1
|
Montero F, Parra-López M, Rodríguez-Martínez A, Murciano-Calles J, Buzon P, Han Z, Lin LY, Ramos MC, Ruiz-Sanz J, Martinez JC, Radi M, Moog C, Diederich S, Harty RN, Pérez-Sánchez H, Vicente F, Castillo F, Luque I. Exploring the druggability of the UEV domain of human TSG101 in search for broad-spectrum antivirals. Protein Sci 2025; 34:e70005. [PMID: 39724449 DOI: 10.1002/pro.70005] [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: 10/11/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024]
Abstract
The ubiquitin E2 variant domain of TSG101 (TSG101-UEV) plays a pivotal role in protein sorting and virus budding by recognizing PTAP motifs within ubiquitinated proteins. Disruption of TSG101-UEV/PTAP interactions has emerged as a promising strategy for the development of host-oriented broad-spectrum antivirals with low susceptibility to resistance. TSG101 is a challenging target characterized by an extended and flat binding interface, low affinity for PTAP ligands, and complex binding energetics. Here, we assess the druggability of the TSG101-UEV/PTAP binding interface by searching for drug-like inhibitors and evaluating their ability to block PTAP recognition, impair budding, and inhibit viral proliferation. A discovery workflow was established by combining in vitro miniaturized HTS assays and a set of cell-based activity assays including high-content bimolecular complementation, virus-like particle release measurement, and antiviral testing in live virus infection. This approach has allowed us to identify a set of chemically diverse molecules that block TSG101-UEV/PTAP binding with IC50s in the low μM range and are able to disrupt the interaction between full-length TSG101 and viral proteins in human cells and inhibit viral replication. State-of-the-art molecular docking studies reveal that the active compounds exploit binding hotspots at the PTAP binding site, unlocking the full binding potential of the TSG101-UEV binding pockets. These inhibitors represent promising hits for the development of novel broad-spectrum antivirals through targeted optimization and are also valuable tools for investigating the involvement of ESCRT in the proliferation of different virus families and study the secondary effects induced by the disruption of ESCRT/virus interactions.
Collapse
Affiliation(s)
- Fernando Montero
- Department of Physical Chemistry, Institute of Biotechnology, and Unit of Excellence in Chemistry Applied to Biomedicine and Environment, School of Sciences, University of Granada, Granada, Spain
| | - Marisa Parra-López
- Department of Physical Chemistry, Institute of Biotechnology, and Unit of Excellence in Chemistry Applied to Biomedicine and Environment, School of Sciences, University of Granada, Granada, Spain
| | - Alejandro Rodríguez-Martínez
- Department of Physical Chemistry, Institute of Biotechnology, and Unit of Excellence in Chemistry Applied to Biomedicine and Environment, School of Sciences, University of Granada, Granada, Spain
- Structural Bioinformatics and High-Performance Computing (BIO-HPC) Research Group, Universidad Católica de Murcia (UCAM), Guadalupe, Spain
| | - Javier Murciano-Calles
- Department of Physical Chemistry, Institute of Biotechnology, and Unit of Excellence in Chemistry Applied to Biomedicine and Environment, School of Sciences, University of Granada, Granada, Spain
| | - Pedro Buzon
- Department of Physical Chemistry, Institute of Biotechnology, and Unit of Excellence in Chemistry Applied to Biomedicine and Environment, School of Sciences, University of Granada, Granada, Spain
| | - Ziying Han
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - L-Y Lin
- Laboratoire d'ImmunoRhumatologie Moléculaire, UMR_S 1109, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | | | - Javier Ruiz-Sanz
- Department of Physical Chemistry, Institute of Biotechnology, and Unit of Excellence in Chemistry Applied to Biomedicine and Environment, School of Sciences, University of Granada, Granada, Spain
| | - Jose C Martinez
- Department of Physical Chemistry, Institute of Biotechnology, and Unit of Excellence in Chemistry Applied to Biomedicine and Environment, School of Sciences, University of Granada, Granada, Spain
| | - Marco Radi
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parma, Italy
| | - Christiane Moog
- Laboratoire d'ImmunoRhumatologie Moléculaire, UMR_S 1109, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Sandra Diederich
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Greifswald, Germany
| | - Ronald N Harty
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Horacio Pérez-Sánchez
- Structural Bioinformatics and High-Performance Computing (BIO-HPC) Research Group, Universidad Católica de Murcia (UCAM), Guadalupe, Spain
| | | | | | - Irene Luque
- Department of Physical Chemistry, Institute of Biotechnology, and Unit of Excellence in Chemistry Applied to Biomedicine and Environment, School of Sciences, University of Granada, Granada, Spain
| |
Collapse
|
2
|
Tang X, Chen J, Cai J, Wang Q. N-substituting perturbation on the interaction affinity and recognition specificity between rheumatic immune-related Abl SH3 domain and its peptoid ligands. J Mol Graph Model 2023; 125:108601. [PMID: 37607432 DOI: 10.1016/j.jmgm.2023.108601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/04/2023] [Accepted: 08/13/2023] [Indexed: 08/24/2023]
Abstract
Abl is a nonreceptor tyrosine kinase involved in a variety of disease pathways such as rheumatic immune. Full-length Abl protein consists of a catalytic tyrosine kinase (TK) domain as well as two regulatory Src homology domains 2 and 3 (SH2 and SH3, respectively); the latter recognizes and binds to those natural proline-rich peptide segments containing a PxxP motif on the protein surface of its interacting partners. However, natural peptides cannot bind effectively to the modular domain in high affinity and strong selectivity due to their small size and broad specificity. Here, a synthetic proline-rich peptide p41 was used as template; its structural diversity was extended by combinationally replacing the Pro0 and Pro+3 residues with a number of N-substituted amino acids. Consequently, peptide affinity change upon the replacement was derived to create a systematic N-substituting perturbation profile, from which we identified several N-substitution combinations at the Pro0 and Pro+3 residues of p41 PxxP motif that may moderately or significantly improve the peptide binding potency to Abl; they represent potent peptoid binders of Abl SH3 domain, with affinity improved considerably relative to p41. More significantly, the designed potent peptoids were also found to exhibit a good SH3-selectivity for their cognate Abl over other noncognate nonreceptor tyrosine kinases, with S = 9.7-fold.
Collapse
Affiliation(s)
- Xiaomin Tang
- Department of Acupuncture Rehabilitation, Danyang Traditional Chinese Medicine Hospital, Zhenjiang 212399, China
| | - Jingjin Chen
- Department of Acupuncture Rehabilitation, Danyang Traditional Chinese Medicine Hospital, Zhenjiang 212399, China
| | - Jiahui Cai
- Department of Acupuncture Rehabilitation, Danyang Traditional Chinese Medicine Hospital, Zhenjiang 212399, China
| | - Qiuqin Wang
- Nursing College, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
3
|
Zhao S, Shen L, Wang Q, Lu W. Dynamics simulation, energetics calculation and experimental analysis of the intermolecular interaction between human neonatal ABL SH3 domain and its N-substituted peptoid ligands. J Biomol Struct Dyn 2023; 42:12637-12644. [PMID: 37909467 DOI: 10.1080/07391102.2023.2272344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 10/08/2023] [Indexed: 11/03/2023]
Abstract
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.
Collapse
Affiliation(s)
- Shijian Zhao
- Department of Gynaecology and Obstetrics, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, China
| | - Lili Shen
- Department of Pediatrics, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, China
| | - Qiuqin Wang
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenxiao Lu
- Department of Gynaecology and Obstetrics, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, China
| |
Collapse
|
4
|
Lin J, Wang S, Wen L, Ye H, Shang S, Li J, Shu J, Zhou P. Targeting peptide-mediated interactions in omics. Proteomics 2023; 23:e2200175. [PMID: 36461811 DOI: 10.1002/pmic.202200175] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022]
Abstract
Peptide-mediated interactions (PMIs) play a crucial role in cell signaling network, which are responsible for about half of cellular protein-protein associations in the human interactome and have recently been recognized as a new kind of promising druggable target for drug development and disease therapy. In this article, we give a systematic review regarding the proteome-wide discovery of PMIs and targeting druggable PMIs (dPMIs) with chemical drugs, self-inhibitory peptides (SIPs) and protein agents, particularly focusing on their implications and applications for therapeutic purpose in omics. We also introduce computational peptidology strategies used to model, analyze, and design PMI-targeted molecular entities and further extend the concepts of protein context, direct/indirect readout, and enthalpy/entropy effect involved in PMIs. Current issues and future perspective on this topic are discussed. There is still a long way to go before establishment of efficient therapeutic strategies to target PMIs on the omics scale.
Collapse
Affiliation(s)
- Jing Lin
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC), Chengdu, China
| | - Shaozhou Wang
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC), Chengdu, China
| | - Li Wen
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC), Chengdu, China
| | - Haiyang Ye
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC), Chengdu, China
| | - Shuyong Shang
- Institute of Ecological Environment Protection, Chengdu Normal University, Chengdu, China
| | - Juelin Li
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC), Chengdu, China
| | - Jianping Shu
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC), Chengdu, China
| | - Peng Zhou
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC), Chengdu, China
| |
Collapse
|
5
|
Martinez JC, Castillo F, Ruiz-Sanz J, Murciano-Calles J, Camara-Artigas A, Luque I. Understanding binding affinity and specificity of modular protein domains: A focus in ligand design for the polyproline-binding families. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 130:161-188. [PMID: 35534107 DOI: 10.1016/bs.apcsb.2021.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Within the modular protein domains there are five families that recognize proline-rich sequences: SH3, WW, EVH1, GYF and UEV domains. This chapter reviews the main strategies developed for the design of ligands for these families, including peptides, peptidomimetics and drugs. We also describe some studies aimed to understand the molecular reasons responsible for the intrinsic affinity and specificity of these domains.
Collapse
Affiliation(s)
- Jose C Martinez
- Departamento de Química Física e Instituto de Biotecnología, Facultad de Ciencias, Universidad de Granada, Granada, Spain.
| | - Francisco Castillo
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Parque Tecnológico Ciencias de la Salud, Granada, Spain
| | - Javier Ruiz-Sanz
- Departamento de Química Física e Instituto de Biotecnología, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - Javier Murciano-Calles
- Departamento de Química Física e Instituto de Biotecnología, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - Ana Camara-Artigas
- Departamento de Química Física, Universidad de Almería, Campus de Excelencia Internacional Agroalimentario ceiA3 y CIAMBITAL, Almeria, Spain
| | - Irene Luque
- Departamento de Química Física e Instituto de Biotecnología, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| |
Collapse
|
6
|
Vincenzi M, Mercurio FA, Leone M. Protein Interaction Domains: Structural Features and Drug Discovery Applications (Part 2). Curr Med Chem 2021; 28:854-892. [PMID: 31942846 DOI: 10.2174/0929867327666200114114142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/28/2019] [Accepted: 11/04/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Proteins present a modular organization made up of several domains. Apart from the domains playing catalytic functions, many others are crucial to recruit interactors. The latter domains can be defined as "PIDs" (Protein Interaction Domains) and are responsible for pivotal outcomes in signal transduction and a certain array of normal physiological and disease-related pathways. Targeting such PIDs with small molecules and peptides able to modulate their interaction networks, may represent a valuable route to discover novel therapeutics. OBJECTIVE This work represents a continuation of a very recent review describing PIDs able to recognize post-translationally modified peptide segments. On the contrary, the second part concerns with PIDs that interact with simple peptide sequences provided with standard amino acids. METHODS Crucial structural information on different domain subfamilies and their interactomes was gained by a wide search in different online available databases (including the PDB (Protein Data Bank), the Pfam (Protein family), and the SMART (Simple Modular Architecture Research Tool)). Pubmed was also searched to explore the most recent literature related to the topic. RESULTS AND CONCLUSION PIDs are multifaceted: they have all diverse structural features and can recognize several consensus sequences. PIDs can be linked to different diseases onset and progression, like cancer or viral infections and find applications in the personalized medicine field. Many efforts have been centered on peptide/peptidomimetic inhibitors of PIDs mediated interactions but much more work needs to be conducted to improve drug-likeness and interaction affinities of identified compounds.
Collapse
Affiliation(s)
- Marian Vincenzi
- Institute of Biostructures and Bioimaging, National Research Council (CNR), Via Mezzocannone 16, 80134 Naples, Italy
| | - Flavia Anna Mercurio
- Institute of Biostructures and Bioimaging, National Research Council (CNR), Via Mezzocannone 16, 80134 Naples, Italy
| | - Marilisa Leone
- Institute of Biostructures and Bioimaging, National Research Council (CNR), Via Mezzocannone 16, 80134 Naples, Italy
| |
Collapse
|
7
|
Chu W, Prodromou R, Day KN, Schneible JD, Bacon KB, Bowen JD, Kilgore RE, Catella CM, Moore BD, Mabe MD, Alashoor K, Xu Y, Xiao Y, Menegatti S. Peptides and pseudopeptide ligands: a powerful toolbox for the affinity purification of current and next-generation biotherapeutics. J Chromatogr A 2020; 1635:461632. [PMID: 33333349 DOI: 10.1016/j.chroma.2020.461632] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 02/08/2023]
Abstract
Following the consolidation of therapeutic proteins in the fight against cancer, autoimmune, and neurodegenerative diseases, recent advancements in biochemistry and biotechnology have introduced a host of next-generation biotherapeutics, such as CRISPR-Cas nucleases, stem and car-T cells, and viral vectors for gene therapy. With these drugs entering the clinical pipeline, a new challenge lies ahead: how to manufacture large quantities of high-purity biotherapeutics that meet the growing demand by clinics and biotech companies worldwide. The protein ligands employed by the industry are inadequate to confront this challenge: while featuring high binding affinity and selectivity, these ligands require laborious engineering and expensive manufacturing, are prone to biochemical degradation, and pose safety concerns related to their bacterial origin. Peptides and pseudopeptides make excellent candidates to form a new cohort of ligands for the purification of next-generation biotherapeutics. Peptide-based ligands feature excellent target biorecognition, low or no toxicity and immunogenicity, and can be manufactured affordably at large scale. This work presents a comprehensive and systematic review of the literature on peptide-based ligands and their use in the affinity purification of established and upcoming biological drugs. A comparative analysis is first presented on peptide engineering principles, the development of ligands targeting different biomolecular targets, and the promises and challenges connected to the industrial implementation of peptide ligands. The reviewed literature is organized in (i) conventional (α-)peptides targeting antibodies and other therapeutic proteins, gene therapy products, and therapeutic cells; (ii) cyclic peptides and pseudo-peptides for protein purification and capture of viral and bacterial pathogens; and (iii) the forefront of peptide mimetics, such as β-/γ-peptides, peptoids, foldamers, and stimuli-responsive peptides for advanced processing of biologics.
Collapse
Affiliation(s)
- Wenning Chu
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Raphael Prodromou
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Kevin N Day
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - John D Schneible
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Kaitlyn B Bacon
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - John D Bowen
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Ryan E Kilgore
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Carly M Catella
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Brandyn D Moore
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Matthew D Mabe
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Kawthar Alashoor
- Department of Biochemistry and Biophysics, University of Rochester, Rochester, NY 14642
| | - Yiman Xu
- College of Material Science and Engineering, Donghua University, 201620 Shanghai, People's Republic of China
| | - Yuanxin Xiao
- College of Textile, Donghua University, Songjiang District, Shanghai, 201620, People's Republic of China
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606.
| |
Collapse
|
8
|
Marciel MP, Hoffmann PR. Molecular Mechanisms by Which Selenoprotein K Regulates Immunity and Cancer. Biol Trace Elem Res 2019; 192:60-68. [PMID: 31187393 PMCID: PMC6801056 DOI: 10.1007/s12011-019-01774-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/05/2019] [Indexed: 02/07/2023]
Abstract
Many of the 25 members of the selenoprotein family function as enzymes that utilize their selenocysteine (Sec) residues to catalyze redox-based reactions. However, some selenoproteins likely do not exert enzymatic activity by themselves and selenoprotein K (SELENOK) is one such selenoprotein family member that uses its Sec residue in an alternative manner. SELENOK is an endoplasmic reticulum (ER) transmembrane protein that has been shown to be important for ER stress and for calcium-dependent signaling. Molecular mechanisms for the latter have recently been elucidated using knockout mice and genetically manipulated cell lines. These studies have shown that SELENOK interacts with an enzyme in the ER membrane, DHHC6 (letters represent the amino acids aspartic acid, histidine, histidine, and cysteine in the catalytic domain), and the SELENOK/DHHC6 complex catalyzes the transfer of acyl groups such as palmitate to cysteine residues in target proteins, i.e., palmitoylation. One protein palmitoylated by SELENOK/DHHC6 is the calcium channel protein, the inositol 1,4,5-trisphosphate receptor (IP3R), which is acylated as a means for stabilizing the tetrameric calcium channel in the ER membrane. Factors that lower SELENOK levels or function impair IP3R-driven calcium flux. This role for SELENOK is important for the activation and proliferation of immune cells, and recently, a critical role for SELENOK in promoting calcium flux for the progression of melanoma has been demonstrated. This review provides a summary of these findings and their implications in terms of designing new therapeutic interventions that target SELENOK for treating cancers like melanoma.
Collapse
Affiliation(s)
- Michael P Marciel
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Peter R Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA.
| |
Collapse
|
9
|
Meirson T, Bomze D, Kahlon L, Gil-Henn H, Samson AO. A helical lock and key model of polyproline II conformation with SH3. Bioinformatics 2019; 36:154-159. [DOI: 10.1093/bioinformatics/btz527] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/09/2019] [Accepted: 06/24/2019] [Indexed: 11/12/2022] Open
Abstract
Abstract
Motivation
More than half of the human proteome contains the proline-rich motif, PxxP. This motif has a high propensity for adopting a left-handed polyproline II (PPII) helix and can potentially bind SH3 domains. SH3 domains are generally grouped into two classes, based on whether the PPII binds in a positive (N-to-C terminal) or negative (C-to-N terminal) orientation. Since the discovery of this structural motif, over six decades ago, a systematic understanding of its binding remains poor and the consensus amino acid sequence that binds SH3 domains is still ill defined.
Results
Here, we show that the PPII interaction with SH3 domains is governed by the helix backbone and its prolines, and their rotation angle around the PPII helical axis. Based on a geometric analysis of 131 experimentally solved SH3 domains in complex with PPIIs, we observed a rotary translation along the helical screw axis, and separated them by 120° into three categories we name α (0–120°), β (120–240°) and γ (240–360°). Furthermore, we found that PPII helices are distinguished by a shifting PxxP motif preceded by positively charged residues which act as a structural reading frame and dictates the organization of SH3 domains; however, there is no one single consensus motif for all classified PPIIs. Our results demonstrate a remarkable apparatus of a lock with a rotating and translating key with no known equivalent machinery in molecular biology. We anticipate our model to be a starting point for deciphering the PPII code, which can unlock an exponential growth in our understanding of the relationship between protein structure and function.
Availability and implementation
We have implemented the proposed methods in the R software environment and in an R package freely available at https://github.com/Grantlab/bio3d.
Supplementary information
Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Tomer Meirson
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Safed 1589, Israel
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1589, Israel
| | - David Bomze
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 39040, Israel
| | - Liron Kahlon
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1589, Israel
| | - Hava Gil-Henn
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1589, Israel
| | - Abraham O Samson
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Safed 1589, Israel
| |
Collapse
|
10
|
Niu J, Dick IE, Yang W, Bamgboye MA, Yue DT, Tomaselli G, Inoue T, Ben-Johny M. Allosteric regulators selectively prevent Ca 2+-feedback of Ca V and Na V channels. eLife 2018; 7:35222. [PMID: 30198845 PMCID: PMC6156082 DOI: 10.7554/elife.35222] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 09/09/2018] [Indexed: 12/31/2022] Open
Abstract
Calmodulin (CaM) serves as a pervasive regulatory subunit of CaV1, CaV2, and NaV1 channels, exploiting a functionally conserved carboxy-tail element to afford dynamic Ca2+-feedback of cellular excitability in neurons and cardiomyocytes. Yet this modularity counters functional adaptability, as global changes in ambient CaM indiscriminately alter its targets. Here, we demonstrate that two structurally unrelated proteins, SH3 and cysteine-rich domain (stac) and fibroblast growth factor homologous factors (fhf) selectively diminish Ca2+/CaM-regulation of CaV1 and NaV1 families, respectively. The two proteins operate on allosteric sites within upstream portions of respective channel carboxy-tails, distinct from the CaM-binding interface. Generalizing this mechanism, insertion of a short RxxK binding motif into CaV1.3 carboxy-tail confers synthetic switching of CaM regulation by Mona SH3 domain. Overall, our findings identify a general class of auxiliary proteins that modify Ca2+/CaM signaling to individual targets allowing spatial and temporal orchestration of feedback, and outline strategies for engineering Ca2+/CaM signaling to individual targets.
Collapse
Affiliation(s)
- Jacqueline Niu
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, United States
| | - Ivy E Dick
- Department of Physiology, University of Maryland, Baltimore, United States
| | - Wanjun Yang
- Department of Cardiology, Johns Hopkins University, Baltimore, United States
| | | | - David T Yue
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, United States
| | - Gordon Tomaselli
- Department of Cardiology, Johns Hopkins University, Baltimore, United States
| | - Takanari Inoue
- Department of Cell Biology, Johns Hopkins University, Baltimore, United States.,Center for Cell Dynamics, Institute for Basic Biomedical Sciences, Johns Hopkins University, Baltimore, United States
| | - Manu Ben-Johny
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, United States
| |
Collapse
|
11
|
Wapling J, Srivastava S, Shehu-Xhilaga M, Tachedjian G. Targeting Human Immunodeficiency Virus Type 1 Assembly, Maturation and Budding. Drug Target Insights 2017. [DOI: 10.1177/117739280700200020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Johanna Wapling
- Molecular Interactions Group, Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, 3004, Australia
- Department of Microbiology, Monash University, Clayton, Victoria 3168, Australia
| | - Seema Srivastava
- Molecular Interactions Group, Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, 3004, Australia
| | - Miranda Shehu-Xhilaga
- Department of Medicine, Monash University, Prahran, Victoria 3181, Australia
- Infectious Diseases Unit, Alfred Hospital, Prahran, Victoria 3181, Australia
| | - Gilda Tachedjian
- Molecular Interactions Group, Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, 3004, Australia
- Department of Microbiology, Monash University, Clayton, Victoria 3168, Australia
- Department of Medicine, Monash University, Prahran, Victoria 3181, Australia
| |
Collapse
|
12
|
Shen Q, Zeng D, Zhao B, Bhatt VS, Li P, Cho JH. The Molecular Mechanisms Underlying the Hijack of Host Proteins by the 1918 Spanish Influenza Virus. ACS Chem Biol 2017; 12:1199-1203. [PMID: 28368102 DOI: 10.1021/acschembio.7b00168] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The 1918 Spanish influenza A virus (IAV) caused one of the most serious pandemics in history. The nonstructural protein 1 (NS1) of the 1918 IAV hijacks the interaction between human CrkII and JNK1. Little is, however, known about its molecular mechanism. Here, we performed X-ray crystallography, NMR relaxation dispersion experiment, and fluorescence spectroscopy to determine the structural, kinetic, and thermodynamic mechanisms underlying the hijacking of CrkII by 1918 IAV NS1. We observed that the interaction between a proline-rich motif in NS1 and the N-terminal SH3 domain of CrkII displays strikingly rapid kinetics and exceptionally high affinity with 100-fold faster kon and 3300-fold lower Kd compared to those for the CrkII-JNK1 interaction. These results provide molecular insight into the mechanism by which 1918 IAV NS1 hijacks CrkII and disrupts its interactions with critical cellular signaling proteins.
Collapse
Affiliation(s)
- Qingliang Shen
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Danyun Zeng
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Baoyu Zhao
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Veer S. Bhatt
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Pingwei Li
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Jae-Hyun Cho
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
13
|
Kim H, Chen W. A non-chromatographic protein purification strategy using Src 3 homology domains as generalized capture domains. J Biotechnol 2016; 234:27-34. [DOI: 10.1016/j.jbiotec.2016.07.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 07/18/2016] [Accepted: 07/22/2016] [Indexed: 10/21/2022]
|
14
|
Butterfoss GL, Drew K, Renfrew PD, Kirshenbaum K, Bonneau R. Conformational preferences of peptide-peptoid hybrid oligomers. Biopolymers 2016; 102:369-78. [PMID: 24919990 DOI: 10.1002/bip.22516] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 06/04/2014] [Accepted: 06/08/2014] [Indexed: 11/07/2022]
Abstract
Peptomers are oligomeric molecules composed of both α-amino acids and N-substituted glycine monomers, thus creating a hybrid of peptide and peptoid units. Peptomers have been used in several applications such as antimicrobials, protease inhibitors, and antibody mimics. Despite the considerable promise of peptomers as chemically diverse molecular scaffolds, we know little about their conformational tendencies. This lack of knowledge limits the ability to implement computational approaches for peptomer design. Here we computationally evaluate the local structural propensities of the peptide-peptoid linkage. We find some general similarities between the peptide residue conformational preferences and the Ramachandran distribution of residues that precede proline in folded protein structures. However, there are notable differences. For example, several β-turn motifs are disallowed when the i+2 residue is also a peptoid monomer. Significantly, the lowest energy geometry, when dispersion forces are accounted for, corresponds to a "cis-Pro touch-turn" conformation, an unusual turn motif that has been observed at protein catalytic centers and binding sites. The peptomer touch-turn thus represents a useful design element for the construction of folded oligomers capable of molecular recognition and as modules in the assembly of structurally complex peptoid-protein hybrid macromolecules.
Collapse
Affiliation(s)
- Glenn L Butterfoss
- Center for Genomics and Systems Biology, New York University Abu Dhabi, P.O. Box, 129188, Abu Dhabi, United Arab Emirates
| | | | | | | | | |
Collapse
|
15
|
Speltz EB, Nathan A, Regan L. Design of Protein-Peptide Interaction Modules for Assembling Supramolecular Structures in Vivo and in Vitro. ACS Chem Biol 2015; 10:2108-15. [PMID: 26131725 DOI: 10.1021/acschembio.5b00415] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Synthetic biology and protein origami both require protein building blocks that behave in a reliable, predictable fashion. In particular, we require protein interaction modules with known specificity and affinity. Here, we describe three designed TRAP (Tetratricopeptide Repeat Affinity Protein)-peptide interaction pairs that are functional in vivo. We show that each TRAP binds to its cognate peptide and exhibits low cross-reactivity with the peptides bound by the other TRAPs. In addition, we demonstrate that the TRAP-peptide interactions are functional in many cellular contexts. In extensions of these designs, we show that the binding affinity of a TRAP-peptide pair can be systematically varied. The TRAP-peptide pairs we present thus represent a powerful set of new building blocks that are suitable for a variety of applications.
Collapse
Affiliation(s)
- Elizabeth B. Speltz
- Department
of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06511, United States
| | - Aparna Nathan
- Department
of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06511, United States
| | - Lynne Regan
- Department
of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06511, United States
- Department
of Chemistry, Yale University, New Haven, Connecticut 06511, United States
- Integrated Graduate Program in Physical and Engineering Biology, New Haven, Connecticut 06511, United States
| |
Collapse
|
16
|
Vohidov F, Knudsen SE, Leonard PG, Ohata J, Wheadon MJ, Popp BV, Ladbury JE, Ball ZT. Potent and selective inhibition of SH3 domains with dirhodium metalloinhibitors. Chem Sci 2015; 6:4778-4783. [PMID: 29142714 PMCID: PMC5667506 DOI: 10.1039/c5sc01602a] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 06/03/2015] [Indexed: 01/23/2023] Open
Abstract
Src-family kinases (SFKs) play important roles in human biology and are key drug targets as well. However, achieving selective inhibition of individual Src-family kinases is challenging due to the high similarity within the protein family. We describe rhodium(ii) conjugates that deliver both potent and selective inhibition of Src-family SH3 domains. Rhodium(ii) conjugates offer dramatic affinity enhancements due to interactions with specific and unique Lewis-basic histidine residues near the SH3 binding interface, allowing predictable, structure-guided inhibition of SH3 targets that are recalcitrant to traditional inhibitors. In one example, a simple metallopeptide binds the Lyn SH3 domain with 6 nM affinity and exhibits functional activation of Lyn kinase under biologically relevant concentrations (EC50 ∼ 200 nM).
Collapse
Affiliation(s)
- Farrukh Vohidov
- Department of Chemistry , Rice University , 6100 Main St. , Houston , Texas , USA .
| | - Sarah E Knudsen
- Department of Chemistry , Rice University , 6100 Main St. , Houston , Texas , USA .
| | - Paul G Leonard
- Department of Genomic Medicine , Core for Biomolecular Structure and Function , University of Texas , M.D. Anderson Cancer Center , Houston , Texas , USA
| | - Jun Ohata
- Department of Chemistry , Rice University , 6100 Main St. , Houston , Texas , USA .
| | - Michael J Wheadon
- Department of Chemistry , Rice University , 6100 Main St. , Houston , Texas , USA .
| | - Brian V Popp
- Eugene Bennett Department of Chemistry , West Virginia University , 217 Clark Hall , Morgantown , West Virginia , USA
| | - John E Ladbury
- Department of Molecular and Cellular Biology , University of Leeds , LS2 9JT , UK
| | - Zachary T Ball
- Department of Chemistry , Rice University , 6100 Main St. , Houston , Texas , USA .
| |
Collapse
|
17
|
Karna N, Dębowski D, Łęgowska A, Bąchor R, Szewczuk Z, Rolka K. Peptide splicing in a double-sequence analogue of trypsin inhibitor SFTI-1 substituted in the P₁ positions by peptoid monomers. Biopolymers 2015; 104:206-12. [PMID: 25904562 DOI: 10.1002/bip.22659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/13/2015] [Accepted: 04/14/2015] [Indexed: 11/12/2022]
Abstract
Recently, we described a process of trypsin-assisted peptide splicing of analogs of trypsin inhibitor SFTI-1, that seems to be very similar to proteasome-catalyzed peptide splicing. Here, we show, for the first time, that a peptide-peptoid hybrid (peptomer) can also be spliced by trypsin. Incubation of a double sequence SFTI-1 analog, containing two peptoid monomers, with equimolar amount of trypsin leads to formation of monocyclic peptomer as the main product. We proved that the peptide bond formed by a peptoid monomer is not only digested by trypsin but also participates in the enzyme-assisted splicing process.
Collapse
Affiliation(s)
- Natalia Karna
- Department of Chemistry, University of Gdansk, Wita Stwosza 63, 80-952, Gdansk, Poland
| | - Dawid Dębowski
- Department of Chemistry, University of Gdansk, Wita Stwosza 63, 80-952, Gdansk, Poland
| | - Anna Łęgowska
- Department of Chemistry, University of Gdansk, Wita Stwosza 63, 80-952, Gdansk, Poland
| | - Remigiusz Bąchor
- Department of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383, Wroclaw, Poland
| | - Zbigniew Szewczuk
- Department of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383, Wroclaw, Poland
| | - Krzysztof Rolka
- Department of Chemistry, University of Gdansk, Wita Stwosza 63, 80-952, Gdansk, Poland
| |
Collapse
|
18
|
Yokoyama N, Miller WT. Molecular characterization of WDCP, a novel fusion partner for the anaplastic lymphoma tyrosine kinase ALK. Biomed Rep 2014; 3:9-13. [PMID: 25469238 DOI: 10.3892/br.2014.374] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 10/20/2014] [Indexed: 11/06/2022] Open
Abstract
Anaplastic lymphoma kinase (ALK) is a member of the receptor tyrosine kinase superfamily. The ALK gene is a site of frequent mutation and chromosomal rearrangement in various types of human cancers. A novel chromosomal translocation was recently identified in human colorectal cancer between the ALK gene and chromosome 2, open reading frame 44 (C2orf44), a gene of unknown function. As a first step in understanding the oncogenic properties of this fusion protein, C2orf44 cDNA was cloned and the encoded protein was characterized, which was designated as WD repeat and coiled coil containing protein (WDCP). A C-terminal proline-rich segment in WDCP was shown to mediate binding to the Src homology 3 domain of the Src family kinase hematopoietic cell kinase (Hck). Co-expression with Hck lead to tyrosine phosphorylation of WDCP. Chromatographic fractionation of WDCP-containing lysates indicates that the protein exists as an oligomer in mammalian cells. These results suggest that, in the context of the ALK-C2orf44 gene fusion, WDCP imposes an oligomeric structure on ALK that results in constitutive kinase activation and signaling.
Collapse
Affiliation(s)
- Noriko Yokoyama
- Department of Physiology and Biophysics, School of Medicine, State University of New York at Stony Brook, Stony Brook, NY 11794, USA
| | - W Todd Miller
- Department of Physiology and Biophysics, School of Medicine, State University of New York at Stony Brook, Stony Brook, NY 11794, USA
| |
Collapse
|
19
|
Register AC, Leonard SE, Maly DJ. SH2-catalytic domain linker heterogeneity influences allosteric coupling across the SFK family. Biochemistry 2014; 53:6910-23. [PMID: 25302671 PMCID: PMC4230323 DOI: 10.1021/bi5008194] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Src-family
kinases (SFKs) make up a family of nine homologous multidomain
tyrosine kinases whose misregulation is responsible for human disease
(cancer, diabetes, inflammation, etc.). Despite overall sequence homology
and identical domain architecture, differences in SH3 and SH2 regulatory
domain accessibility and ability to allosterically autoinhibit the
ATP-binding site have been observed for the prototypical SFKs Src
and Hck. Biochemical and structural studies indicate that the SH2-catalytic
domain (SH2-CD) linker, the intramolecular binding epitope for SFK
SH3 domains, is responsible for allosterically coupling SH3 domain
engagement to autoinhibition of the ATP-binding site through the conformation
of the αC helix. As a relatively unconserved region between
SFK family members, SH2-CD linker sequence variability across the
SFK family is likely a source of nonredundant cellular functions between
individual SFKs via its effect on the availability of SH3 and SH2
domains for intermolecular interactions and post-translational modification.
Using a combination of SFKs engineered with enhanced or weakened regulatory
domain intramolecular interactions and conformation-selective inhibitors
that report αC helix conformation, this study explores how SH2-CD
sequence heterogeneity affects allosteric coupling across the SFK
family by examining Lyn, Fyn1, and Fyn2. Analyses of Fyn1 and Fyn2,
isoforms that are identical but for a 50-residue sequence spanning
the SH2-CD linker, demonstrate that SH2-CD linker sequence differences
can have profound effects on allosteric coupling between otherwise
identical kinases. Most notably, a dampened allosteric connection
between the SH3 domain and αC helix leads to greater autoinhibitory
phosphorylation by Csk, illustrating the complex effects of SH2-CD
linker sequence on cellular function.
Collapse
Affiliation(s)
- A C Register
- Department of Chemistry, University of Washington , Seattle, Washington 98195, United States
| | | | | |
Collapse
|
20
|
Li W, Gigante A, Perez-Perez MJ, Yue H, Hirano M, McIntyre TM, Silverstein RL. Thymidine phosphorylase participates in platelet signaling and promotes thrombosis. Circ Res 2014; 115:997-1006. [PMID: 25287063 DOI: 10.1161/circresaha.115.304591] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Platelets contain abundant thymidine phosphorylase (TYMP), which is highly expressed in diseases with high risk of thrombosis, such as atherosclerosis and type II diabetes mellitus. OBJECTIVE To test the hypothesis that TYMP participates in platelet signaling and promotes thrombosis. METHODS AND RESULTS By using a ferric chloride (FeCl3)-induced carotid artery injury thrombosis model, we found time to blood flow cessation was significantly prolonged in Tymp(-/-) and Tymp(+/-) mice compared with wild-type mice. Bone marrow transplantation and platelet transfusion studies demonstrated that platelet TYMP was responsible for the antithrombotic phenomenon in the TYMP-deficient mice. Collagen-, collagen-related peptide-, adenosine diphosphate-, or thrombin-induced platelet aggregation were significantly attenuated in Tymp(+/-) and Tymp(-/-) platelets, and in wild type or human platelets pretreated with TYMP inhibitor KIN59. Tymp deficiency also significantly decreased agonist-induced P-selectin expression. TYMP contains an N-terminal SH3 domain-binding proline-rich motif and forms a complex with the tyrosine kinases Lyn, Fyn, and Yes in platelets. TYMP-associated Lyn was inactive in resting platelets, and TYMP trapped and diminished active Lyn after collagen stimulation. Tymp/Lyn double haploinsufficiency diminished the antithrombotic phenotype of Tymp(+/-) mice. TYMP deletion or inhibition of TYMP with KIN59 dramatically increased platelet-endothelial cell adhesion molecule 1 tyrosine phosphorylation and diminished collagen-related peptide- or collagen-induced AKT phosphorylation. In vivo administration of KIN59 significantly inhibited FeCl3-induced carotid artery thrombosis without affecting hemostasis. CONCLUSIONS TYMP participates in multiple platelet signaling pathways and regulates platelet activation and thrombosis. Targeting TYMP might be a novel antiplatelet and antithrombosis therapy.
Collapse
Affiliation(s)
- Wei Li
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute, The Cleveland Clinic, OH (W.L., T.M.M.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, OH (W.L., T.M.M.); Instituto de Quimica Medica, Consejo Superior De Investigaciones Cientificas (IQM-CSIC), Madrid, Spain (A.G.,M.-J.P.-P.); Department of Biological Sciences, Case Western Reserve University, Cleveland, OH (H.Y.); Department of Neurology, Columbia University Medical Center, New York, NY (M.H.); and Department of Medicine, Medical College of Wisconsin and Blood Research Institute, Blood Center of Wisconsin, Milwaukee (R.L.S.)
| | - Alba Gigante
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute, The Cleveland Clinic, OH (W.L., T.M.M.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, OH (W.L., T.M.M.); Instituto de Quimica Medica, Consejo Superior De Investigaciones Cientificas (IQM-CSIC), Madrid, Spain (A.G.,M.-J.P.-P.); Department of Biological Sciences, Case Western Reserve University, Cleveland, OH (H.Y.); Department of Neurology, Columbia University Medical Center, New York, NY (M.H.); and Department of Medicine, Medical College of Wisconsin and Blood Research Institute, Blood Center of Wisconsin, Milwaukee (R.L.S.)
| | - Maria-Jesus Perez-Perez
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute, The Cleveland Clinic, OH (W.L., T.M.M.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, OH (W.L., T.M.M.); Instituto de Quimica Medica, Consejo Superior De Investigaciones Cientificas (IQM-CSIC), Madrid, Spain (A.G.,M.-J.P.-P.); Department of Biological Sciences, Case Western Reserve University, Cleveland, OH (H.Y.); Department of Neurology, Columbia University Medical Center, New York, NY (M.H.); and Department of Medicine, Medical College of Wisconsin and Blood Research Institute, Blood Center of Wisconsin, Milwaukee (R.L.S.)
| | - Hong Yue
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute, The Cleveland Clinic, OH (W.L., T.M.M.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, OH (W.L., T.M.M.); Instituto de Quimica Medica, Consejo Superior De Investigaciones Cientificas (IQM-CSIC), Madrid, Spain (A.G.,M.-J.P.-P.); Department of Biological Sciences, Case Western Reserve University, Cleveland, OH (H.Y.); Department of Neurology, Columbia University Medical Center, New York, NY (M.H.); and Department of Medicine, Medical College of Wisconsin and Blood Research Institute, Blood Center of Wisconsin, Milwaukee (R.L.S.)
| | - Michio Hirano
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute, The Cleveland Clinic, OH (W.L., T.M.M.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, OH (W.L., T.M.M.); Instituto de Quimica Medica, Consejo Superior De Investigaciones Cientificas (IQM-CSIC), Madrid, Spain (A.G.,M.-J.P.-P.); Department of Biological Sciences, Case Western Reserve University, Cleveland, OH (H.Y.); Department of Neurology, Columbia University Medical Center, New York, NY (M.H.); and Department of Medicine, Medical College of Wisconsin and Blood Research Institute, Blood Center of Wisconsin, Milwaukee (R.L.S.)
| | - Thomas M McIntyre
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute, The Cleveland Clinic, OH (W.L., T.M.M.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, OH (W.L., T.M.M.); Instituto de Quimica Medica, Consejo Superior De Investigaciones Cientificas (IQM-CSIC), Madrid, Spain (A.G.,M.-J.P.-P.); Department of Biological Sciences, Case Western Reserve University, Cleveland, OH (H.Y.); Department of Neurology, Columbia University Medical Center, New York, NY (M.H.); and Department of Medicine, Medical College of Wisconsin and Blood Research Institute, Blood Center of Wisconsin, Milwaukee (R.L.S.)
| | - Roy L Silverstein
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute, The Cleveland Clinic, OH (W.L., T.M.M.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, OH (W.L., T.M.M.); Instituto de Quimica Medica, Consejo Superior De Investigaciones Cientificas (IQM-CSIC), Madrid, Spain (A.G.,M.-J.P.-P.); Department of Biological Sciences, Case Western Reserve University, Cleveland, OH (H.Y.); Department of Neurology, Columbia University Medical Center, New York, NY (M.H.); and Department of Medicine, Medical College of Wisconsin and Blood Research Institute, Blood Center of Wisconsin, Milwaukee (R.L.S.)
| |
Collapse
|
21
|
Renfrew PD, Craven TW, Butterfoss G, Kirshenbaum K, Bonneau R. A rotamer library to enable modeling and design of peptoid foldamers. J Am Chem Soc 2014; 136:8772-82. [PMID: 24823488 PMCID: PMC4227732 DOI: 10.1021/ja503776z] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Indexed: 01/08/2023]
Abstract
Peptoids are a family of synthetic oligomers composed of N-substituted glycine units. Along with other "foldamer" systems, peptoid oligomer sequences can be predictably designed to form a variety of stable secondary structures. It is not yet evident if foldamer design can be extended to reliably create tertiary structure features that mimic more complex biomolecular folds and functions. Computational modeling and prediction of peptoid conformations will likely play a critical role in enabling complex biomimetic designs. We introduce a computational approach to provide accurate conformational and energetic parameters for peptoid side chains needed for successful modeling and design. We find that peptoids can be described by a "rotamer" treatment, similar to that established for proteins, in which the peptoid side chains display rotational isomerism to populate discrete regions of the conformational landscape. Because of the insufficient number of solved peptoid structures, we have calculated the relative energies of side-chain conformational states to provide a backbone-dependent (BBD) rotamer library for a set of 54 different peptoid side chains. We evaluated two rotamer library development methods that employ quantum mechanics (QM) and/or molecular mechanics (MM) energy calculations to identify side-chain rotamers. We show by comparison to experimental peptoid structures that both methods provide an accurate prediction of peptoid side chain placements in folded peptoid oligomers and at protein interfaces. We have incorporated our peptoid rotamer libraries into ROSETTA, a molecular design package previously validated in the context of protein design and structure prediction.
Collapse
Affiliation(s)
- P. Douglas Renfrew
- Center for Genomics and
Systems Biology, Department
of Biology, Department of Chemistry, and Courant Institute of Mathematical
Sciences, Computer Science Department, New
York University, New York, New York 10003, United States
| | - Timothy W. Craven
- Center for Genomics and
Systems Biology, Department
of Biology, Department of Chemistry, and Courant Institute of Mathematical
Sciences, Computer Science Department, New
York University, New York, New York 10003, United States
| | - Glenn
L. Butterfoss
- Center
for Genomics and Systems Biology, New York
University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Kent Kirshenbaum
- Center for Genomics and
Systems Biology, Department
of Biology, Department of Chemistry, and Courant Institute of Mathematical
Sciences, Computer Science Department, New
York University, New York, New York 10003, United States
| | - Richard Bonneau
- Center for Genomics and
Systems Biology, Department
of Biology, Department of Chemistry, and Courant Institute of Mathematical
Sciences, Computer Science Department, New
York University, New York, New York 10003, United States
| |
Collapse
|
22
|
He P, Wu W, Wang HD, Liao KL, Zhang W, Lv FL, Yang K. Why ligand cross-reactivity is high within peptide recognition domain families? A case study on human c-Src SH3 domain. J Theor Biol 2014; 340:30-7. [DOI: 10.1016/j.jtbi.2013.08.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 07/26/2013] [Accepted: 08/21/2013] [Indexed: 10/26/2022]
|
23
|
Fuller AA, Holmes CA, Seidl FJ. A fluorescent peptoid pH-sensor. Biopolymers 2013; 100:380-6. [DOI: 10.1002/bip.22242] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 03/17/2013] [Indexed: 01/30/2023]
Affiliation(s)
- Amelia A. Fuller
- Department of Chemistry & Biochemistry; Santa Clara University; 500 El Camino Real; Santa Clara; CA; 95053
| | - Courtney A. Holmes
- Department of Chemistry & Biochemistry; Santa Clara University; 500 El Camino Real; Santa Clara; CA; 95053
| | - Frederick J. Seidl
- Department of Chemistry & Biochemistry; Santa Clara University; 500 El Camino Real; Santa Clara; CA; 95053
| |
Collapse
|
24
|
Sun J, Zuckermann RN. Peptoid polymers: a highly designable bioinspired material. ACS NANO 2013; 7:4715-32. [PMID: 23721608 DOI: 10.1021/nn4015714] [Citation(s) in RCA: 317] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Bioinspired polymeric materials are attracting increasing attention due to significant advantages over their natural counterparts: the ability to precisely tune their structures over a broad range of chemical and physical properties, increased stability, and improved processability. Polypeptoids, a promising class of bioinspired polymer based on a N-substituted glycine backbone, have a number of unique properties that bridge the material gap between proteins and bulk polymers. Peptoids combine the sequence specificity of biopolymers with the simpler intra/intermolecular interactions and robustness of traditional synthetic polymers. They are highly designable because hundreds of chemically diverse side chains can be introduced from simple building blocks. Peptoid polymers can be prepared by two distinct synthetic techniques offering access to two material subclasses: (1) automated solid-phase synthesis which enables precision sequence control and near absolute monodispersity up to chain lengths of ~50 monomers, and (2) a classical polymerization approach which allows access to higher molecular weights and larger-scale yields, but with less control over length and sequence. This combination of facile synthetic approaches makes polypeptoids a highly tunable, rapid polymer prototyping platform to investigate new materials that are intermediate between proteins and bulk polymers, in both their structure and their properties. In this paper, we review the methods to synthesize peptoid polymers and their applications in biomedicine and nanoscience, as both sequence-specific materials and as bulk polymers.
Collapse
Affiliation(s)
- Jing Sun
- Molecular Foundry, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, California 94720, USA
| | | |
Collapse
|
25
|
Park M, Wetzler M, Jardetzky TS, Barron AE. A readily applicable strategy to convert peptides to peptoid-based therapeutics. PLoS One 2013; 8:e58874. [PMID: 23555603 PMCID: PMC3605428 DOI: 10.1371/journal.pone.0058874] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Accepted: 02/07/2013] [Indexed: 01/23/2023] Open
Abstract
Incorporation of unnatural amino acids and peptidomimetic residues into therapeutic peptides is highly efficacious and commonly employed, but generally requires laborious trial-and-error approaches. Previously, we demonstrated that C20 peptide has the potential to be a potential antiviral agent. Herein we report our attempt to improve the biological properties of this peptide by introducing peptidomimetics. Through combined alanine, proline, and sarcosine scans coupled with a competitive fluorescence polarization assay developed for identifying antiviral peptides, we enabled to pinpoint peptoid-tolerant peptide residues within C20 peptide. The synergistic benefits of combining these (and other) commonly employed methods could lead to a easily applicable strategy for designing and refining therapeutically-attractive peptidomimetics.
Collapse
Affiliation(s)
- Minyoung Park
- Department of Chemical and Systems Biology, School of Medicine, Stanford University, Stanford, California, United States of America
| | - Modi Wetzler
- Department of Bioengineering, Schools of Engineering and Medicine, Stanford University, Stanford, California, United States of America
| | - Theodore S. Jardetzky
- Department of Structural Biology, School of Medicine, Stanford University, Stanford, California, United States of America
| | - Annelise E. Barron
- Department of Bioengineering, Schools of Engineering and Medicine, Stanford University, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
26
|
Wendt MD. Discovery of ABT-263, a Bcl-family protein inhibitor: observations on targeting a large protein-protein interaction. Expert Opin Drug Discov 2013; 3:1123-43. [PMID: 23506184 DOI: 10.1517/17460441.3.9.1123] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The discovery of ABT-263, a rationally designed Bcl-2/Bcl-xL inhibitor at present in Phase I clinical trials for cancer, is described. Emphasis is placed on the specific hurdles overcome throughout the discovery process that relate to the nature of the targeted protein-protein interaction (PPI). OBJECTIVE/METHODS This review draws on observations from the experience of discovering ABT-263 and discusses them within the framework of the larger issue of discovering drugs targeting PPIs. Issues discussed include the 'hot spot' paradigm, hit and lead generation, serum protein binding, structure-based design, and in particular, hydrophobicity and molecular size and their relation to pharmacokinetic/pharmacodynamic properties. RESULTS/CONCLUSION Approaches to understanding obstacles thought of as being specifically attached to PPIs, and existing techniques to combat these obstacles, were very helpful in overcoming them. The example of ABT-263 provides evidence that the larger family of PPI targets is more tractable than may have been thought.
Collapse
Affiliation(s)
- Michael D Wendt
- Abbott Laboratories, Cancer Research, Global Pharmaceutical R&D, Dept R4N6, Bldg. AP10-3, 100 Abbott Park Road, Abbott Park, Illinois 60064, USA +1 847 937 9305 ; +1 847 938 1004 ;
| |
Collapse
|
27
|
Wu Z, Tan M, Chen X, Yang Z, Wang L. Molecular hydrogelators of peptoid-peptide conjugates with superior stability against enzyme digestion. NANOSCALE 2012; 4:3644-3646. [PMID: 22581113 DOI: 10.1039/c2nr30408b] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
We report on molecular hydrogelators based on peptoid-peptide conjugates with good biocompatibility to different cells and superior stability against proteinase K digestion.
Collapse
Affiliation(s)
- Zhidan Wu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, PR China
| | | | | | | | | |
Collapse
|
28
|
Liu F, Park JE, Qian WJ, Lim D, Scharow A, Berg T, Yaffe MB, Lee KS, Burke TR. Peptoid-Peptide hybrid ligands targeting the polo box domain of polo-like kinase 1. Chembiochem 2012; 13:1291-6. [PMID: 22570300 PMCID: PMC4536914 DOI: 10.1002/cbic.201200206] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Indexed: 11/10/2022]
Abstract
We replaced the amino terminal Pro residue of the Plk1 polo-box-domain-binding pentapeptide (PLHSpT) with a library of N-alkyl-Gly "peptoids", and identified long-chain tethered phenyl moieties giving greater than two-orders-of-magnitude affinity enhancement. Further simplification by replacing the peptoid residue with appropriate amides gave low-nanomolar affinity N-acylated tetrapeptides. Binding of the N-terminal long-chain phenyl extension was demonstrated by X-ray co-crystal data.
Collapse
Affiliation(s)
- Fa Liu
- Chemical Biology Laboratory, Frederick National Lab, NCI, NIH, Frederick, MD 21702 (USA), Fax: (+1) 301-846-6033
| | - Jung-Eun Park
- Laboratory of Metabolism, National Cancer Institute, NIH, Bethesda, MD 20892 (USA)
| | - Wen-Jian Qian
- Chemical Biology Laboratory, Frederick National Lab, NCI, NIH, Frederick, MD 21702 (USA), Fax: (+1) 301-846-6033
| | - Dan Lim
- Center for Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 (USA)
| | - Andrej Scharow
- Institute of Organic Chemistry, University of Leipzig, 04103 Leipzig (Germany)
| | - Thorsten Berg
- Institute of Organic Chemistry, University of Leipzig, 04103 Leipzig (Germany)
| | - Michael B. Yaffe
- Center for Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 (USA)
| | - Kyung S. Lee
- Laboratory of Metabolism, National Cancer Institute, NIH, Bethesda, MD 20892 (USA)
| | - Terrence R. Burke
- Chemical Biology Laboratory, Frederick National Lab, NCI, NIH, Frederick, MD 21702 (USA), Fax: (+1) 301-846-6033
| |
Collapse
|
29
|
Halova I, Draberova L, Draber P. Mast cell chemotaxis - chemoattractants and signaling pathways. Front Immunol 2012; 3:119. [PMID: 22654878 PMCID: PMC3360162 DOI: 10.3389/fimmu.2012.00119] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 04/24/2012] [Indexed: 01/09/2023] Open
Abstract
Migration of mast cells is essential for their recruitment within target tissues where they play an important role in innate and adaptive immune responses. These processes rely on the ability of mast cells to recognize appropriate chemotactic stimuli and react to them by a chemotactic response. Another level of intercellular communication is attained by production of chemoattractants by activated mast cells, which results in accumulation of mast cells and other hematopoietic cells at the sites of inflammation. Mast cells express numerous surface receptors for various ligands with properties of potent chemoattractants. They include the stem cell factor (SCF) recognized by c-Kit, antigen, which binds to immunoglobulin E (IgE) anchored to the high affinity IgE receptor (FcεRI), highly cytokinergic (HC) IgE recognized by FcεRI, lipid mediator sphingosine-1-phosphate (S1P), which binds to G protein-coupled receptors (GPCRs). Other large groups of chemoattractants are eicosanoids [prostaglandin E2 and D2, leukotriene (LT) B4, LTD4, and LTC4, and others] and chemokines (CC, CXC, C, and CX3C), which also bind to various GPCRs. Further noteworthy chemoattractants are isoforms of transforming growth factor (TGF) β1–3, which are sensitively recognized by TGF-β serine/threonine type I and II β receptors, adenosine, C1q, C3a, and C5a components of the complement, 5-hydroxytryptamine, neuroendocrine peptide catestatin, tumor necrosis factor-α, and others. Here we discuss the major types of chemoattractants recognized by mast cells, their target receptors, as well as signaling pathways they utilize. We also briefly deal with methods used for studies of mast cell chemotaxis and with ways of how these studies profited from the results obtained in other cellular systems.
Collapse
Affiliation(s)
- Ivana Halova
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic Prague, Czech Republic
| | | | | |
Collapse
|
30
|
Interfacial water molecules in SH3 interactions: Getting the full picture on polyproline recognition by protein-protein interaction domains. FEBS Lett 2012; 586:2619-30. [DOI: 10.1016/j.febslet.2012.04.057] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 04/27/2012] [Accepted: 04/30/2012] [Indexed: 01/16/2023]
|
31
|
Abstract
Signaling cascades are managed in time and space by interactions between and among proteins. These interactions are often aided by adaptor proteins, which guide enzyme-substrate pairs into proximity. Miniature proteins are a class of small, well-folded protein domains possessing engineered binding properties. Here we made use of two miniature proteins with complementary binding properties to create a synthetic adaptor protein that effectively redirects a ubiquitous signaling event: tyrosine phosphorylation. We report that miniature-protein-based adaptor 3 uses templated catalysis to redirect the Src family kinase Hck to phosphorylate hDM2, a negative regulator of the p53 tumor suppressor and a poor Hck substrate. Phosphorylation occurs with multiple turnover and at a single site targeted by c-Abl kinase in the cell.
Collapse
Affiliation(s)
- Elissa M. Hobert
- Department of Chemistry, Cellular and Developmental Biology, Yale University, New Haven, CT 06520-8107, United States
| | - Alanna Schepartz
- Department of Chemistry, Cellular and Developmental Biology, Yale University, New Haven, CT 06520-8107, United States
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520-8107, United States
| |
Collapse
|
32
|
Abstract
Peptoid oligomers were initially developed as part of a larger basic research effort to accelerate the drug-discovery process in the biotech/biopharma industry. Their ease of synthesis, stability, and structural similarity to polypeptides made them ideal candidates for the combinatorial discovery of novel peptidomimetic drug candidates. Diverse libraries of short peptoid oligomers provided one of the first demonstrations in the mid-1990s that high-affinity ligands to pharmaceutically relevant receptors could be discovered from combinatorial libraries of synthetic compounds. The solid-phase submonomer method of peptoid synthesis was so efficient and general that it soon became possible to explore the properties of longer polypeptoid chains in a variety of areas beyond drug discovery (e.g., diagnostics, drug delivery, and materials science). Exploration into protein-mimetic materials soon followed, with the fundamental goal of folding a non-natural sequence-specific heteropolymer into defined secondary or tertiary structures. This effort first yielded the peptoid helix and much later the peptoid sheet, both of which are secondary-structure mimetics that are close relatives to their natural counterparts. These crucial discoveries have brought us closer to building proteinlike structure and function from a non-natural polymer and have provided great insight into the rules governing polymer and protein folding. The accessibility of peptoid synthesis to chemists and nonchemists alike, along with a lack of information-rich non-natural polymers available to study, has led to a rapid growth in the field of peptoid science by many new investigators. This work provides an overview of the initial discovery and early developments in the peptoid field.
Collapse
Affiliation(s)
- Ronald N Zuckermann
- The Molecular Foundry, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA.
| |
Collapse
|
33
|
Balakrishnan S, Scheuermann MJ, Zondlo NJ. Arginine mimetics using α-guanidino acids: introduction of functional groups and stereochemistry adjacent to recognition guanidiniums in peptides. Chembiochem 2012; 13:259-70. [PMID: 22213184 PMCID: PMC3712784 DOI: 10.1002/cbic.201100638] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Indexed: 01/19/2023]
Abstract
Arginine residues are broadly employed for specific biomolecular recognition, including in protein-protein, protein-DNA, and protein-RNA interactions. Arginine recognition commonly exploits the potential for bidentate electrostatic and hydrogen-bonding interactions. However, in arginine residues, the guanidinium functional group is located at the terminus of a flexible hydrocarbon side chain, which lacks the functionality to contribute to specific arginine-mediated recognition and may entropically disfavor binding. In order to enhance the potential for specificity and affinity in arginine-mediated molecular recognition, we have developed an approach to the synthesis of peptides that incorporates an α-guanidino acid as a novel arginine mimetic. α-Guanidino acids, derived from α-amino acids, with guanidinylation of the amino group, were incorporated stereospecifically into peptides on solid phase via coupling of an Fmoc amino acid to diaminopropionic acid (Dap), Fmoc deprotection, guanidinylation of the amine on solid phase, and deprotection, generating a peptide containing an α-functionalized arginine mimetic. This approach was examined by incorporating arginine mimetics into ligands for the Src, Grb, and Crk SH3 domains at the site of the key recognition arginine. Protein binding was examined for peptides containing guanidino acids derived from Gly, L-Val, L-Phe, L-Trp, D-Val, D-Phe, and D-Trp. We demonstrate that paralogue specificity and target site affinity may be modulated with the use of α-guanidino acid-derived arginine mimetics, generating peptides that exhibit enhanced Src specificity by selection against Grb and peptides that reverse the specificity of the native peptide ligand, with enhancements in Src target specificity of up to 15-fold (1.6 kcal mol(-1)).
Collapse
Affiliation(s)
- Shalini Balakrishnan
- Department of Chemistry and Biochemistry University of Delaware Newark, Delaware 19716 (USA)
| | - Michael J. Scheuermann
- Department of Chemistry and Biochemistry University of Delaware Newark, Delaware 19716 (USA)
| | - Neal J. Zondlo
- Department of Chemistry and Biochemistry University of Delaware Newark, Delaware 19716 (USA)
| |
Collapse
|
34
|
Suppiah S, Mousa HA, Tzeng WP, Matthews JD, Frey TK. Binding of cellular p32 protein to the rubella virus P150 replicase protein via PxxPxR motifs. J Gen Virol 2012; 93:807-816. [PMID: 22238231 DOI: 10.1099/vir.0.038901-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A proline-rich region (PRR) within the rubella virus (RUBV) P150 replicase protein that contains three SH3 domain-binding motifs (PxxPxR) was investigated for its ability to bind cell proteins. Pull-down experiments using a glutathione S-transferase-PRR fusion revealed PxxPxR motif-specific binding with human p32 protein (gC1qR), which could be mediated by either of the first two motifs. This finding was of interest because p32 protein also binds to the RUBV capsid protein. Binding of p32 to P150 was confirmed and was abolished by mutation of the first two motifs. When mutations in the first two motifs were introduced into a RUBV cDNA infectious clone, virus replication was significantly impaired. However, virus RNA synthesis was found to be unaffected, and subsequent immunofluorescence analysis of RUBV-infected cells revealed co-localization of p32 and P150 but little overlap of p32 with RNA replication complexes, indicating that p32 does not participate directly in virus RNA synthesis. Thus, the role of p32 in RUBV replication remains unresolved.
Collapse
Affiliation(s)
- Suganthi Suppiah
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Heather A Mousa
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Wen-Pin Tzeng
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Jason D Matthews
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Teryl K Frey
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
35
|
Millward SW, Henning RK, Kwong GA, Pitram S, Agnew HD, Deyle KM, Nag A, Hein J, Lee SS, Lim J, Pfeilsticker JA, Sharpless KB, Heath JR. Iterative in situ click chemistry assembles a branched capture agent and allosteric inhibitor for Akt1. J Am Chem Soc 2011; 133:18280-8. [PMID: 21962254 DOI: 10.1021/ja2064389] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
We describe the use of iterative in situ click chemistry to design an Akt-specific branched peptide triligand that is a drop-in replacement for monoclonal antibodies in multiple biochemical assays. Each peptide module in the branched structure makes unique contributions to affinity and/or specificity resulting in a 200 nM affinity ligand that efficiently immunoprecipitates Akt from cancer cell lysates and labels Akt in fixed cells. Our use of a small molecule to preinhibit Akt prior to screening resulted in low micromolar inhibitory potency and an allosteric mode of inhibition, which is evidenced through a series of competitive enzyme kinetic assays. To demonstrate the efficiency and selectivity of the protein-templated in situ click reaction, we developed a novel QPCR-based methodology that enabled a quantitative assessment of its yield. These results point to the potential for iterative in situ click chemistry to generate potent, synthetically accessible antibody replacements with novel inhibitory properties.
Collapse
Affiliation(s)
- Steven W Millward
- Nanosystems Biology Cancer Center, Division of Chemistry and Chemical Engineering, MC-127-72, California Institute of Technology, Pasadena, California 91125, United States
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Park SH, Szleifer I. Structural and dynamical characteristics of peptoid oligomers with achiral aliphatic side chains studied by molecular dynamics simulation. J Phys Chem B 2011; 115:10967-75. [PMID: 21819112 PMCID: PMC3177600 DOI: 10.1021/jp2025957] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
All-atom molecular dynamics simulations of N-substituted glycine peptoid oligomers with methyl and methoxyethyl side chains have been carried out for chain lengths of 5, 10, 20, and 50 residues in aqueous phase at room temperature. The (ϕ, ψ) backbone dihedral angle distributions in the Ramachandran plots show that helical structures, similar to polyproline type I and type II helices, are the most favorable conformations in most peptoid oligomers studied. The left-handed helical structures are shown to be increasingly favored as the oligomer chain length grows. A significant population of cis amide bond configurations has been identified in the peptoid oligomers. By combining the analysis of ϕ and ω backbone dihedral angles, we determined the relative composition of the four major conformations favored by the backbone dihedral angles. The trans α(D) conformation is found to be most favored for all peptoid oligomers studies. The time correlation functions of the end-to-end distance highlight a rigid backbone structure relative to side chains for peptoid oligomers. The transition between right-handed and left-handed helical conformations is found to be very rare and between cis and trans isomerism in the amide bond completely absent in the simulation time scale. The radii of gyration for all peptoid oligomers have been found to be consistently larger in comparison to the peptide counterparts, suggesting slightly open structures for peptoids relative to peptides, whereas the fluctuations in the radius of gyration support a rigid backbone structure of peptoids.
Collapse
Affiliation(s)
- Sung Hyun Park
- Department of Biomedical Engineering and Chemistry of Life Processes, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, U.S.A
| | - Igal Szleifer
- Department of Biomedical Engineering and Chemistry of Life Processes, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, U.S.A
| |
Collapse
|
37
|
Moure A, Sanclimens G, Bujons J, Masip I, Alvarez-Larena A, Pérez-Payá E, Alfonso I, Messeguer A. Chemical modulation of peptoids: synthesis and conformational studies on partially constrained derivatives. Chemistry 2011; 17:7927-39. [PMID: 21611988 DOI: 10.1002/chem.201100216] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Indexed: 01/16/2023]
Abstract
The high conformational flexibility of peptoids can generate problems in biomolecular selectivity as a result of undesired off-target interactions. This drawback can be counterbalanced by restricting the original flexibility to a certain extent, thus leading to new peptidomimetics. By starting from the structure of an active peptoid as an apoptosis inhibitor, we designed two families of peptidomimetics that bear either 7-substituted perhydro-1,4-diazepine-2,5-dione 2 or 3-substituted 1,4-piperazine-2,5-dione 3 moieties. We report an efficient, solid-phase-based synthesis for both peptidomimetic families 2 and 3 from a common intermediate. An NMR spectroscopic study of 2a,b and 3a,b showed two species in solution in different solvents that interconvert slowly on the NMR timescale. The cis/trans isomerization around the exocyclic tertiary amide bond is responsible for this conformational behavior. The cis isomers are more favored in nonpolar environments, and this preference is higher for the six-membered-ring derivative 3a,b. We propose that the hydrogen-bonding pattern could play an important role in the cis/trans equilibrium process. These hydrogen bonds were characterized in solution, in the solid state (i.e., by using X-ray studies), and by molecular modeling of simplified systems. A comparative study of a model peptoid 10 containing the isolated tertiary amide bond under study outlined the importance of the heterocyclic moiety for the prevalence of the cis configuration in 2a and 3a. The kinetics of the cis/trans interconversion in 2a, 3a, and 10 was also studied by variable-temperature NMR spectroscopic analysis. The full line-shape analysis of the NMR spectra of 10 revealed negligible entropic contribution to the energetic barrier in this conformational process. A theoretical analysis of 10 supported the results observed by NMR spectroscopic analysis. Overall, these results are relevant for the study of the peptidomimetic/biological-target interactions.
Collapse
Affiliation(s)
- Alejandra Moure
- Department of Chemical and Biomolecular Nanotechnology, Instituto de Química Avanzada de Cataluña, Consejo Superior de Investigaciones Científicas, J. Girona, 18, 08034 Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Kim SE, Liu F, Im YJ, Stephen AG, Fivash MJ, Waheed AA, Freed EO, Fisher RJ, Hurley JH, Burke TR. Elucidation of New Binding Interactions with the Tumor Susceptibility Gene 101 (Tsg101) Protein Using Modified HIV-1 Gag-p6 Derived Peptide Ligands. ACS Med Chem Lett 2011; 2:337-341. [PMID: 21643473 DOI: 10.1021/ml1002579] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Targeting protein-protein interactions is gaining greater recognition as an attractive approach to therapeutic development. An example of this may be found with the human cellular protein encoded by the tumor susceptibility gene 101 (Tsg101), where interaction with the p6 C-terminal domain of the nascent viral Gag protein is required for HIV-1 particle budding and release. This association of Gag with Tsg101 is highly dependent on a "Pro-Thr-Ala-Pro" ("PTAP") peptide sequence within the p6 protein. Although p6-derived peptides offer potential starting points for developing Tsg101-binding inhibitors, the affinities of canonical peptides are outside the useful range (K(d) values greater than 50 μM). Reported herein are crystal structures of Tsg101 in complex with two structurally-modified PTAP-derived peptides. This data define new regions of ligand interaction not previously identified with canonical peptide sequences. This information could be highly useful in the design of Tsg101-binding antagonists.
Collapse
Affiliation(s)
- Sung-Eun Kim
- Chemical Biology Laboratory, Molecular, Discovery Program, CCR, NCI-Frederick, Frederick, Maryland 21702, United States
| | - Fa Liu
- Chemical Biology Laboratory, Molecular, Discovery Program, CCR, NCI-Frederick, Frederick, Maryland 21702, United States
| | | | | | - Matthew J. Fivash
- Data Management Systems, Inc., NCI-Frederick, Frederick, Maryland 21702, United States
| | - Abdul A. Waheed
- HIV Drug Resistance Program, CCR, NCI-Frederick, Frederick, Maryland 21702, United States
| | - Eric O. Freed
- HIV Drug Resistance Program, CCR, NCI-Frederick, Frederick, Maryland 21702, United States
| | | | | | - Terrence R. Burke
- Chemical Biology Laboratory, Molecular, Discovery Program, CCR, NCI-Frederick, Frederick, Maryland 21702, United States
| |
Collapse
|
39
|
Park M, Jardetzky TS, Barron AE. NMEGylation: A novel modification to enhance the bioavailability of therapeutic peptides. Biopolymers 2011; 96:688-93. [DOI: 10.1002/bip.21607] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
40
|
Fuller AA, Seidl FJ, Bruno PA, Plescia MA, Palla KS. Use of the environmentally sensitive fluorophore 4-N,N-dimethylamino-1,8-naphthalimide to study peptoid helix structures. Biopolymers 2011; 96:627-38. [DOI: 10.1002/bip.21605] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
41
|
Guo L, Li J, Brown Z, Ghale K, Zhang D. Synthesis and characterization of cyclic and linear helical poly(α-peptoid)s by N-heterocyclic carbene-mediated ring-opening polymerizations of N-substituted N-carboxyanhydrides. Biopolymers 2011; 96:596-603. [DOI: 10.1002/bip.21597] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
42
|
Gao CM, Yam AY, Wang X, Magdangal E, Salisbury C, Peretz D, Zuckermann RN, Connolly MD, Hansson O, Minthon L, Zetterberg H, Blennow K, Fedynyshyn JP, Allauzen S. Aβ40 oligomers identified as a potential biomarker for the diagnosis of Alzheimer's disease. PLoS One 2010; 5:e15725. [PMID: 21209907 PMCID: PMC3012719 DOI: 10.1371/journal.pone.0015725] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Accepted: 11/21/2010] [Indexed: 12/02/2022] Open
Abstract
Alzheimer's Disease (AD) is the most prevalent form of dementia worldwide, yet the development of therapeutics has been hampered by the absence of suitable biomarkers to diagnose the disease in its early stages prior to the formation of amyloid plaques and the occurrence of irreversible neuronal damage. Since oligomeric Aβ species have been implicated in the pathophysiology of AD, we reasoned that they may correlate with the onset of disease. As such, we have developed a novel misfolded protein assay for the detection of soluble oligomers composed of Aβ x-40 and x-42 peptide (hereafter Aβ40 and Aβ42) from cerebrospinal fluid (CSF). Preliminary validation of this assay with 36 clinical samples demonstrated the presence of aggregated Aβ40 in the CSF of AD patients. Together with measurements of total Aβ42, diagnostic sensitivity and specificity greater than 95% and 90%, respectively, were achieved. Although larger sample populations will be needed to confirm this diagnostic sensitivity, our studies demonstrate a sensitive method of detecting circulating Aβ40 oligomers from AD CSF and suggest that these oligomers could be a powerful new biomarker for the early detection of AD.
Collapse
Affiliation(s)
- Carol Man Gao
- Research and Development, Novartis Vaccines and Diagnostics, Emeryville, California, United States of America
| | - Alice Y. Yam
- Research and Development, Novartis Vaccines and Diagnostics, Emeryville, California, United States of America
| | - Xuemei Wang
- Research and Development, Novartis Vaccines and Diagnostics, Emeryville, California, United States of America
| | - Erika Magdangal
- Research and Development, Novartis Vaccines and Diagnostics, Emeryville, California, United States of America
| | - Cleo Salisbury
- Research and Development, Novartis Vaccines and Diagnostics, Emeryville, California, United States of America
| | - David Peretz
- Research and Development, Novartis Vaccines and Diagnostics, Emeryville, California, United States of America
| | - Ronald N. Zuckermann
- Research and Development, Novartis Vaccines and Diagnostics, Emeryville, California, United States of America
| | - Michael D. Connolly
- Research and Development, Novartis Vaccines and Diagnostics, Emeryville, California, United States of America
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Lennart Minthon
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Department of Neuroscience and Physiology, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Department of Neuroscience and Physiology, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Joseph P. Fedynyshyn
- Research and Development, Novartis Vaccines and Diagnostics, Emeryville, California, United States of America
- * E-mail:
| | - Sophie Allauzen
- Research and Development, Novartis Vaccines and Diagnostics, Emeryville, California, United States of America
| |
Collapse
|
43
|
Yoo B, Shin SBY, Huang ML, Kirshenbaum K. Peptoid macrocycles: making the rounds with peptidomimetic oligomers. Chemistry 2010; 16:5528-37. [PMID: 20414912 DOI: 10.1002/chem.200903549] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Macrocyclic constraints are often employed to rigidify the conformation of flexible oligomeric systems. This approach has recently been used to organize the structure of peptoid oligomers, which are peptidomimetics composed of chemically diverse N-substituted glycine monomer units. In this review, we describe advances in the synthesis and characterization of cyclic peptoids. We evaluate how the installation of covalent constraints between the oligomer termini or side chains has been effective in defining peptoid conformations. We also discuss the potential applications for this promising family of macrocyclic peptidomimetics.
Collapse
Affiliation(s)
- Barney Yoo
- Department of Chemistry, New York University, 100 Washington Square East, New York, NY 10003-6688, USA
| | | | | | | |
Collapse
|
44
|
Culf AS, Ouellette RJ. Solid-phase synthesis of N-substituted glycine oligomers (alpha-peptoids) and derivatives. Molecules 2010; 15:5282-335. [PMID: 20714299 PMCID: PMC6257730 DOI: 10.3390/molecules15085282] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Revised: 07/14/2010] [Accepted: 08/02/2010] [Indexed: 12/02/2022] Open
Abstract
Peptoids (N-substituted polyglycines and extended peptoids with variant backbone amino-acid monomer units) are oligomeric synthetic polymers that are becoming a valuable molecular tool in the biosciences. Of particular interest are their applications to the exploration of peptoid secondary structures and drug design. Major advantages of peptoids as research and pharmaceutical tools include the ease and economy of synthesis, highly variable backbone and side-chain chemistry possibilities. At the same time, peptoids have been demonstrated as highly active in biological systems while resistant to proteolytic decay. This review with 227 references considers the solid-phase synthetic aspects of peptoid preparation and utilization up to 2010 from the instigation, by R. N. Zuckermann et al., of peptoid chemistry in 1992.
Collapse
Affiliation(s)
- Adrian S Culf
- Atlantic Cancer Research Institute, Moncton, NB, Canada.
| | | |
Collapse
|
45
|
Abstract
Peptidomimetic oligomers and foldamers have received considerable attention for over a decade, with beta-peptides and the so-called peptoids (N-alkylglycine oligomers) representing prominent examples of such architectures. Lately, hybrid or mixed backbones consisting of both alpha- and beta-amino acids (alpha/beta-peptides) have been investigated in some detail as well. The present Minireview is a survey of the literature concerning hybrid structures of alpha-amino acids and peptoids, including beta-peptoids (N-alkyl-beta-alanine oligomers), and is intended to give an overview of this area of research within the field of peptidomimetic science.
Collapse
Affiliation(s)
- Christian A Olsen
- Department of Medicinal Chemistry, University of Copenhagen, Universitetsparken 2, Copenhagen, 2100 Denmark.
| |
Collapse
|
46
|
Havrylov S, Redowicz MJ, Buchman VL. Emerging roles of Ruk/CIN85 in vesicle-mediated transport, adhesion, migration and malignancy. Traffic 2010; 11:721-31. [PMID: 20331533 DOI: 10.1111/j.1600-0854.2010.01061.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Ruk/CIN85 is an adaptor protein. Similar to many other proteins of this type, Ruk/CIN85 is known to take part in multiple cellular processes including signal transduction, vesicle-mediated transport, cytoskeleton remodelling, programmed cell death and viral infection. Recent studies have also revealed the potential importance of Ruk/CIN85 in cancer cell invasiveness. In this review we summarize the various roles of this protein as well as the potential contribution of Ruk/CIN85 to malignancy and the invasiveness of cancer cells. In the last section of the paper we also speculate on the utility of Ruk/CIN85 as a target for novel anti-cancer therapies.
Collapse
Affiliation(s)
- Serhiy Havrylov
- Nencki Institute of Experimental Biology, Pasteura 3 Street, 02-093 Warsaw, Poland
| | | | | |
Collapse
|
47
|
Gorske BC, Stringer JR, Bastian BL, Fowler SA, Blackwell HE. New strategies for the design of folded peptoids revealed by a survey of noncovalent interactions in model systems. J Am Chem Soc 2010; 131:16555-67. [PMID: 19860427 DOI: 10.1021/ja907184g] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Controlling the equilibria between backbone cis- and trans-amides in peptoids, or N-substituted glycine oligomers, constitutes a significant challenge in the construction of discretely folded peptoid structures. Through the analysis of a set of monomeric peptoid model systems, we have developed new and general strategies for controlling peptoid conformation that utilize local noncovalent interactions to regulate backbone amide rotameric equilibria, including n-->pi*, steric, and hydrogen bonding interactions. The chemical functionalities required to implement these strategies are typically confined to the peptoid side chains, preserve chirality at the side chain N-alpha-carbon known to engender peptoid structure, and are fully compatible with standard peptoid synthesis techniques. Our examinations of peptoid model systems have also elucidated how solvents affect various side chain-backbone interactions, revealing fundamental aspects of these noncovalent interactions in peptoids that were largely uncharacterized previously. As validation of our monomeric model systems, we extended the scope of this study to include peptoid oligomers and have now demonstrated the importance of local steric and n-->pi* interactions in dictating the structures of larger, folded peptoids. This new, modular design strategy has guided the construction of peptoids containing 1-naphthylethyl side chains, which we show can be utilized to effectively eliminate trans-amide rotamers from the peptoid backbone, yielding the most conformationally homogeneous class of peptoid structures yet reported in terms of amide rotamerism. Overall, this research has afforded a valuable and expansive set of design tools for the construction of both discretely folded peptoids and structurally biased peptoid libraries and should shape our understanding of peptoid folding.
Collapse
Affiliation(s)
- Benjamin C Gorske
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, Wisconsin 53706-1322, USA
| | | | | | | | | |
Collapse
|
48
|
Hoffmann S, Funke SA, Wiesehan K, Moedder S, Glück JM, Feuerstein S, Gerdts M, Mötter J, Willbold D. Competitively selected protein ligands pay their increase in specificity by a decrease in affinity. MOLECULAR BIOSYSTEMS 2009; 6:126-33. [PMID: 20024074 DOI: 10.1039/b910945e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Protein-ligand interactions characterise and govern the current state and fate of a living cell. The specificity of proteins is mainly determined by the relative affinities to each potential ligand. To investigate the consequences and potentials of ligands with increased specificity in comparison with ligands optimised solely for affinity, it was necessary to identify ligands that are optimised towards specificity instead of a barely optimised affinity to a given target. In the presented example, a modified phage display screening procedure yielded specific ligands for the LckSH3 domain. We found that increased specificity of one of the hereby obtained ligands for LckSH3 is achieved at the cost of a slightly reduced affinity to LckSH3 and a drastically reduced affinity to other SH3 domains. A surface plasmon resonance experiment simulating in vivo-like realistic competitive binding conditions exerted enhanced binding behaviour of the specific ligand under these binding conditions. The experimental data, together with a mathematical model describing the complex experimental situation, and theoretical considerations lead to the conclusion that increased specificity is achieved at the cost of reduced affinity, but after all, it pays if the ligand is applied under realistic, i.e. competitive, conditions.
Collapse
Affiliation(s)
- Silke Hoffmann
- ISB-3, Structural Biochemistry, Forschungszentrum Jülich, 52425 Jülich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Fowler SA, Blackwell HE. Structure-function relationships in peptoids: recent advances toward deciphering the structural requirements for biological function. Org Biomol Chem 2009; 7:1508-24. [PMID: 19343235 DOI: 10.1039/b817980h] [Citation(s) in RCA: 233] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Oligomers of N-substituted glycine, or peptoids, are versatile tools to probe biological processes and hold promise as therapeutic agents. An underlying theme in the majority of recent peptoid research is the connection between peptoid function and peptoid structure. For certain applications, well-folded peptoids are essential for activity, while unstructured peptoids appear to suffice, or even are superior, for other applications. Currently, these structure-function connections are largely made after the design, synthesis, and characterization process. However, as guidelines for peptoid folding are elucidated and the known biological activities are expanded, we anticipate these connections will provide a pathway toward the de novo design of functional peptoids. In this perspective, we review several of the peptoid structure-function relationships that have been delineated over the past five years.
Collapse
Affiliation(s)
- Sarah A Fowler
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, WI 53706-1322, USA
| | | |
Collapse
|
50
|
Abstract
The tremendous challenge of inhibiting therapeutically important protein-protein interactions has created the opportunity to extend traditional medicinal chemistry to a new class of targets and to explore nontraditional strategies. Here we review a widely studied system, the interaction between tumor suppressor p53 and its natural antagonist MDM2, for which both traditional and nontraditional approaches have been reported. This system has been a testing ground for novel proteomimetic scaffold-based strategies, i.e., for attempts to mimic the recognition surface displayed by a folded protein with unnatural oligomers. Retroinverso peptides, peptoids, terphenyls, beta-hairpins, p-oligobenzamides, beta-peptides, and miniproteins have all been explored as inhibitors of the p53/MDM2 interaction, and we focus on these oligomer-based efforts. Traditional approaches have been successful as well, and we briefly review small molecule inhibitors along with other strategies for reactivation of the p53 pathway, for comparison with oligomer- based approaches. We close with comments on an emerging dichotomy among protein-protein interaction targets.
Collapse
Affiliation(s)
- Justin K Murray
- Department of Chemistry, University of Wisconsin, 1101 University Avenue, Madison, WI 53706, USA
| | | |
Collapse
|