes M, Loyter A, Friedler A. Peptides that inhibit HIV-1 integrase by blocking its protein-protein interactions. FEBS J 2012; 279:2795-809. [PMID: 22742518 DOI: 10.1111/j.1742-4658.2012.08680.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
HIV-1 integrase (IN) is one of the key enzymes in the viral replication cycle. It mediates the integration of viral cDNA into the host cell genome. IN activity requires interactions with several viral and cellular proteins, as well as IN oligomerization. Inhibition of IN is an important target for the development of anti-HIV therapies, but there is currently only one anti-HIV drug used in the clinic that targets IN. Several other small-molecule anti-IN drug leads are either undergoing clinical trials or in earlier stages of development. These molecules specifically inhibit one of the IN-mediated reactions necessary for successful integration. However, small-molecule inhibitors of protein-protein interactions are difficult to develop. In this review, we focus on peptides that inhibit IN. Peptides have advantages over small-molecule inhibitors of protein-protein interactions: they can mimic the structures of the binding domains within proteins, and are large enough to competitively inhibit protein-protein interactions. The development of peptides that bind IN and inhibit its protein-protein interactions will increase our understanding of the IN mode of action, and lead to the development of new drug leads, such as small molecules derived from these peptides, for better anti-HIV therapy.
Collapse
Affiliation(s)
- Michal Maes
- Institute of Chemistry, The Hebrew University of Jerusalem, Israel
| | | | | |
Collapse
|
37
|
Abstract
INTRODUCTION The interpretation of high-throughput profiling data depends on the pathway analysis database. Currently, pathway analysis often has to rely on a set of interactions and pathways measured in every possible human tissue, due to insufficient knowledge about interactions and pathways in the context of the profiling experiment. However, a recent global scale analysis of human tissue proteomes and interactomes reveals significant differences among tissues, suggesting that interaction and pathway data that are used out of biological context are the major source of inaccuracies and noise in the analysis of profiling data. AREAS COVERED In this review, the major classes of biological context used for experimental detection of molecular interactions and pathways in molecular biology are described. Furthermore, the author reviews methods for predicting biological interactions in order to evaluate the applicability of various contextual interaction data in pathway analysis. Using the results from recent publications that study large-scale tissue composition, the article provides an estimation of the gain in pathway analysis accuracy if only the interactions predicted for the context of a molecular profiling experiment are used, relative to the analysis performed with a context-independent knowledge base. EXPERT OPINION It is of the author's opinion that the major source of inaccuracy in pathway analysis is the lack of knowledge about tissue-specific transcriptional regulation. It is therefore suggested that the accuracy of the analysis can be substantially improved if only context-specific interactions and pathways are used for interpretation.
Collapse
Affiliation(s)
- Anton Yuryev
- Elsevier, Ariadne Genomics, Inc., Rockville, MD 20878, USA.
| |
Collapse
|
38
|
Wang MC, Chen FC, Chen YZ, Huang YT, Chuang TJ. LDGIdb: a database of gene interactions inferred from long-range strong linkage disequilibrium between pairs of SNPs. BMC Res Notes 2012; 5:212. [PMID: 22551073 PMCID: PMC3441865 DOI: 10.1186/1756-0500-5-212] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 04/26/2012] [Indexed: 12/22/2022] Open
Abstract
Background Complex human diseases may be associated with many gene interactions. Gene interactions take several different forms and it is difficult to identify all of the interactions that are potentially associated with human diseases. One approach that may fill this knowledge gap is to infer previously unknown gene interactions via identification of non-physical linkages between different mutations (or single nucleotide polymorphisms, SNPs) to avoid hitchhiking effect or lack of recombination. Strong non-physical SNP linkages are considered to be an indication of biological (gene) interactions. These interactions can be physical protein interactions, regulatory interactions, functional compensation/antagonization or many other forms of interactions. Previous studies have shown that mutations in different genes can be linked to the same disorders. Therefore, non-physical SNP linkages, coupled with knowledge of SNP-disease associations may shed more light on the role of gene interactions in human disorders. A user-friendly web resource that integrates information about non-physical SNP linkages, gene annotations, SNP information, and SNP-disease associations may thus be a good reference for biomedical research. Findings Here we extracted the SNPs located within the promoter or exonic regions of protein-coding genes from the HapMap database to construct a database named the Linkage-Disequilibrium-based Gene Interaction database (LDGIdb). The database stores 646,203 potential human gene interactions, which are potential interactions inferred from SNP pairs that are subject to long-range strong linkage disequilibrium (LD), or non-physical linkages. To minimize the possibility of hitchhiking, SNP pairs inferred to be non-physically linked were required to be located in different chromosomes or in different LD blocks of the same chromosomes. According to the genomic locations of the involved SNPs (i.e., promoter, untranslated region (UTR) and coding region (CDS)), the SNP linkages inferred were categorized into promoter-promoter, promoter-UTR, promoter-CDS, CDS-CDS, CDS-UTR and UTR-UTR linkages. For the CDS-related linkages, the coding SNPs were further classified into nonsynonymous and synonymous variations, which represent potential gene interactions at the protein and RNA level, respectively. The LDGIdb also incorporates human disease-association databases such as Genome-Wide Association Studies (GWAS) and Online Mendelian Inheritance in Man (OMIM), so that the user can search for potential disease-associated SNP linkages. The inferred SNP linkages are also classified in the context of population stratification to provide a resource for investigating potential population-specific gene interactions. Conclusion The LDGIdb is a user-friendly resource that integrates non-physical SNP linkages and SNP-disease associations for studies of gene interactions in human diseases. With the help of the LDGIdb, it is plausible to infer population-specific SNP linkages for more focused studies, an avenue that is potentially important for pharmacogenetics. Moreover, by referring to disease-association information such as the GWAS data, the LDGIdb may help identify previously uncharacterized disease-associated gene interactions and potentially lead to new discoveries in studies of human diseases. Keywords Gene interaction, SNP, Linkage disequilibrium, Systems biology, Bioinformatics
Collapse
Affiliation(s)
- Ming-Chih Wang
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | | | | | | | | |
Collapse
|
39
|
Studying protein–peptide interactions using benzophenone units: A case study of protein kinase B/Akt and its inhibitor PTR6154. Anal Biochem 2012; 421:750-4. [DOI: 10.1016/j.ab.2011.11.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 11/17/2011] [Accepted: 11/23/2011] [Indexed: 11/20/2022]
|
40
|
Developing potent backbone cyclic peptides bearing the shared epitope sequence as rheumatoid arthritis drug-leads. Bioorg Med Chem Lett 2011; 22:493-6. [PMID: 22113111 DOI: 10.1016/j.bmcl.2011.10.098] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 10/18/2011] [Accepted: 10/27/2011] [Indexed: 01/06/2023]
Abstract
Rheumatoid arthritis (RA) is a common human leukocyte antigen-associated disease. Most RA patients have a five-residue sequence motif called the shared epitope (SE) in the DRβ-chain of the HLA-DRB1 protein. The SE was found to activate nitric oxide (NO) production, suggesting a possible mechanism for RA development. The native conformation of the SE is presumed to be an α-helix, thus using cyclic peptides to stabilize this conformation may produce a potent SE mimetic which will have drug-like properties. We present the development of a backbone cyclic SE mimetic that activates NO production in the low nM range. Circular dichroism analysis revealed a conformational change from for the parent linear peptides to the cyclic analogs. The most active cyclic analog is completely stable towards trypsin/chymotrypsin degradation while the linear 15-mer analogs completely degraded within 30 min. The outcome of this study is a potent cyclic peptide with drug-like properties that can be used as a template for drug development.
Collapse
|
41
|
Abstract
Rapid advances in our collective understanding of biomolecular structure and, in concert, of biochemical systems, coupled with developments in computational methods, have massively impacted the field of medicinal chemistry over the past two decades, with even greater changes appearing on the horizon. In this perspective, we endeavor to profile some of the most prominent determinants of change and speculate as to further evolution that may consequently occur during the next decade. The five main angles to be addressed are: protein-protein interactions; peptides and peptidomimetics; molecular diversity and pharmacological space; molecular pharmacodynamics (significance, potential and challenges); and early-stage clinical efficacy and safety. We then consider, in light of these, the future of medicinal chemistry and the educational preparation that will be required for future medicinal chemists.
Collapse
Affiliation(s)
- Seetharama D Satyanarayanajois
- Basic Pharmaceutical Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe LA 71201, USA.
| | | |
Collapse
|
42
|
Friedler A. From peptides to proteins: lessons from my years at the Centre for Protein Engineering. Protein Eng Des Sel 2010; 24:241-5. [PMID: 21062759 DOI: 10.1093/protein/gzq080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The MRC Centre for Protein Engineering (CPE) hosted and trained many scientists over the years. It is a unique research environment that shaped the career of many scientists in all aspects. These include research directions and methodologies, but even more important--issues such as how to approach scientific problems and how to manage a research team. Alan Fersht was the director of the CPE when I joined it as a postdoc in the year 2000. In the current article for the PEDS special CPE issue, I will review how my scientific research and my approach to science developed from the days I arrived to the CPE as a young peptide chemist and throughout the years I spent at the CPE, and how it shaped my current research interests and attitude. I will focus on two major fields: (i) Using peptides to study and modulate the structure and interactions of proteins; (ii) Using quantitative biophysical methods to study proteins and their interactions at the molecular level.
Collapse
Affiliation(s)
- Assaf Friedler
- Institute of Chemistry, The Hebrew University of Jerusalem, Safra Campus, Givat Ram, Jerusalem 91904, Israel.
| |
Collapse
|