1
|
Vornweg JR, Maier TM, Jacob CR. The density-based many-body expansion for poly-peptides and proteins. Phys Chem Chem Phys 2025; 27:8719-8730. [PMID: 40235457 DOI: 10.1039/d5cp00727e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Fragmentation schemes enable the efficient quantum-chemical treatment of large biomolecular systems, and provide an ideal starting point for the development of accurate machine-learning potentials for proteins. Here, we present a fragment-based method that only uses calculations for single-amino acids and their dimers, and is able to reduce the fragmentation error in total energies to ca. 1 kJ mol-1 per amino acid for polypeptides and proteins across different structural motifs. This is achieved by combining a two-body extension of the molecular fractionation with conjugate caps (MFCC) scheme with the density-based many-body expansion (db-MBE), thus extending the applicability of the db-MBE from molecular clusters to polypeptides and proteins.
Collapse
Affiliation(s)
- Johannes R Vornweg
- Technische Universität Braunschweig, Institute of Physical and Theoretical Chemistry, Gaußstraße 17, 38106 Braunschweig, Germany.
| | - Toni M Maier
- Technische Universität Braunschweig, Institute of Physical and Theoretical Chemistry, Gaußstraße 17, 38106 Braunschweig, Germany.
| | - Christoph R Jacob
- Technische Universität Braunschweig, Institute of Physical and Theoretical Chemistry, Gaußstraße 17, 38106 Braunschweig, Germany.
| |
Collapse
|
2
|
Kang K, do Espirito Santo É, Diaz CJ, Mayfield S, Molino JVD. Engineering microalgal cell wall-anchored proteins using GP1 PPSPX motifs and releasing with intein-mediated fusion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.634604. [PMID: 39896471 PMCID: PMC11785195 DOI: 10.1101/2025.01.23.634604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Harnessing and controlling the localization of recombinant proteins is critical for advancing applications in synthetic biology, industrial biotechnology, and drug delivery. This study explores protein anchoring and controlled release in Chlamydomonas reinhardtii, providing innovative tools for these fields. Using truncated variants of the GP1 glycoprotein fused to the plastic-degrading enzyme PHL7, we identified the PPSPX motif as essential for anchoring proteins to the cell wall. Constructs with increased PPSPX content exhibited reduced secretion but improved anchoring, pinpointing the potential anchor-signal sites of GP1 and highlighting the distinct roles of these motifs in protein localization. Building on the anchoring capabilities established with these glycomodules, we also demonstrated a controlled release system using a pH-sensitive intein derived from RecA from Mycobacterium tuberculosis. This intein efficiently cleaved and released PHL7 and mCherry that was fused to GP1 under acidic conditions, enabling precise temporal and environmental control. At pH 5.5, fluorescence kinetics demonstrated significant mCherry release from the pJPW4mCherry construct within 4 hours. In contrast, release was minimal under pH 8.0 conditions and negligible for the pJPW2mCherry (W2) control, irrespective of the pH. Additionally, bands on the Western blot at the expected size of mCherry also showed its efficient release from the mCherry::intein::GP1 fusion protein at pH 5.5. Conversely, at pH 8.0, no bands were detected. This anchor-release approach offers significant potential for drug delivery, biocatalysis, and environmental monitoring applications. By integrating glycomodules and pH-sensitive inteins, this study establishes a versatile framework for optimizing protein localization and release in C. reinhardtii, with broad implications for proteomics, biofilm engineering, and scalable therapeutic delivery systems.
Collapse
Affiliation(s)
- Kalisa Kang
- Department of Molecular Biology, School of Biological Sciences, University of California San Diego, La Jolla, CA, United States of America
| | - Évellin do Espirito Santo
- Department of Molecular Biology, School of Biological Sciences, University of California San Diego, La Jolla, CA, United States of America
- Department of Biochemical and Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Crisandra Jade Diaz
- Department of Molecular Biology, School of Biological Sciences, University of California San Diego, La Jolla, CA, United States of America
| | - Stephen Mayfield
- Department of Molecular Biology, School of Biological Sciences, University of California San Diego, La Jolla, CA, United States of America
- Algenesis Inc., 1238 Sea Village Dr., Cardiff, CA, United States of America
| | - João Vitor Dutra Molino
- Department of Molecular Biology, School of Biological Sciences, University of California San Diego, La Jolla, CA, United States of America
| |
Collapse
|
3
|
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
|
4
|
Behar AE, Maayan G. A cocktail of Cu 2+- and Zn 2+-peptoid-based chelators can stop ROS formation for Alzheimer's disease therapy. Chem Sci 2024:d4sc04313h. [PMID: 39464602 PMCID: PMC11503657 DOI: 10.1039/d4sc04313h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024] Open
Abstract
The formation of reactive oxygen species (ROS) in the brain is a major cause of neuropathologic degradation associated with Alzheimer's Disease (AD). It has been suggested that the copper (Cu)-amyloid-β (Aβ) peptide complex can lead to ROS formation in the brain. An external chelator for Cu that can extract Cu from the CuAβ complex should inhibit the formation of ROS, making Cu chelation an excellent therapeutic approach for AD. Such a chelator should possess high selectivity for Cu over zinc (Zn), which is also present within the synaptic cleft. However, such selectivity is generally hard to achieve in one molecule due to the similarities in the binding preferences of these two metal ions. As an alternative to monotherapy (where Cu extraction is performed using a single chelator), herein we describe a variation of combination therapy - a novel cocktail approach, which is based on the co-administration of two structurally different peptidomimetic chelators, aiming to target both Cu2+ and Zn2+ ions simultaneously but independently from each other. Based on rigorous spectroscopic experiments, we demonstrate that our peptidomimetic cocktail allows, for the first time, the complete and immediate inhibition of ROS production by the CuAβ complex in the presence of Zn2+. In addition, we further demonstrate the high stability of the cocktail under simulated physiological conditions and its resistance to proteolytic degradation by trypsin and report the water/n-octanol partition coefficient, initially assessing the blood-brain barrier (BBB) permeability potential of the chelators.
Collapse
Affiliation(s)
- Anastasia E Behar
- Schulich Faculty of Chemistry, Technion - Israel Institute of Technology, Technion City 3200008 Haifa Israel
| | - Galia Maayan
- Schulich Faculty of Chemistry, Technion - Israel Institute of Technology, Technion City 3200008 Haifa Israel
| |
Collapse
|
5
|
Tennett JC, Epstein SR, Sawyer N. Enhanced EphB2-Specific Peptide Inhibitors through Stabilization of Polyproline II Helical Structure. ACS Chem Biol 2024; 19:1214-1221. [PMID: 38739742 DOI: 10.1021/acschembio.3c00791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Ephrin (Eph) receptors are the largest family of receptor tyrosine kinases. Interactions between Eph receptors and their membrane-bound ephrin protein ligands are associated with many developmental processes as well as various cancers and neurodegenerative diseases. With significant crosstalk between different Eph receptors and ephrin ligands, there is an urgent need for high-affinity ligands that bind specifically to individual Eph receptors to interrogate and modulate their functions. Here, we describe the rational development of potent EphB2 receptor inhibitors derived from the EphB2 receptor-specific SNEW peptide. To improve inhibitory potency, we evaluated 20+ cross-linkers with the goal of spanning and stabilizing a single polyproline II helical turn observed when SNEW binds to the EphB2 receptor. Of the cross-linkers evaluated, an 11-atom cross-linker, composed of a rigid 2,7-dimethylnaphthyl moiety between two cysteine residues, was found to yield the most potent inhibitor. Analysis of the cyclized region of this peptide by NMR and molecular dynamics simulations suggests that cross-linking stabilizes the receptor-bound polyproline II helix structure observed in the receptor-peptide complex. Cross-linked SNEW variants retained binding specificity for EphB2 and showed cross-linker-dependent resistance to trypsin proteolysis. Beyond the discovery of more potent EphB2 receptor inhibitors, these studies illustrate a novel cyclization approach with potential to stabilize polyproline II helical structure in various peptides for specific targeting of the myriad protein-protein interactions (PPIs) mediated by polyproline II helices.
Collapse
Affiliation(s)
- Jessica C Tennett
- Department of Chemistry, Fordham University, 441 E. Fordham Rd., Bronx, New York 10458, United States
| | - Sophie R Epstein
- Department of Chemistry, Fordham University, 441 E. Fordham Rd., Bronx, New York 10458, United States
| | - Nicholas Sawyer
- Department of Chemistry, Fordham University, 441 E. Fordham Rd., Bronx, New York 10458, United States
| |
Collapse
|
6
|
Jos S, Poulose R, Kambaru A, Gogoi H, Dalavaikodihalli Nanjaiah N, Padmanabhan B, Mehta B, Padavattan S. Tau-S214 Phosphorylation Inhibits Fyn Kinase Interaction and Increases the Decay Time of NMDAR-mediated Current. J Mol Biol 2024; 436:168445. [PMID: 38218365 DOI: 10.1016/j.jmb.2024.168445] [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: 08/17/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/15/2024]
Abstract
Fyn kinase SH3 domain interaction with PXXP motif in the Tau protein is implicated in AD pathology and is central to NMDAR function. Among seven PXXP motifs localized in proline-rich domain of Tau protein, tandem 5th and 6th PXXP motifs are critical to Fyn-SH3 domain interaction. Here, we report the crystal structure of Fyn-SH3 -Tau (207-221) peptide consisting of 5th and 6th PXXP motif complex to 1.01 Å resolution. Among five AD-specific phosphorylation sites encompassing the 5th and 6th PXXP motifs, only S214 residue showed interaction with SH3 domain. Biophysical studies showed that Tau (207-221) with S214-phosphorylation (pS214) inhibits its interaction with Fyn-SH3 domain. The individual administration of Tau (207-221) with/without pS214 peptides to a single neuron increased the decay time of evoked NMDA current response. Recordings of spontaneous NMDA EPSCs at +40 mV indicate an increase in frequency and amplitude of events for the Tau (207-221) peptide. Conversely, the Tau (207-221) with pS214 peptide exhibited a noteworthy amplitude increase alongside a prolonged decay time. These outcomes underscore the distinctive modalities of action associated with each peptide in the study. Overall, this study provides insights into how Tau (207-221) with/without pS214 affects the molecular framework of NMDAR signaling, indicating its involvement in Tau-related pathogenesis.
Collapse
Affiliation(s)
- Sneha Jos
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Roshni Poulose
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Archanalakshmi Kambaru
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Hemanga Gogoi
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | | | - Balasundaram Padmanabhan
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Bhupesh Mehta
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India.
| | - Sivaraman Padavattan
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India.
| |
Collapse
|
7
|
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
|
8
|
Behar AE, Maayan G. A Peptoid-Chelator Selective to Cu 2+ That Can Extract Copper from Metallothionein-2 and Lead to the Production of ROS. Antioxidants (Basel) 2023; 12:2031. [PMID: 38136151 PMCID: PMC10741037 DOI: 10.3390/antiox12122031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 12/24/2023] Open
Abstract
Copper is an essential metal ion that is involved in critical cellular processes, but which can also exhibit toxic effects through its ability to catalyze reactive oxygen species (ROS) formation. Dysregulation of copper homeostasis has been implicated in the progression of several diseases, including cancer. A novel therapeutic approach, extensively studied in recent years, is to capitalize on the increased copper uptake and dependency exhibited by cancer cells and to promote copper-associated ROS production within the tumor microenvironment, leading to the apoptosis of cancer cells. Such an effect can be achieved by selectively chelating copper from copper-bearing metalloproteins in cancer cells, thereby forming a copper-chelator complex that produces ROS and, through this, induces oxidative stress and initiates apoptosis. Herein, we describe a peptoid chelator, TB, that is highly suitable to carry this task. Peptoids are N-substituted glycine oligomers that can be efficiently synthesized on a solid support and are also biocompatible; thus, they are considered promising drug candidates. We show, by rigorous spectroscopic techniques, that TB is not only selective for Cu(II) ions, but can also effectively extract copper from metallothionein-2, and the formed complex CuTB can promote ROS production. Our findings present a promising first example for the future development of peptoid-based chelators for applications in anti-cancer chelation therapy, highlighting the potential for the prospect of peptoid chelators as therapeutics.
Collapse
Affiliation(s)
| | - Galia Maayan
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology Technion City, Haifa 3200008, Israel
| |
Collapse
|
9
|
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
|
10
|
Mehrabipour M, Jasemi NSK, Dvorsky R, Ahmadian MR. A Systematic Compilation of Human SH3 Domains: A Versatile Superfamily in Cellular Signaling. Cells 2023; 12:2054. [PMID: 37626864 PMCID: PMC10453029 DOI: 10.3390/cells12162054] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
SRC homology 3 (SH3) domains are fundamental modules that enable the assembly of protein complexes through physical interactions with a pool of proline-rich/noncanonical motifs from partner proteins. They are widely studied modular building blocks across all five kingdoms of life and viruses, mediating various biological processes. The SH3 domains are also implicated in the development of human diseases, such as cancer, leukemia, osteoporosis, Alzheimer's disease, and various infections. A database search of the human proteome reveals the existence of 298 SH3 domains in 221 SH3 domain-containing proteins (SH3DCPs), ranging from 13 to 720 kilodaltons. A phylogenetic analysis of human SH3DCPs based on their multi-domain architecture seems to be the most practical way to classify them functionally, with regard to various physiological pathways. This review further summarizes the achievements made in the classification of SH3 domain functions, their binding specificity, and their significance for various diseases when exploiting SH3 protein modular interactions as drug targets.
Collapse
Affiliation(s)
- Mehrnaz Mehrabipour
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (M.M.); (N.S.K.J.)
| | - Neda S. Kazemein Jasemi
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (M.M.); (N.S.K.J.)
| | - Radovan Dvorsky
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (M.M.); (N.S.K.J.)
- Center for Interdisciplinary Biosciences, P. J. Šafárik University, 040 01 Košice, Slovakia
| | - Mohammad R. Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (M.M.); (N.S.K.J.)
| |
Collapse
|
11
|
Behar AE, Maayan G. The First Cu(I)-Peptoid Complex: Enabling Metal Ion Stability and Selectivity via Backbone Helicity. Chemistry 2023; 29:e202301118. [PMID: 37221918 DOI: 10.1002/chem.202301118] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 05/25/2023]
Abstract
Stabilization of Cu(I) is ubiquitous within native copper proteins. Understanding how to stabilize Cu(I) within synthetic biomimetic systems is therefore desired towards biological applications. Peptoids are an important class of peptodomimetics, that can bind metal ions and stabilize them in their high oxidation state. Thus, to date, they were not used for Cu(I) binding. Here we show how the helical peptoid hexamer, having two 2,2'-bipyridine (Bipy) groups that face the same side of the helix, forms the intramolecular air stable Cu(I) complex. Further study of the binding site by rigorous spectroscopic techniques suggests that Cu(I) is tetracoordinated, binding to only three N atoms from the Bipy ligands and to the N-terminus of the peptoid's backbone. A set of control peptoids and experiments indicates that the Cu(I) stability and selectivity are dictated by the intramolecular binding, forced by the helicity of the peptoid, which can be defined as the second coordination sphere of the metal center.
Collapse
Affiliation(s)
- Anastasia E Behar
- Schulich Faculty of Chemistry, Technion - Israel Institute of Technology, Technion City, 3200008 Haifa, Israel
| | - Galia Maayan
- Schulich Faculty of Chemistry, Technion - Israel Institute of Technology, Technion City, 3200008 Haifa, Israel
| |
Collapse
|
12
|
Vornweg JR, Wolter M, Jacob CR. A simple and consistent quantum-chemical fragmentation scheme for proteins that includes two-body contributions. J Comput Chem 2023; 44:1634-1644. [PMID: 37171574 DOI: 10.1002/jcc.27114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 05/13/2023]
Abstract
The Molecular Fractionation with Conjugate Caps (MFCC) method is a popular fragmentation method for the quantum-chemical treatment of proteins. However, it does not account for interactions between the amino acid fragments, such as intramolecular hydrogen bonding. Here, we present a combination of the MFCC fragmentation scheme with a second-order many-body expansion (MBE) that consistently accounts for all fragment-fragment, fragment-cap, and cap-cap interactions, while retaining the overall simplicity of the MFCC scheme with its chemically meaningful fragments. We show that with the resulting MFCC-MBE(2) scheme, the errors in the total energies of selected polypeptides and proteins can be reduced by up to one order of magnitude and relative energies of different protein conformers can be predicted accurately.
Collapse
Affiliation(s)
- Johannes R Vornweg
- Institute of Physical and Theoretical Chemistry, Technische Universität Braunschweig, Braunschweig, Germany
| | - Mario Wolter
- Institute of Physical and Theoretical Chemistry, Technische Universität Braunschweig, Braunschweig, Germany
| | - Christoph R Jacob
- Institute of Physical and Theoretical Chemistry, Technische Universität Braunschweig, Braunschweig, Germany
| |
Collapse
|
13
|
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
|
14
|
Gorges J, Bädorf B, Hansen A, Grimme S. Efficient Computation of the Interaction Energies of Very Large Non-covalently Bound Complexes. Synlett 2022. [DOI: 10.1055/s-0042-1753141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
AbstractWe present a new benchmark set consisting of 16 large non-covalently bound systems (LNCI16) ranging from 380 up to 1988 atoms and featuring diverse interaction motives. Gas-phase interaction energies are calculated with various composite DFT, semi-empirical quantum mechanical (SQM), and force field (FF) methods and are evaluated using accurate DFT reference values. Of the employed QM methods, PBEh-3c proves to be the most robust for large systems with a relative mean absolute deviation (relMAD) of 8.5% with respect to the reference interaction energies. r2SCAN-3c yields an even smaller relMAD, at least for the subset of complexes for which the calculation could be converged, but is less robust for systems with smaller HOMO–LUMO gaps. The inclusion of Fock-exchange is therefore important for the description of very large non-covalent interaction (NCI) complexes in the gas phase. GFN2-xTB was found to be the best performer of the SQM methods with an excellent result of only 11.1% deviation. From the assessed force fields, GFN-FF and GAFF achieve the best accuracy. Considering their low computational costs, both can be recommended for routine calculations of very large NCI complexes, with GFN-FF being clearly superior in terms of general applicability. Hence, GFN-FF may be routinely applied in supramolecular synthesis planning.1 Introduction2 The LNCI16 Benchmark Set3 Computational Details4 Generation of Reference Values5 Results and Discussion6 Conclusions
Collapse
|
15
|
Helical Foldamers and Stapled Peptides as New Modalities in Drug Discovery: Modulators of Protein-Protein Interactions. Processes (Basel) 2022. [DOI: 10.3390/pr10050924] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
A “foldamer” is an artificial oligomeric molecule with a regular secondary or tertiary structure consisting of various building blocks. A “stapled peptide” is a peptide with stabilized secondary structures, in particular, helical structures by intramolecular covalent side-chain cross-linking. Helical foldamers and stapled peptides are potential drug candidates that can target protein-protein interactions because they enable multipoint molecular recognition, which is difficult to achieve with low-molecular-weight compounds. This mini-review describes a variety of peptide-based foldamers and stapled peptides with a view to their applications in drug discovery, including our recent progress.
Collapse
|
16
|
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
|
17
|
Garsi JB, Aguiar PM, Hanessian S. Design of Pseudodiproline Dimers as Mimetics of Pro-Pro Units: Stereocontrolled Synthesis, Configurational Relevance, and Structural Properties. J Org Chem 2021; 86:16834-16847. [PMID: 34749500 DOI: 10.1021/acs.joc.1c02061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Stereocontrolled methods are described for the synthesis of hitherto unreported pseudodiproline dimers in which a cyclopentane carboxylic acid is linked to a pyrrolidine residue by a stereochemically defined hydroxymethylene tether. These proline-cyclopentane (Pro-Cyp) dimers have interesting structural characteristics as seen in their X-ray crystal structures as well as their nuclear magnetic resonance (NMR) spectra in CDCl3. They can be considered to be novel Pro-Pro mimetics, which can be used to replace natural diproline sequences with potential applications in medicinal chemistry. They also represent a new concept in the peptidomimetic design of chimeric proline-based amino acids as carbocyclic hydroxyethylene isosteres of inhibitor molecules, in which the stereodefined bridging hydroxyl group can simulate a tetrahedral intermediate in an enzyme complex.
Collapse
Affiliation(s)
- Jean-Baptiste Garsi
- Department of Chemistry, Université de Montréal, 1375 Ave. Thérèse-Lavoie-Roux, Montréal, H2V 0B3 QC, Canada
| | - Pedro M Aguiar
- Department of Chemistry, Université de Montréal, 1375 Ave. Thérèse-Lavoie-Roux, Montréal, H2V 0B3 QC, Canada
| | - Stephen Hanessian
- Department of Chemistry, Université de Montréal, 1375 Ave. Thérèse-Lavoie-Roux, Montréal, H2V 0B3 QC, Canada
| |
Collapse
|
18
|
Behar AE, Sabater L, Baskin M, Hureau C, Maayan G. A Water-Soluble Peptoid Chelator that Can Remove Cu 2+ from Amyloid-β Peptides and Stop the Formation of Reactive Oxygen Species Associated with Alzheimer's Disease. Angew Chem Int Ed Engl 2021; 60:24588-24597. [PMID: 34510664 DOI: 10.1002/anie.202109758] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/01/2021] [Indexed: 12/25/2022]
Abstract
Cu bound to amyloid-β (Aβ) peptides can act as a catalyst for the formation of reactive oxygen species (ROS), leading to neuropathologic degradation associated with Alzheimer's disease (AD). An excellent therapeutic approach is to use a chelator that can selectively remove Cu from Cu-Aβ. This chelator should compete with Zn2+ ions (Zn) that are present in the synaptic cleft while forming a nontoxic Cu complex. Herein we describe P3, a water-soluble peptidomimetic chelator that selectively removes Cu2+ from Cu-Aβ in the presence of Zn and prevent the formation of ROS even in a reductive environment. We demonstrate, based on extensive spectroscopic analysis, that although P3 extracts Zn from Cu,Zn-Aβ faster than it removes Cu, the formed Zn complexes are kinetic products that further dissociate, while CuP3 is formed as an exclusive stable thermodynamic product. Our unique findings, combined with the bioavailability of peptoids, make P3 an excellent drug candidate in the context of AD.
Collapse
Affiliation(s)
- Anastasia E Behar
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, Technion City, 3200008, Haifa, Israel
| | - Laurent Sabater
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, 31077, Toulouse, France.,Université de Toulouse, 31077, Toulouse, France
| | - Maria Baskin
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, Technion City, 3200008, Haifa, Israel
| | - Christelle Hureau
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, 31077, Toulouse, France.,Université de Toulouse, 31077, Toulouse, France
| | - Galia Maayan
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, Technion City, 3200008, Haifa, Israel
| |
Collapse
|
19
|
Behar AE, Sabater L, Baskin M, Hureau C, Maayan G. A Water‐Soluble Peptoid Chelator that Can Remove Cu
2+
from Amyloid‐β Peptides and Stop the Formation of Reactive Oxygen Species Associated with Alzheimer's Disease. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202109758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Anastasia E. Behar
- Schulich Faculty of Chemistry Technion—Israel Institute of Technology Technion City 3200008 Haifa Israel
| | - Laurent Sabater
- CNRS LCC (Laboratoire de Chimie de Coordination) 205 route de Narbonne 31077 Toulouse France
- Université de Toulouse 31077 Toulouse France
| | - Maria Baskin
- Schulich Faculty of Chemistry Technion—Israel Institute of Technology Technion City 3200008 Haifa Israel
| | - Christelle Hureau
- CNRS LCC (Laboratoire de Chimie de Coordination) 205 route de Narbonne 31077 Toulouse France
- Université de Toulouse 31077 Toulouse France
| | - Galia Maayan
- Schulich Faculty of Chemistry Technion—Israel Institute of Technology Technion City 3200008 Haifa Israel
| |
Collapse
|
20
|
Wu J, Wang X, Xiao L, Wang F, Zhang Y, Li X. Synthetic Protein Scaffolds for Improving R-(-)-Linalool Production in Escherichia coli. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:5663-5670. [PMID: 33983023 DOI: 10.1021/acs.jafc.1c01101] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
R-(-)-Linalool is widely used in the pharmaceutical, agrochemical, and fragrance industries; however, its applications are limited owing to low yield and high cost of production. To improve the production efficiency of R-(-)-linalool in Escherichia coli, three enzymes [E. coli-derived isopentenyl diphosphate isomerase, Abies grandis-derived geranyl diphosphate synthase, and Streptomyces clavuligerus-derived (3R)-linalool synthases] were physically colocalized to synthetic complexes using synthetic protein scaffolds of GTPase-binding domain, Src homology 3, and PSD95/DlgA/Zo-1. R-(-)-Linalool was produced at the highest concentration in the strain IGL114 containing a scaffold ratio of 1:1:4. By further optimizing the inducer, temperature, and glycerol concentration, the production titer of R-(-)-linalool in the shake flask was increased by approximately 10 times compared with that of the scaffold-free control and was 2.78 times the previously reported yield. The production in the fermenter was about 1.5 times the previous highest production. In general, the final strain accumulated 277.8 and 1523.2 mg/L R-(-)-linalool under the conditions of shake-flask and fed-batch fermentation, respectively. This study provides a foundation for the assembly of bacterial intracellular protein scaffolds.
Collapse
Affiliation(s)
- Jing Wu
- Jiangsu Provincial Key Laboratory for the Chemistry and Utilization of Agro-Forest Biomass, Nanjing Forestry University, Nanjing 210037, China
- Jiangsu Key Laboratory of Biomass-Based Green Fuels and Chemicals, Nanjing Forestry University, Nanjing 210037, China
- Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
- International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing 210037, China
| | - Xun Wang
- Jiangsu Provincial Key Laboratory for the Chemistry and Utilization of Agro-Forest Biomass, Nanjing Forestry University, Nanjing 210037, China
- Jiangsu Key Laboratory of Biomass-Based Green Fuels and Chemicals, Nanjing Forestry University, Nanjing 210037, China
- Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
- International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing 210037, China
| | - Longjie Xiao
- Jiangsu Provincial Key Laboratory for the Chemistry and Utilization of Agro-Forest Biomass, Nanjing Forestry University, Nanjing 210037, China
- Jiangsu Key Laboratory of Biomass-Based Green Fuels and Chemicals, Nanjing Forestry University, Nanjing 210037, China
- Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
- International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing 210037, China
| | - Fei Wang
- Jiangsu Provincial Key Laboratory for the Chemistry and Utilization of Agro-Forest Biomass, Nanjing Forestry University, Nanjing 210037, China
- Jiangsu Key Laboratory of Biomass-Based Green Fuels and Chemicals, Nanjing Forestry University, Nanjing 210037, China
- Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
- International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing 210037, China
| | - Yu Zhang
- Jiangsu Provincial Key Laboratory for the Chemistry and Utilization of Agro-Forest Biomass, Nanjing Forestry University, Nanjing 210037, China
- Jiangsu Key Laboratory of Biomass-Based Green Fuels and Chemicals, Nanjing Forestry University, Nanjing 210037, China
- Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
- International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing 210037, China
| | - Xun Li
- Jiangsu Provincial Key Laboratory for the Chemistry and Utilization of Agro-Forest Biomass, Nanjing Forestry University, Nanjing 210037, China
- Jiangsu Key Laboratory of Biomass-Based Green Fuels and Chemicals, Nanjing Forestry University, Nanjing 210037, China
- Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
- International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing 210037, China
| |
Collapse
|
21
|
Wolter M, von Looz M, Meyerhenke H, Jacob CR. Systematic Partitioning of Proteins for Quantum-Chemical Fragmentation Methods Using Graph Algorithms. J Chem Theory Comput 2021; 17:1355-1367. [PMID: 33591754 DOI: 10.1021/acs.jctc.0c01054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Quantum-chemical fragmentation methods offer an efficient approach for the treatment of large proteins, in particular if local target quantities such as protein-ligand interaction energies, enzymatic reaction energies, or spectroscopic properties of embedded chromophores are sought. However, the accuracy that is achievable for such local target quantities intricately depends on how the protein is partitioned into smaller fragments. While the commonly employed naı̈ve approach of using fragments with a fixed size is widely used, it can result in large and unpredictable errors when varying the fragment size. Here, we present a systematic partitioning scheme that aims at minimizing the fragmentation error of a local target quantity for a given maximum fragment size. To this end, we construct a weighted graph representation of the protein, in which the amino acids constitute the nodes. These nodes are connected by edges weighted with an estimate for the fragmentation error that is expected when cutting this edge. This allows us to employ graph partitioning algorithms provided by computer science to determine near-optimal partitions of the protein. We apply this scheme to a test set of six proteins representing various prototypical applications of quantum-chemical fragmentation methods using a simplified molecular fractionation with conjugate caps (MFCC) approach with hydrogen caps. We show that our graph-based scheme consistently improves upon the naı̈ve approach.
Collapse
Affiliation(s)
- Mario Wolter
- Institute of Physical and Theoretical Chemistry, Technische Universität Braunschweig, Gaußstrasse 17, 38106 Braunschweig, Germany
| | - Moritz von Looz
- Department of Computer Science, Humboldt-Universität zu Berlin, Unter den Linden 6, 10099 Berlin, Germany
| | - Henning Meyerhenke
- Department of Computer Science, Humboldt-Universität zu Berlin, Unter den Linden 6, 10099 Berlin, Germany
| | - Christoph R Jacob
- Institute of Physical and Theoretical Chemistry, Technische Universität Braunschweig, Gaußstrasse 17, 38106 Braunschweig, Germany
| |
Collapse
|
22
|
Romero JA, Nawrocka EK, Shchukina A, Blanco FJ, Diercks T, Kazimierczuk K. Non‐Stationary Complementary Non‐Uniform Sampling (NOSCO NUS) for Fast Acquisition of Serial 2D NMR Titration Data. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202009479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Javier A. Romero
- Centre of New Technologies University of Warsaw Banacha 2C 02-097 Warsaw Poland
| | - Ewa K. Nawrocka
- Centre of New Technologies University of Warsaw Banacha 2C 02-097 Warsaw Poland
- Faculty of Chemistry University of Warsaw Pasteura 1 02-093 Warsaw Poland
| | - Alexandra Shchukina
- Faculty of Chemistry Biological and Chemical Research Centre University of Warsaw Zwirki i Wigury 101 02-089 Warsaw Poland
| | - Francisco J. Blanco
- Structural and Chemical Biology Department Centro de Investigaciones Biológicas CIB-CSIC 28040 Madrid Spain
| | - Tammo Diercks
- CIC bioGUNE Parque Tecnológico de Bizkaia, Ed. 800 48160- Derio Spain
| | | |
Collapse
|
23
|
Romero JA, Nawrocka EK, Shchukina A, Blanco FJ, Diercks T, Kazimierczuk K. Non-Stationary Complementary Non-Uniform Sampling (NOSCO NUS) for Fast Acquisition of Serial 2D NMR Titration Data. Angew Chem Int Ed Engl 2020; 59:23496-23499. [PMID: 32852098 PMCID: PMC7756666 DOI: 10.1002/anie.202009479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Indexed: 11/13/2022]
Abstract
NMR spectroscopy offers unique benefits for ligand binding studies on isotopically labelled target proteins. These benefits include atomic resolution, direct distinction of binding sites and modes, a lowest detectable affinity limit, and function independent setup. Yet, retracing protein signal assignments from apo to holo states to derive exact dissociation constants and chemical shift perturbation amplitudes (for ligand docking and structure‐based optimization) requires lengthy titration series of 2D heteronuclear correlation spectra at variable ligand concentration that may exceed the protein's lifetime and available spectrometer time. We present a novel method to overcome this critical limitation, based on non‐stationary complementary non‐uniform sampling (NOSCO NUS) combined with a robust particle swarm optimization algorithm. We illustrate its potential in two challenging studies with very distinct protein sizes and binding affinities, showing that NOSCO NUS can reduce measurement times by an order of magnitude to make such highly informative NMR titration studies more broadly feasible.
Collapse
Affiliation(s)
- Javier A Romero
- Centre of New Technologies, University of Warsaw, Banacha 2C, 02-097, Warsaw, Poland
| | - Ewa K Nawrocka
- Centre of New Technologies, University of Warsaw, Banacha 2C, 02-097, Warsaw, Poland.,Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093, Warsaw, Poland
| | - Alexandra Shchukina
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Zwirki i Wigury 101, 02-089, Warsaw, Poland
| | - Francisco J Blanco
- Structural and Chemical Biology Department, Centro de Investigaciones Biológicas, CIB-CSIC, 28040, Madrid, Spain
| | - Tammo Diercks
- CIC bioGUNE, Parque Tecnológico de Bizkaia, Ed. 800, 48160-, Derio, Spain
| | | |
Collapse
|
24
|
Hu K, Xie L, Hanyu M, Zhang Y, Li L, Ma X, Nagatsu K, Suzuki H, Wang W, Zhang MR. Harnessing the PD-L1 interface peptide for positron emission tomography imaging of the PD-1 immune checkpoint. RSC Chem Biol 2020; 1:214-224. [PMID: 34458761 PMCID: PMC8341843 DOI: 10.1039/d0cb00070a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/04/2020] [Indexed: 12/18/2022] Open
Abstract
Interface peptides that mediate protein-protein interactions (PPI) are a class of important lead compounds for designing PPI inhibitors. However, their potential as precursors for radiotracers has never been exploited. Here we report that the interface peptides from programmed death-ligand 1 (PD-L1) can be used in positron emission tomography (PET) imaging of programmed cell death 1 (PD-1) with high accuracy and sensitivity. Moreover, the performance differentiation between murine PD-L1 derived interface peptide (mPep-1) and human PD-L1 derived interface peptide (hPep-1) as PET tracers for PD-1 unveiled an unprecedented role of a non-critical residue in target binding, highlighting the significance of PET imaging as a companion diagnostic in drug development. Collectively, this study not only provided a first-of-its-kind peptide-based PET tracer for PD-1 but also conveyed a unique paradigm for developing imaging agents for highly challenging protein targets, which could be used to identify other protein biomarkers involved in the PPI networks.
Collapse
Affiliation(s)
- Kuan Hu
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology Chiba, 263-8555 Japan
| | - Lin Xie
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology Chiba, 263-8555 Japan
| | - Masayuki Hanyu
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology Chiba, 263-8555 Japan
| | - Yiding Zhang
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology Chiba, 263-8555 Japan
| | - Lingyun Li
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology Beijing 100081 P. R. China
| | - Xiaohui Ma
- Department of Vascular Surgery, General Hospital of People's Liberation Army Beijing 100853 P. R. China
| | - Kotaro Nagatsu
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology Chiba, 263-8555 Japan
| | - Hisashi Suzuki
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology Chiba, 263-8555 Japan
| | - Weizhi Wang
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology Beijing 100081 P. R. China
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology Chiba, 263-8555 Japan
| |
Collapse
|
25
|
Liao TJ, Jang H, Fushman D, Nussinov R. SOS1 interacts with Grb2 through regions that induce closed nSH3 conformations. J Chem Phys 2020; 153:045106. [PMID: 32752665 PMCID: PMC7390601 DOI: 10.1063/5.0013926] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 07/12/2020] [Indexed: 12/18/2022] Open
Abstract
Grb2 is an adaptor protein connecting the epidermal growth factor receptor and the downstream Son of sevenless 1 (SOS1), a Ras-specific guanine nucleotide exchange factor (RasGEF), which exchanges GDP by GTP. Grb2 contains three SH domains: N-terminal SH3 (nSH3), SH2, and C-terminal SH3 (cSH3). The C-terminal proline-rich (PR) domain of SOS1 regulates nSH3 open/closed conformations. Earlier, several nSH3 binding motifs were identified in the PR domain. More recently, we characterized by nuclear magnetic resonance and replica exchange simulations possible cSH3 binding regions. Among them, we discovered a cSH3-specific binding region. However, how PR binding at these sites regulates the nSH3/cSH3 conformation has been unclear. Here, we explore the nSH3/cSH3 interaction with linked and truncated PR segments using molecular dynamics simulations. Our 248 μs simulations include 620 distinct trajectories, each 400 ns. We construct the effective free energy landscape to validate the nSH3/cSH3 binding sites. The nSH3/cSH3-SOS1 peptide complex models indicate that strong peptide binders attract the flexible nSH3 n-Src loop, inducing a closed conformation of nSH3; by contrast, the cSH3 conformation remains unchanged. Inhibitors that disrupt the Ras-SOS1 interaction have been designed; the conformational details uncovered here may assist in the design of polypeptides inhibiting Grb2-SOS1 interaction, thus SOS1 recruitment to the membrane where Ras resides.
Collapse
Affiliation(s)
| | - Hyunbum Jang
- Computational Structural Biology Section, Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, USA
| | | | | |
Collapse
|
26
|
Liao TJ, Jang H, Nussinov R, Fushman D. High-Affinity Interactions of the nSH3/cSH3 Domains of Grb2 with the C-Terminal Proline-Rich Domain of SOS1. J Am Chem Soc 2020; 142:3401-3411. [PMID: 31970984 PMCID: PMC8459210 DOI: 10.1021/jacs.9b10710] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Grb2 is an adaptor protein that recruits Ras-specific guanine nucleotide exchange factor, Son of Sevenless 1 (SOS1), to the plasma membrane. SOS1 exchanges GDP by GTP, activating Ras. Grb2 consists of an SH2 domain flanked by N- and C-terminal SH3 domains (nSH3/cSH3). Grb2 nSH3/cSH3 domains have strong binding affinity for the SOS1 proline-rich (PR) domain that mediates the Grb2-SOS1 interaction. The nSH3/cSH3 domains have distinct preferred binding motifs: PxxPxR for nSH3 and PxxxRxxKP for cSH3 (x represents any natural amino acid). Several nSH3-binding motifs have been identified in the SOS1 PR domain but none specific for cSH3 binding. Even though both nSH3 and cSH3 exhibit the strongest binding to the SOS1 peptide PVPPPVPPRRRP, this mutually exclusive binding combined with other potential nSH3/cSH3 binding regions in SOS1 makes understanding the Grb2-SOS1 interaction challenging. To identify the SOS1-cSH3 binding sites, we selected seven potential binding segments in SOS1. The synthesized peptides were tested for their binding to nSH3/cSH3. Our NMR data reveal that the PKLPPKTYKREH peptide has strong binding affinity for cSH3, but very weak for nSH3. The binding specificity suggests that the most likely Grb2-SOS1 binding mode is through nSH3-PVPPPVPPRRRP and cSH3-PKLPPKTYKREH interactions, which is supported by replica-exchange simulations for the Grb2-SOS1 complex models. We propose that nSH3/cSH3 binding peptides, which effectively interrupt Grb2-SOS1 association, can serve as tumor suppressors. The Grb2-SOS1 mechanism outlined here offers new venues for future therapeutic strategies for upstream mutations in cancer, such as in EGFR.
Collapse
Affiliation(s)
- Tsung-Jen Liao
- Biophysics Program, Institute for Physical Science and Technology , University of Maryland , College Park , Maryland 20742 , United States
- Computational Structural Biology Section, Basic Science Program , Frederick National Laboratory for Cancer Research , Frederick , Maryland 21702 , United States
| | - Hyunbum Jang
- Computational Structural Biology Section, Basic Science Program , Frederick National Laboratory for Cancer Research , Frederick , Maryland 21702 , United States
| | - Ruth Nussinov
- Computational Structural Biology Section, Basic Science Program , Frederick National Laboratory for Cancer Research , Frederick , Maryland 21702 , United States
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine , Tel Aviv University , Tel Aviv 69978 , Israel
| | - David Fushman
- Biophysics Program, Institute for Physical Science and Technology , University of Maryland , College Park , Maryland 20742 , United States
- Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Organization , University of Maryland , College Park , Maryland 20742 , United States
| |
Collapse
|
27
|
Molecular Dynamics model of peptide-protein conjugation: case study of covalent complex between Sos1 peptide and N-terminal SH3 domain from Grb2. Sci Rep 2019; 9:20219. [PMID: 31882608 PMCID: PMC6934455 DOI: 10.1038/s41598-019-56078-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 12/04/2019] [Indexed: 12/31/2022] Open
Abstract
We have investigated covalent conjugation of VPPPVPPRRRX′ peptide (where X′ denotes Nε-chloroacetyl lysine) to N-terminal SH3 domain from adapter protein Grb2. Our experimental results confirmed that the peptide first binds to the SH3 domain noncovalently before establishing a covalent linkage through reaction of X′ with the target cysteine residue C32. We have also confirmed that this reaction involves a thiolate-anion form of C32 and follows the SN2 mechanism. For this system, we have developed a new MD-based protocol to model the formation of covalent conjugate. The simulation starts with the known coordinates of the noncovalent complex. When two reactive groups come into contact during the course of the simulation, the reaction is initiated. The reaction is modeled via gradual interpolation between the two sets of force field parameters that are representative of the noncovalent and covalent complexes. The simulation proceeds smoothly, with no appreciable perturbations to temperature, pressure or volume, and results in a high-quality MD model of the covalent complex. The validity of this model is confirmed using the experimental chemical shift data. The new MD-based approach offers a valuable tool to explore the mechanics of protein-peptide conjugation and build accurate models of covalent complexes.
Collapse
|
28
|
Tang W, Zhao Z, Wang C, Ye T, Yang B. Molecular design and optimization of hepatic cancer SLP76-derived PLCγ1 SH3-binding peptide with the systematic N-substitution of peptide PXXP motif. J Mol Recognit 2019; 32:e2806. [PMID: 31397025 DOI: 10.1002/jmr.2806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/24/2019] [Accepted: 07/01/2019] [Indexed: 12/23/2022]
Abstract
The phospholipase Cγ1 (PLCγ1) is essential for T-cell signaling and activation in hepatic cancer immune response, which has a regulatory Src homology 3 (SH3) domain that can specifically recognize and interact with the PXXP-containing decapeptide segment (185 QPPVPPQRPM194 , termed as SLP76185-194 peptide) of adaptor protein SLP76 following T-cell receptor ligation. The isolated peptide can only bind to the PLCγ1 SH3 domain with a moderate affinity due to lack of protein context support. Instead of the traditional natural residue mutagenesis that is limited by low structural diversity and shifted target specificity, we herein attempt to improve the peptide affinity by replacing the two key proline residues Pro187 and Pro190 of SLP76185-194 PXXP motif with nonnatural N-substituted amino acids, as the proline is the only endogenous N-substituted amino acid. The replacement would increase peptide flexibility but can restore peptide activity by establishing additional interactions with the domain. Structural analysis reveals that the domain pocket can be divided into a large amphipathic region and a small negatively charged region; they accommodate hydrophobic, aromatic, polar, and moderate-sized N-substituted amino acid types. A systematic replacement combination profile between the peptide residues Pro187 and Pro190 is created by structural modeling, dynamics simulation, and energetics analysis, from which six improved and two reduced N-substituted peptides as well as native SLP76185-194 peptide are identified and tested for their binding affinity to the recombinant protein of the human PLCγ1 SH3 domain using fluorescence-based assays. Two N-substituted peptides, SLP76185-194 (N-Leu187/N-Gln190) and SLP76185-194 (N-Thr187/N-Gln190), are designed to have high potency (Kd = 0.67 ± 0.18 and 1.7 ± 0.3 μM, respectively), with affinity improvement by, respectively, 8.5-fold and 3.4-fold relative to native peptide (Kd = 5.7 ± 1.2 μM).
Collapse
Affiliation(s)
- Wenqing Tang
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhiying Zhao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Wang
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tao Ye
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Biwei Yang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
29
|
Fu Y, He P, Zhou Y, Huang S, Liang L, Liu S. Exploring the systematic effect of
N
‐substituted PxxP motifs on peptoid affinity to ARHGEF5/TIM SH3 domain and its relationship with ARHGEF5/TIM activation. Proteins 2019; 87:979-991. [PMID: 31197859 DOI: 10.1002/prot.25760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 06/04/2019] [Accepted: 06/07/2019] [Indexed: 11/09/2022]
Affiliation(s)
- Yong Fu
- Department of Endocrine and Breast SurgeryThe First Affiliated Hospital of Chongqing Medical University Chongqing China
- Department of Cardiothoracic SurgeryDianjiang People's Hospital of Chongqing Chongqing China
| | - Ping He
- Department of Cardiac SurgerySouthwest Hospital, Third Army Medical University Chongqing China
| | - Yu Zhou
- Department of Cardiothoracic SurgeryDianjiang People's Hospital of Chongqing Chongqing China
| | - Shengyuan Huang
- Department of Cardiothoracic SurgeryDianjiang People's Hospital of Chongqing Chongqing China
| | - Lin Liang
- Department of Cardiothoracic SurgeryDianjiang People's Hospital of Chongqing Chongqing China
| | - Shengchun Liu
- Department of Endocrine and Breast SurgeryThe First Affiliated Hospital of Chongqing Medical University Chongqing China
| |
Collapse
|
30
|
Wellhöfer I, Frydenvang K, Kotesova S, Christiansen AM, Laursen JS, Olsen CA. Functionalized Helical β-Peptoids. J Org Chem 2019; 84:3762-3779. [DOI: 10.1021/acs.joc.9b00218] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Isabelle Wellhöfer
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Karla Frydenvang
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Simona Kotesova
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Andreas M. Christiansen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Jonas S. Laursen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Christian A. Olsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| |
Collapse
|
31
|
Enhancing Aromatic Foldamer Helix Dynamics to Probe Interactions with Protein Surfaces. European J Org Chem 2018. [DOI: 10.1002/ejoc.201800855] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
32
|
Ke M, Liu J, Chen W, Chen L, Gao W, Qin Y, He A, Chu B, Tang J, Xu R, Deng Y, Tian R. Integrated and Quantitative Proteomic Approach for Charting Temporal and Endogenous Protein Complexes. Anal Chem 2018; 90:12574-12583. [PMID: 30280895 DOI: 10.1021/acs.analchem.8b02667] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | - Jun Tang
- Shenzhen People’s Hospital, The Second Clinical Medical College of Jinan University, Shenzhen 518020, China
| | - Ruilian Xu
- Shenzhen People’s Hospital, The Second Clinical Medical College of Jinan University, Shenzhen 518020, China
| | - Yi Deng
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen 518055, China
| | - Ruijun Tian
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen 518055, China
| |
Collapse
|
33
|
Li T, Chen X, Cai Y, Dai J. Artificial Protein Scaffold System (AProSS): An efficient method to optimize exogenous metabolic pathways in Saccharomyces cerevisiae. Metab Eng 2018; 49:13-20. [PMID: 30010058 DOI: 10.1016/j.ymben.2018.07.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/11/2018] [Accepted: 07/12/2018] [Indexed: 01/15/2023]
Abstract
Scaffold proteins influence cellular signaling by orchestrating multiple enzymes, receptors or ion channels, and could be tailored to enhance the efficiency of biochemical reactions by positioning related enzymes physically together. However, the number of applicable domains remains small, and the construction of scaffold proteins with optimal domain ratio could be tedious and time-consuming. In this study, we outlined a modular design to quickly assemble scaffold proteins using protein interaction domains, which have been constructed into a standardized vector. We generated multiple protein interaction domains and ligands for making artificial scaffold proteins. At the same time, we developed a robust Golden-Gate-based molecular toolkit for the construction of artificial scaffold proteins, allowing a variance of domain types, number, and positions. The synthesized domain-ligand interaction was verified by yeast two-hybrid and split-GFP assays. Using synthetic scaffolds, we demonstrated an increase in the yield of two target products by 29% and 63% respectively. Moreover, we demonstrated that the synthetic scaffold could be applied to rewire the metabolic flux. Our system could be a useful tool for metabolic engineering and beyond.
Collapse
Affiliation(s)
- Tianyi Li
- Key Laboratory of Industrial Biocatalysis (Ministry of Education) and Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiuqi Chen
- Key Laboratory of Industrial Biocatalysis (Ministry of Education) and Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Department of Biology, Johns Hopkins University, 3400N. Charles Street, Baltimore, MD, USA
| | - Yizhi Cai
- Shenzhen Key Laboratory of Synthetic Genomics and Center for Synthetic Genomics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, M1 7DN, Manchester, UK.
| | - Junbiao Dai
- Key Laboratory of Industrial Biocatalysis (Ministry of Education) and Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Shenzhen Key Laboratory of Synthetic Genomics and Center for Synthetic Genomics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
34
|
Haidar M, Latré de Laté P, Kennedy EJ, Langsley G. Cell penetrating peptides to dissect host-pathogen protein-protein interactions in Theileria-transformed leukocytes. Bioorg Med Chem 2018; 26:1127-1134. [PMID: 28917447 PMCID: PMC5842112 DOI: 10.1016/j.bmc.2017.08.056] [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: 06/27/2017] [Revised: 08/24/2017] [Accepted: 08/30/2017] [Indexed: 10/18/2022]
Abstract
One powerful application of cell penetrating peptides is the delivery into cells of molecules that function as specific competitors or inhibitors of protein-protein interactions. Ablating defined protein-protein interactions is a refined way to explore their contribution to a particular cellular phenotype in a given disease context. Cell-penetrating peptides can be synthetically constrained through various chemical modifications that stabilize a given structural fold with the potential to improve competitive binding to specific targets. Theileria-transformed leukocytes display high PKA activity, but PKA is an enzyme that plays key roles in multiple cellular processes; consequently genetic ablation of kinase activity gives rise to a myriad of confounding phenotypes. By contrast, ablation of a specific kinase-substrate interaction has the potential to give more refined information and we illustrate this here by describing how surgically ablating PKA interactions with BAD gives precise information on the type of glycolysis performed by Theileria-transformed leukocytes. In addition, we provide two other examples of how ablating specific protein-protein interactions in Theileria-infected leukocytes leads to precise phenotypes and argue that constrained penetrating peptides have great therapeutic potential to combat infectious diseases in general.
Collapse
Affiliation(s)
- Malak Haidar
- Inserm U1016, Cnrs UMR8104, Cochin Institute, Paris 75014, France; Laboratoire de Biologie Cellulaire Comparative des Apicomplexes, Faculté de Médecine, Université Paris Descartes - Sorbonne Paris Cité, 75014, France; Pathogen Genomics Laboratory, Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Perle Latré de Laté
- Inserm U1016, Cnrs UMR8104, Cochin Institute, Paris 75014, France; Laboratoire de Biologie Cellulaire Comparative des Apicomplexes, Faculté de Médecine, Université Paris Descartes - Sorbonne Paris Cité, 75014, France
| | - Eileen J Kennedy
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, United States
| | - Gordon Langsley
- Inserm U1016, Cnrs UMR8104, Cochin Institute, Paris 75014, France; Laboratoire de Biologie Cellulaire Comparative des Apicomplexes, Faculté de Médecine, Université Paris Descartes - Sorbonne Paris Cité, 75014, France.
| |
Collapse
|
35
|
Darapaneni CM, Kaniraj PJ, Maayan G. Water soluble hydrophobic peptoids via a minor backbone modification. Org Biomol Chem 2018; 16:1480-1488. [DOI: 10.1039/c7ob02928d] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The incorporation of piperazine or homopiperazine within hydrophobic peptoid scaffolds leads to their water solubility while increasing their overall conformational order in water.
Collapse
Affiliation(s)
| | - Prathap Jeya Kaniraj
- Schulich Faculty of Chemistry
- Technion – Israel Institute of Technology
- Technion City
- Israel
| | - Galia Maayan
- Schulich Faculty of Chemistry
- Technion – Israel Institute of Technology
- Technion City
- Israel
| |
Collapse
|
36
|
Lella M, Mahalakshmi R. Solvation driven conformational transitions in the second transmembrane domain of mycobacteriophage holin. Biopolymers 2017; 108. [DOI: 10.1002/bip.22894] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/11/2016] [Accepted: 05/31/2016] [Indexed: 12/29/2022]
Affiliation(s)
- Muralikrishna Lella
- Molecular Biophysics Laboratory, Department of Biological Sciences; Indian Institute of Science Education and Research; Bhopal 462023 India
| | - Radhakrishnan Mahalakshmi
- Molecular Biophysics Laboratory, Department of Biological Sciences; Indian Institute of Science Education and Research; Bhopal 462023 India
| |
Collapse
|
37
|
Altamirano S, Chandrasekaran S, Kozubowski L. Mechanisms of Cytokinesis in Basidiomycetous Yeasts. FUNGAL BIOL REV 2017; 31:73-87. [PMID: 28943887 DOI: 10.1016/j.fbr.2016.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
While mechanisms of cytokinesis exhibit considerable plasticity, it is difficult to precisely define the level of conservation of this essential part of cell division in fungi, as majority of our knowledge is based on ascomycetous yeasts. However, in the last decade more details have been uncovered regarding cytokinesis in the second largest fungal phylum, basidiomycetes, specifically in two yeasts, Cryptococcus neoformans and Ustilago maydis. Based on these findings, and current sequenced genomes, we summarize cytokinesis in basidiomycetous yeasts, indicating features that may be unique to this phylum, species-specific characteristics, as well as mechanisms that may be common to all eukaryotes.
Collapse
Affiliation(s)
- Sophie Altamirano
- Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, USA
| | | | - Lukasz Kozubowski
- Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
38
|
Vasa SK, Rovó P, Giller K, Becker S, Linser R. Access to aliphatic protons as reporters in non-deuterated proteins by solid-state NMR. Phys Chem Chem Phys 2017; 18:8359-63. [PMID: 26686237 DOI: 10.1039/c5cp06601h] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Interactions within proteins, with their surrounding, and with other molecules are mediated mostly by hydrogen atoms. In fully protonated, inhomogeneous, or larger proteins, however, aliphatic proton shifts tend to show little dispersion despite fast Magic-Angle Spinning. 3D correlations dispersing aliphatic proton shifts by their better resolved amide N/H shifts can alleviate this problem. Using inverse second-order cross-polarization (iSOCP), we here introduce dedicated and improved means to sensitively link site-specific chemical shift information from aliphatic protons with a backbone amide resolution. Thus, even in cases where protein deuteration is impossible, this approach may enable access to various aspects of protein functions that are reported on by protons.
Collapse
Affiliation(s)
- Suresh Kumar Vasa
- Max-Planck Institute for Biophysical Chemistry, Department NMR-Based Structural Biology, Am Fassberg 11, 37077 Göttingen, Germany.
| | - Petra Rovó
- Max-Planck Institute for Biophysical Chemistry, Department NMR-Based Structural Biology, Am Fassberg 11, 37077 Göttingen, Germany.
| | - Karin Giller
- Max-Planck Institute for Biophysical Chemistry, Department NMR-Based Structural Biology, Am Fassberg 11, 37077 Göttingen, Germany.
| | - Stefan Becker
- Max-Planck Institute for Biophysical Chemistry, Department NMR-Based Structural Biology, Am Fassberg 11, 37077 Göttingen, Germany.
| | - Rasmus Linser
- Max-Planck Institute for Biophysical Chemistry, Department NMR-Based Structural Biology, Am Fassberg 11, 37077 Göttingen, Germany.
| |
Collapse
|
39
|
Perez-Lopez AM, Valero E, Bradley M. Synthesis and optimization of a reactive oxygen species responsive cellular delivery system. NEW J CHEM 2017. [DOI: 10.1039/c6nj02985j] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Reactive oxygen species responsive delivery systems for the detection of peroxides in live macrophages have been designed. The oxidative cleavage of a boronic ester to a phenol triggered by hydrogen peroxide followed by self-immolation of a ROS-sensitive cleavable linkervia1,6-elimination allowed the disturbance of the fluorescence resonance energy transfer turning on the near-infrared fluorescence.
Collapse
Affiliation(s)
| | - Elsa Valero
- School of Chemistry
- University of Edinburgh
- Edinburgh EH9 3JJ
- UK
| | - Mark Bradley
- School of Chemistry
- University of Edinburgh
- Edinburgh EH9 3JJ
- UK
| |
Collapse
|
40
|
Craven TW, Bonneau R, Kirshenbaum K. PPII Helical Peptidomimetics Templated by Cation-π Interactions. Chembiochem 2016; 17:1824-1828. [PMID: 27539882 DOI: 10.1002/cbic.201600248] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Indexed: 11/06/2022]
Abstract
Poly-proline type II (PPII) helical PXXP motifs are the recognition elements for a variety of protein-protein interactions that are critical for cellular signaling. Despite development of protocols for locking peptides into α-helical and β-strand conformations, there remains a lack of analogous methods for generating mimics of PPII helical structures. We describe herein a strategy to enforce PPII helical secondary structure in the 19-residue TrpPlexus miniature protein. Through sequence variation, we showed that a network of cation-π interactions could drive the formation of PPII helical conformations for both peptide and N-substituted glycine peptoid residues. The achievement of chemically diverse PPII helical scaffolds provides a new route towards discovering peptidomimetic inhibitors of protein-protein interactions mediated by PXXP motifs.
Collapse
Affiliation(s)
- Timothy W Craven
- Department of Chemistry, New York University, 100 Washington Square East, New York, NY, 10003, USA.,Department of Biology, Center for Genomics and Systems Biology, New York University, 12 Waverly Pl., New York, NY, 10003, USA
| | - Richard Bonneau
- Department of Biology, Center for Genomics and Systems Biology, New York University, 12 Waverly Pl., New York, NY, 10003, USA.,Department of Computer Science, Courant Institute of Mathematical Sciences, New York University, New York, NY, 10003, USA.,Simons Center for Data Analysis, 160 5th Ave., New York, NY, 10010, USA
| | - Kent Kirshenbaum
- Department of Chemistry, New York University, 100 Washington Square East, New York, NY, 10003, USA.
| |
Collapse
|
41
|
Han S, Liu Q, Wang F, Yuan Z. Targeting the SH3 domain of human osteoclast-stimulating factor with rationally designed peptoid inhibitors. J Pept Sci 2016; 22:533-9. [PMID: 27443979 DOI: 10.1002/psc.2901] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 05/24/2016] [Accepted: 05/31/2016] [Indexed: 11/12/2022]
Affiliation(s)
- Shijie Han
- Department of Orthopedics; Shandong Provincial Hospital; Jinan 250021 China
| | - Qian Liu
- Department of Pain Management; Qilu Hospital of Shandong University; Jinan 250012 China
| | - Feng Wang
- Department of Orthopedics; Shandong Provincial Hospital; Jinan 250021 China
| | - Zenong Yuan
- Department of Orthopedics; Shandong Provincial Hospital; Jinan 250021 China
| |
Collapse
|
42
|
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
|
43
|
Endophilin B2 promotes inner mitochondrial membrane degradation by forming heterodimers with Endophilin B1 during mitophagy. Sci Rep 2016; 6:25153. [PMID: 27112121 PMCID: PMC4845016 DOI: 10.1038/srep25153] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 04/11/2016] [Indexed: 11/21/2022] Open
Abstract
Mitochondrial sequestration by autophagosomes is a key step in mitophagy while the mechanisms mediating this process are not fully understood. It has been reported that Endophilin B1 (EB1) promotes mitochondrial sequestration by binding and shaping membrane. However, the role of EB1 homolog Endophilin B2 (EB2) in mitophagy remains unclear. Here we report that EB2 plays an indispensable role in mitochondria sequestration and inner mitochondrial membrane (IMM) protein degradation during mitophagy. Similar to EB1, EB2 aggregates into foci and then translocates to damaged mitochondria. Loss of either EB2 and/or EB1 significantly enervates the foci translocation to fragmented mitochondria and IMM degradation, and the EB1/EB2 heterodimer formed by EB1/EB2 interaction promotes the above process. We noticed that, it is the dimer domain of EB2 but not that of EB1 mediating the heterodimer formation, manifesting the importance of EB2 in mitophagy. Furthermore, we demonstrate that the EB foci formation is closely regulated by the PINK1-Parkin signaling pathway. From these results, we propose that EB1/EB2 heterodimers may serve as linkers between damaged mitochondria and phagophores during mitophagy.
Collapse
|
44
|
Ren J, Tian Y, Hossain E, Connolly MD. Fragmentation Patterns and Mechanisms of Singly and Doubly Protonated Peptoids Studied by Collision Induced Dissociation. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2016; 27:646-661. [PMID: 26832347 DOI: 10.1007/s13361-016-1341-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 01/07/2016] [Accepted: 01/09/2016] [Indexed: 06/05/2023]
Abstract
Peptoids are peptide-mimicking oligomers consisting of N-alkylated glycine units. The fragmentation patterns for six singly and doubly protonated model peptoids were studied via collision-induced dissociation tandem mass spectrometry. The experiments were carried out on a triple quadrupole mass spectrometer with an electrospray ionization source. Both singly and doubly protonated peptoids were found to fragment mainly at the backbone amide bonds to produce peptoid B-type N-terminal fragment ions and Y-type C-terminal fragment ions. However, the relative abundances of B- versus Y-ions were significantly different. The singly protonated peptoids fragmented by producing highly abundant Y-ions and lesser abundant B-ions. The Y-ion formation mechanism was studied through calculating the energetics of truncated peptoid fragment ions using density functional theory and by controlled experiments. The results indicated that Y-ions were likely formed by transferring a proton from the C-H bond of the N-terminal fragments to the secondary amine of the C-terminal fragments. This proton transfer is energetically favored, and is in accord with the observation of abundant Y-ions. The calculations also indicated that doubly protonated peptoids would fragment at an amide bond close to the N-terminus to yield a high abundance of low-mass B-ions and high-mass Y-ions. The results of this study provide further understanding of the mechanisms of peptoid fragmentation and, therefore, are a valuable guide for de novo sequencing of peptoid libraries synthesized via combinatorial chemistry. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Jianhua Ren
- Department of Chemistry, University of the Pacific, 3601 Pacific Avenue, Stockton, CA, 95211, USA.
| | - Yuan Tian
- Department of Chemistry, University of the Pacific, 3601 Pacific Avenue, Stockton, CA, 95211, USA
| | - Ekram Hossain
- Department of Chemistry, University of the Pacific, 3601 Pacific Avenue, Stockton, CA, 95211, USA
| | - Michael D Connolly
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| |
Collapse
|
45
|
Leithe E. Regulation of connexins by the ubiquitin system: Implications for intercellular communication and cancer. Biochim Biophys Acta Rev Cancer 2016; 1865:133-46. [DOI: 10.1016/j.bbcan.2016.02.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/15/2016] [Accepted: 02/04/2016] [Indexed: 12/31/2022]
|
46
|
Craven TW, Cho MK, Traaseth NJ, Bonneau R, Kirshenbaum K. A Miniature Protein Stabilized by a Cation-π Interaction Network. J Am Chem Soc 2016; 138:1543-50. [PMID: 26812069 PMCID: PMC4867217 DOI: 10.1021/jacs.5b10285] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The design of folded miniature proteins is predicated on establishing noncovalent interactions that direct the self-assembly of discrete thermostable tertiary structures. In this work, we describe how a network of cation-π interactions present in proteins containing "WSXWS motifs" can be emulated to stabilize the core of a miniature protein. This 19-residue protein sequence recapitulates a set of interdigitated arginine and tryptophan residues that stabilize a distinctive β-strand:loop:PPII-helix topology. Validation of the compact fold determined by NMR was carried out by mutagenesis of the cation-π network and by comparison to the corresponding disulfide-bridged structure. These results support the involvement of a coordinated set of cation-π interactions that stabilize the tertiary structure.
Collapse
Affiliation(s)
- Timothy W. Craven
- Department of Chemistry, New York University, 100 Washington Square East, New York, NY
- Department of Biology, Center for Genomics and Systems Biology, New York University, 12 Waverly Pl., New York, NY
| | - Min-Kyu Cho
- Department of Chemistry, New York University, 100 Washington Square East, New York, NY
| | - Nathaniel J. Traaseth
- Department of Chemistry, New York University, 100 Washington Square East, New York, NY
| | - Richard Bonneau
- Department of Biology, Center for Genomics and Systems Biology, New York University, 12 Waverly Pl., New York, NY
- Department of Computer Science, Courant Institute of Mathematical Sciences, New York University, New York, NY
- Simons Center for Data Analysis, New York, NY
| | - Kent Kirshenbaum
- Department of Chemistry, New York University, 100 Washington Square East, New York, NY
| |
Collapse
|
47
|
Efficient production of gamma-aminobutyric acid using Escherichia coli by co-localization of glutamate synthase, glutamate decarboxylase, and GABA transporter. J Ind Microbiol Biotechnol 2015; 43:79-86. [PMID: 26620318 DOI: 10.1007/s10295-015-1712-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 11/13/2015] [Indexed: 10/22/2022]
Abstract
Gamma-aminobutyric acid (GABA) is an important bio-product, which is used in pharmaceutical formulations, nutritional supplements, and biopolymer monomer. The traditional GABA process involves the decarboxylation of glutamate. However, the direct production of GABA from glucose is a more efficient process. To construct the recombinant strains of Escherichia coli, a novel synthetic scaffold was introduced. By carrying out the co-localization of glutamate synthase, glutamate decarboxylase, and GABA transporter, we redirected the TCA cycle flux to GABA pathway. The genetically engineered E. coli strain produced 1.08 g/L of GABA from 10 g/L of initial glucose. Thus, with the introduction of a synthetic scaffold, we increased GABA production by 2.2-fold. The final GABA concentration was increased by 21.8% by inactivating competing pathways.
Collapse
|
48
|
Pelay-Gimeno M, Glas A, Koch O, Grossmann TN. Structure-Based Design of Inhibitors of Protein-Protein Interactions: Mimicking Peptide Binding Epitopes. Angew Chem Int Ed Engl 2015; 54:8896-927. [PMID: 26119925 PMCID: PMC4557054 DOI: 10.1002/anie.201412070] [Citation(s) in RCA: 537] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Indexed: 12/15/2022]
Abstract
Protein-protein interactions (PPIs) are involved at all levels of cellular organization, thus making the development of PPI inhibitors extremely valuable. The identification of selective inhibitors is challenging because of the shallow and extended nature of PPI interfaces. Inhibitors can be obtained by mimicking peptide binding epitopes in their bioactive conformation. For this purpose, several strategies have been evolved to enable a projection of side chain functionalities in analogy to peptide secondary structures, thereby yielding molecules that are generally referred to as peptidomimetics. Herein, we introduce a new classification of peptidomimetics (classes A-D) that enables a clear assignment of available approaches. Based on this classification, the Review summarizes strategies that have been applied for the structure-based design of PPI inhibitors through stabilizing or mimicking turns, β-sheets, and helices.
Collapse
Affiliation(s)
- Marta Pelay-Gimeno
- Chemical Genomics Centre of the Max Planck SocietyOtto-Hahn-Strasse 15, 44227 Dortmund (Germany) E-mail:
| | - Adrian Glas
- Chemical Genomics Centre of the Max Planck SocietyOtto-Hahn-Strasse 15, 44227 Dortmund (Germany) E-mail:
| | - Oliver Koch
- TU Dortmund University, Department of Chemistry and Chemical BiologyOtto-Hahn-Strasse 6, 44227 Dortmund (Germany)
| | - Tom N Grossmann
- Chemical Genomics Centre of the Max Planck SocietyOtto-Hahn-Strasse 15, 44227 Dortmund (Germany) E-mail:
- TU Dortmund University, Department of Chemistry and Chemical BiologyOtto-Hahn-Strasse 6, 44227 Dortmund (Germany)
| |
Collapse
|
49
|
Pelay-Gimeno M, Glas A, Koch O, Grossmann TN. Strukturbasierte Entwicklung von Protein-Protein-Interaktionsinhibitoren: Stabilisierung und Nachahmung von Peptidliganden. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201412070] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
50
|
Engel-Andreasen J, Wich K, Laursen JS, Harris P, Olsen CA. Effects of Thionation and Fluorination on Cis–Trans Isomerization in Tertiary Amides: An Investigation of N-Alkylglycine (Peptoid) Rotamers. J Org Chem 2015; 80:5415-27. [DOI: 10.1021/acs.joc.5b00048] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Jens Engel-Andreasen
- Department
of Chemistry, Technical University of Denmark, Kemitorvet 207, DK-2800 Kongens Lyngby, Denmark
| | - Kathrine Wich
- Center for Biopharmaceuticals and Department of Drug Design & Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Jonas S. Laursen
- Center for Biopharmaceuticals and Department of Drug Design & Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Pernille Harris
- Department
of Chemistry, Technical University of Denmark, Kemitorvet 207, DK-2800 Kongens Lyngby, Denmark
| | - Christian A. Olsen
- Department
of Chemistry, Technical University of Denmark, Kemitorvet 207, DK-2800 Kongens Lyngby, Denmark
- Center for Biopharmaceuticals and Department of Drug Design & Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| |
Collapse
|