301
|
Chen J, Cheong HH, Siu SWI. xDeep-AcPEP: Deep Learning Method for Anticancer Peptide Activity Prediction Based on Convolutional Neural Network and Multitask Learning. J Chem Inf Model 2021; 61:3789-3803. [PMID: 34327990 DOI: 10.1021/acs.jcim.1c00181] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cancer is one of the leading causes of death worldwide. Conventional cancer treatment relies on radiotherapy and chemotherapy, but both methods bring severe side effects to patients, as these therapies not only attack cancer cells but also damage normal cells. Anticancer peptides (ACPs) are a promising alternative as therapeutic agents that are efficient and selective against tumor cells. Here, we propose a deep learning method based on convolutional neural networks to predict biological activity (EC50, LC50, IC50, and LD50) against six tumor cells, including breast, colon, cervix, lung, skin, and prostate. We show that models derived with multitask learning achieve better performance than conventional single-task models. In repeated 5-fold cross validation using the CancerPPD data set, the best models with the applicability domain defined obtain an average mean squared error of 0.1758, Pearson's correlation coefficient of 0.8086, and Kendall's correlation coefficient of 0.6156. As a step toward model interpretability, we infer the contribution of each residue in the sequence to the predicted activity by means of feature importance weights derived from the convolutional layers of the model. The present method, referred to as xDeep-AcPEP, will help to identify effective ACPs in rational peptide design for therapeutic purposes. The data, script files for reproducing the experiments, and the final prediction models can be downloaded from http://github.com/chen709847237/xDeep-AcPEP. The web server to directly access this prediction method is at https://app.cbbio.online/acpep/home.
Collapse
Affiliation(s)
- Jiarui Chen
- Department of Computer and Information Science, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Hong Hin Cheong
- Department of Computer and Information Science, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Shirley W I Siu
- Department of Computer and Information Science, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China.,School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 USM, Penang, Malaysia
| |
Collapse
|
302
|
Herlan CN, Sonnefeld A, Gloge T, Brückel J, Schlee LC, Muhle-Goll C, Nieger M, Bräse S. Macrocyclic Tetramers-Structural Investigation of Peptide-Peptoid Hybrids. Molecules 2021; 26:molecules26154548. [PMID: 34361700 PMCID: PMC8348019 DOI: 10.3390/molecules26154548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/23/2021] [Accepted: 07/24/2021] [Indexed: 11/16/2022] Open
Abstract
Outstanding affinity and specificity are the main characteristics of peptides, rendering them interesting compounds for basic and medicinal research. However, their biological applicability is limited due to fast proteolytic degradation. The use of mimetic peptoids overcomes this disadvantage, though they lack stereochemical information at the α-carbon. Hybrids composed of amino acids and peptoid monomers combine the unique properties of both parent classes. Rigidification of the backbone increases the affinity towards various targets. However, only little is known about the spatial structure of such constrained hybrids. The determination of the three-dimensional structure is a key step for the identification of new targets as well as the rational design of bioactive compounds. Herein, we report the synthesis and the structural elucidation of novel tetrameric macrocycles. Measurements were taken in solid and solution states with the help of X-ray scattering and NMR spectroscopy. The investigations made will help to find diverse applications for this new, promising compound class.
Collapse
Affiliation(s)
- Claudine Nicole Herlan
- Institute of Organic Chemistry, Karlsruhe Institute of Technology, Fritz-Haber-Weg 6, 76131 Karlsruhe, Germany; (C.N.H.); (J.B.); (L.C.S.)
| | - Anna Sonnefeld
- Institute for Biological Interfaces 4, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany; (A.S.); (T.G.); (C.M.-G.)
| | - Thomas Gloge
- Institute for Biological Interfaces 4, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany; (A.S.); (T.G.); (C.M.-G.)
| | - Julian Brückel
- Institute of Organic Chemistry, Karlsruhe Institute of Technology, Fritz-Haber-Weg 6, 76131 Karlsruhe, Germany; (C.N.H.); (J.B.); (L.C.S.)
| | - Luisa Chiara Schlee
- Institute of Organic Chemistry, Karlsruhe Institute of Technology, Fritz-Haber-Weg 6, 76131 Karlsruhe, Germany; (C.N.H.); (J.B.); (L.C.S.)
| | - Claudia Muhle-Goll
- Institute for Biological Interfaces 4, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany; (A.S.); (T.G.); (C.M.-G.)
| | - Martin Nieger
- Department of Chemistry, University of Helsinki, P.O. Box 55 (A.I. Virtasen aukio 1), FIN-00014 Helsinki, Finland;
| | - Stefan Bräse
- Institute of Organic Chemistry, Karlsruhe Institute of Technology, Fritz-Haber-Weg 6, 76131 Karlsruhe, Germany; (C.N.H.); (J.B.); (L.C.S.)
- Institute of Biological and Chemical Systems—Functional Molecular Systems, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
- Correspondence:
| |
Collapse
|
303
|
Liu J, Zou S, Zhang Y, Lin R, Duan Y, He W, Yang Z. Discovery of Antitumor Active Peptides Derived from Peroxiredoxin 5. ChemMedChem 2021; 16:3477-3483. [PMID: 34313010 DOI: 10.1002/cmdc.202100323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/16/2021] [Indexed: 01/21/2023]
Abstract
The peroxiredoxin 5 (PRDX5) is a member of peroxiredoxins with antitumor activity. However, as a recombinant protein, PRDX5 is restricted in clinic due to high cost and keeping high dose in medication. The alternative way is to explore the antitumor active fragments of PRDX5 for potential of peptide drugs. According to the sequence, crystal structure and enzyme function of PRDX5, seven peptides were designed and named as IMB-P1∼7. The peptide IMB-P1 (AFTPGCSKTHLPGFVEQAEAL) containing critical residue C47 exhibited antitumor activity similar to PRDX5 in vivo. Transcriptome analysis showed peptide IMB-P1 could make influence on expression of multiple genes involved in tumorigenesis and deterioration. Besides, an important discovery is the down-regulation of oxidation-related genes. In CT26 cells, IMB-P1 carried similar antitumor activity with increasing ROS level to intact PRDX5. The results demonstrated that peptide IMB-P1 with easier synthesis from PRDX5 may serve as a promising antitumor candidate.
Collapse
Affiliation(s)
- Juanjuan Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Tiantanxili, Dongcheng District, Beijing, China
| | - Sen Zou
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Tiantanxili, Dongcheng District, Beijing, China
| | - Yan Zhang
- Shen Yang Tonglian Group Co., Ltd., Dadong District, Shen Yang, China
| | - Ru Lin
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Tiantanxili, Dongcheng District, Beijing, China
| | - Yanbo Duan
- Shen Yang Tonglian Group Co., Ltd., Dadong District, Shen Yang, China
| | - Weiqing He
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Tiantanxili, Dongcheng District, Beijing, China
| | - Zhaoyong Yang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Tiantanxili, Dongcheng District, Beijing, China
| |
Collapse
|
304
|
Vithani N, Ward MD, Zimmerman MI, Novak B, Borowsky JH, Singh S, Bowman GR. SARS-CoV-2 Nsp16 activation mechanism and a cryptic pocket with pan-coronavirus antiviral potential. Biophys J 2021; 120:2880-2889. [PMID: 33794150 PMCID: PMC8007187 DOI: 10.1016/j.bpj.2021.03.024] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/17/2021] [Accepted: 03/25/2021] [Indexed: 01/12/2023] Open
Abstract
Coronaviruses have caused multiple epidemics in the past two decades, in addition to the current COVID-19 pandemic that is severely damaging global health and the economy. Coronaviruses employ between 20 and 30 proteins to carry out their viral replication cycle, including infection, immune evasion, and replication. Among these, nonstructural protein 16 (Nsp16), a 2'-O-methyltransferase, plays an essential role in immune evasion. Nsp16 achieves this by mimicking its human homolog, CMTr1, which methylates mRNA to enhance translation efficiency and distinguish self from other. Unlike human CMTr1, Nsp16 requires a binding partner, Nsp10, to activate its enzymatic activity. The requirement of this binding partner presents two questions that we investigate in this manuscript. First, how does Nsp10 activate Nsp16? Although experimentally derived structures of the active Nsp16/Nsp10 complex exist, structures of inactive, monomeric Nsp16 have yet to be solved. Therefore, it is unclear how Nsp10 activates Nsp16. Using over 1 ms of molecular dynamics simulations of both Nsp16 and its complex with Nsp10, we investigate how the presence of Nsp10 shifts Nsp16's conformational ensemble to activate it. Second, guided by this activation mechanism and Markov state models, we investigate whether Nsp16 adopts inactive structures with cryptic pockets that, if targeted with a small molecule, could inhibit Nsp16 by stabilizing its inactive state. After identifying such a pocket in SARS-CoV2 Nsp16, we show that this cryptic pocket also opens in SARS-CoV1 and MERS but not in human CMTr1. Therefore, it may be possible to develop pan-coronavirus antivirals that target this cryptic pocket.
Collapse
Affiliation(s)
- Neha Vithani
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri; Center for Science and Engineering of Living Systems, Washington University in St. Louis, St. Louis, Missouri
| | - Michael D Ward
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri; Center for Science and Engineering of Living Systems, Washington University in St. Louis, St. Louis, Missouri
| | - Maxwell I Zimmerman
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri; Center for Science and Engineering of Living Systems, Washington University in St. Louis, St. Louis, Missouri
| | - Borna Novak
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri; Center for Science and Engineering of Living Systems, Washington University in St. Louis, St. Louis, Missouri; Medical Scientist Training Program, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Jonathan H Borowsky
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri; Center for Science and Engineering of Living Systems, Washington University in St. Louis, St. Louis, Missouri
| | - Sukrit Singh
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri; Center for Science and Engineering of Living Systems, Washington University in St. Louis, St. Louis, Missouri
| | - Gregory R Bowman
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri; Center for Science and Engineering of Living Systems, Washington University in St. Louis, St. Louis, Missouri.
| |
Collapse
|
305
|
Kesharwani K, Singh R, Khan MJ, Vinayak V, Joshi KB. Hydrophobized Short Peptide Amphiphile Functionalized Gold Nanoparticles as Antibacterial Biomaterials. ChemistrySelect 2021. [DOI: 10.1002/slct.202102204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Khushboo Kesharwani
- Department of Chemistry School of Chemical Science and Technology Dr.Harisingh Gour Vishwavidyalaya (A Central University) Sagar MP 470003 India
| | - Ramesh Singh
- Department of Chemistry School of Chemical Science and Technology Dr.Harisingh Gour Vishwavidyalaya (A Central University) Sagar MP 470003 India
| | - Mohd Jahir Khan
- Diatom Nanoengineering and metabolism lab (DNM) School of Applied Sciences Department of Criminology and Forensic Science Dr. Harisingh Gour Vishwavidyalaya (A Central University) Sagar MP
| | - Vandana Vinayak
- Diatom Nanoengineering and metabolism lab (DNM) School of Applied Sciences Department of Criminology and Forensic Science Dr. Harisingh Gour Vishwavidyalaya (A Central University) Sagar MP
| | - Khashti Ballabh Joshi
- Department of Chemistry School of Chemical Science and Technology Dr.Harisingh Gour Vishwavidyalaya (A Central University) Sagar MP 470003 India
| |
Collapse
|
306
|
Mazzoccanti G, Manetto S, Bassan M, Macis M, Iazzetti A, Cabri W, Ricci A, Gasparrini F. Expanding the Use of Dynamic Electrostatic Repulsion Reversed-Phase Chromatography: An Effective Elution Mode for Peptides Control and Analysis. Molecules 2021; 26:4348. [PMID: 34299626 PMCID: PMC8303375 DOI: 10.3390/molecules26144348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/16/2021] [Accepted: 07/16/2021] [Indexed: 11/17/2022] Open
Abstract
Bioactive peptides are increasingly used in clinical practice. Reversed-phase chromatography using formic or trifluoroacetic acid in the mobile phase is the most widely used technique for their analytical control. However, sometimes it does not prove sufficient to solve challenging chromatographic problems. In the search for alternative elution modes, the dynamic electrostatic repulsion reversed-phase was evaluated to separate eight probe peptides characterised by different molecular weights and isoelectric points. This technique, which involves TBAHSO4 in the mobile phase, provided the lowest asymmetry and peak width at half height values and the highest in peak capacity (about 200 for a gradient of 30 min) and resolution concerning the classic reversed-phase. All analyses were performed using cutting-edge columns developed for peptide separation, and the comparison of the chromatograms obtained shows how the dynamic electrostatic repulsion reversed-phase is an attractive alternative to the classic reversed-phase.
Collapse
Affiliation(s)
- Giulia Mazzoccanti
- Department of Drug Chemistry and Technology, “Sapienza” University of Rome, 00185 Rome, Italy; (S.M.); (F.G.)
| | - Simone Manetto
- Department of Drug Chemistry and Technology, “Sapienza” University of Rome, 00185 Rome, Italy; (S.M.); (F.G.)
| | - Michele Bassan
- Fresenius Kabi iPSUM, Piazza Maestri del Lavoro 7, 20063 Cernusco sul Naviglio, Italy; (M.B.); (M.M.)
| | - Marco Macis
- Fresenius Kabi iPSUM, Piazza Maestri del Lavoro 7, 20063 Cernusco sul Naviglio, Italy; (M.B.); (M.M.)
| | - Antonia Iazzetti
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy;
| | - Walter Cabri
- Department of Chemistry, Alma Mater Studiorum-University of Bologna, Via Selmi 2, 40126 Bologna, Italy;
| | - Antonio Ricci
- Fresenius Kabi iPSUM, Piazza Maestri del Lavoro 7, 20063 Cernusco sul Naviglio, Italy; (M.B.); (M.M.)
| | - Francesco Gasparrini
- Department of Drug Chemistry and Technology, “Sapienza” University of Rome, 00185 Rome, Italy; (S.M.); (F.G.)
| |
Collapse
|
307
|
Zhou J, Li Y, Huang W, Shi W, Qian H. Source and exploration of the peptides used to construct peptide-drug conjugates. Eur J Med Chem 2021; 224:113712. [PMID: 34303870 DOI: 10.1016/j.ejmech.2021.113712] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 07/12/2021] [Accepted: 07/17/2021] [Indexed: 12/16/2022]
Abstract
Peptide-drug conjugates (PDCs) are a class of novel molecules widely designed and synthesized for delivering payload drugs. The peptide part plays a vital role in the whole molecule, because they determine the ability of the molecules to penetrate the membrane and target to the specific targets. Here, we introduce the source of different kinds of cell-penetrating peptides (CPPs) and cell-targeting peptides (CTPs) that have been used or could be used in constructing PDCs as well as their latest application in delivering drugs. What's more, the approaches of developing CPPs and CTPs and the techniques to discover novel peptides are focused on and summarized in the review. This review aims to help relevant researchers fast understand the research status of peptides in PDCs and carry forward the process of novel peptides discovery.
Collapse
Affiliation(s)
- Jiaqi Zhou
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Yuanyuan Li
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Wenlong Huang
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Wei Shi
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China.
| | - Hai Qian
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China.
| |
Collapse
|
308
|
Leonard DJ, Zieleniewski F, Wellhöfer I, Baker EG, Ward JW, Woolfson DN, Clayden J. Scalable synthesis and coupling of quaternary α-arylated amino acids: α-aryl substituents are tolerated in α-helical peptides. Chem Sci 2021; 12:9386-9390. [PMID: 34349911 PMCID: PMC8278958 DOI: 10.1039/d1sc01378e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/08/2021] [Indexed: 11/21/2022] Open
Abstract
Quaternary amino acids are important tools for the modification and stabilisation of peptide secondary structures. Here we describe a practical and scalable synthesis applicable to quaternary alpha-arylated amino acids (Q4As), and the development of solid-phase synthesis conditions for their incorporation into peptides. Monomeric and dimeric α-helical peptides are synthesised with varying degrees of Q4A substitution and their structures examined using biophysical methods. Both enantiomers of the Q4As are tolerated in folded monomeric and oligomeric α-helical peptides, with the (R)-enantiomer slightly more so than the (S).
Collapse
Affiliation(s)
- Daniel J Leonard
- School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
| | | | - Isabelle Wellhöfer
- School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
| | - Emily G Baker
- School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
- School of Biochemistry, University of Bristol, Medical Sciences Building, University Walk Bristol BS8 1TS UK
- Bristol BioDesign Institute, University of Bristol, Life Sciences Building Tyndall Avenue Bristol BS8 1TQ UK
| | - John W Ward
- School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
| | - Derek N Woolfson
- School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
- School of Biochemistry, University of Bristol, Medical Sciences Building, University Walk Bristol BS8 1TS UK
- Bristol BioDesign Institute, University of Bristol, Life Sciences Building Tyndall Avenue Bristol BS8 1TQ UK
| | - Jonathan Clayden
- School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
| |
Collapse
|
309
|
Sompol P, Gollihue JL, Kraner SD, Artiushin IA, Cloyd RA, Chishti EA, Koren SA, Nation GK, Abisambra JF, Huzian O, Nagy LI, Santha M, Hackler L, Puskas LG, Norris CM. Q134R: Small chemical compound with NFAT inhibitory properties improves behavioral performance and synapse function in mouse models of amyloid pathology. Aging Cell 2021; 20:e13416. [PMID: 34117818 PMCID: PMC8282246 DOI: 10.1111/acel.13416] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 03/30/2021] [Accepted: 05/19/2021] [Indexed: 12/27/2022] Open
Abstract
Inhibition of the protein phosphatase calcineurin (CN) ameliorates pathophysiologic and cognitive changes in aging rodents and mice with aging-related Alzheimer's disease (AD)-like pathology. However, concerns over adverse effects have slowed the transition of common CN-inhibiting drugs to the clinic for the treatment of AD and AD-related disorders. Targeting substrates of CN, like the nuclear factor of activated T cells (NFATs), has been suggested as an alternative, safer approach to CN inhibitors. However, small chemical inhibitors of NFATs have only rarely been described. Here, we investigate a newly developed neuroprotective hydroxyquinoline derivative (Q134R) that suppresses NFAT signaling, without inhibiting CN activity. Q134R partially inhibited NFAT activity in primary rat astrocytes, but did not prevent CN-mediated dephosphorylation of a non-NFAT target, either in vivo, or in vitro. Acute (≤1 week) oral delivery of Q134R to APP/PS1 (12 months old) or wild-type mice (3-4 months old) infused with oligomeric Aβ peptides led to improved Y maze performance. Chronic (≥3 months) oral delivery of Q134R appeared to be safe, and, in fact, promoted survival in wild-type (WT) mice when given for many months beyond middle age. Finally, chronic delivery of Q134R to APP/PS1 mice during the early stages of amyloid pathology (i.e., between 6 and 9 months) tended to reduce signs of glial reactivity, prevented the upregulation of astrocytic NFAT4, and ameliorated deficits in synaptic strength and plasticity, without noticeably altering parenchymal Aβ plaque pathology. The results suggest that Q134R is a promising drug for treating AD and aging-related disorders.
Collapse
Affiliation(s)
- Pradoldej Sompol
- Sanders‐Brown Center on Aging University of Kentucky College of Medicine Lexington KY USA
| | - Jenna L. Gollihue
- Sanders‐Brown Center on Aging University of Kentucky College of Medicine Lexington KY USA
| | - Susan D. Kraner
- Sanders‐Brown Center on Aging University of Kentucky College of Medicine Lexington KY USA
| | - Irina A. Artiushin
- Sanders‐Brown Center on Aging University of Kentucky College of Medicine Lexington KY USA
| | - Ryan A. Cloyd
- Sanders‐Brown Center on Aging University of Kentucky College of Medicine Lexington KY USA
| | - Emad A. Chishti
- Sanders‐Brown Center on Aging University of Kentucky College of Medicine Lexington KY USA
| | - Shon A. Koren
- Sanders‐Brown Center on Aging University of Kentucky College of Medicine Lexington KY USA
| | - Grant K. Nation
- Sanders‐Brown Center on Aging University of Kentucky College of Medicine Lexington KY USA
| | - Jose F. Abisambra
- Center for Translational Research in Neurodegenerative Disease University of Florida Gainesville FL USA
| | | | | | | | | | | | - Christopher M. Norris
- Sanders‐Brown Center on Aging University of Kentucky College of Medicine Lexington KY USA
| |
Collapse
|
310
|
Lai X, Tang J, ElSayed MEH. Recent advances in proteolytic stability for peptide, protein, and antibody drug discovery. Expert Opin Drug Discov 2021; 16:1467-1482. [PMID: 34187273 DOI: 10.1080/17460441.2021.1942837] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: To discover and develop a peptide, protein, or antibody into a drug requires overcoming multiple challenges to obtain desired properties. Proteolytic stability is one of the challenges and deserves a focused investigation.Areas covered: This review concentrates on improving proteolytic stability by engineering the amino acids around the cleavage sites of a liable peptide, protein, or antibody. Peptidases are discussed on three levels including all peptidases in databases, mixtures based on organ and tissue types, and individual peptidases. The technique to identify cleavage sites is spotlighted on mass spectrometry-based approaches such as MALDI-TOF and LC-MS. For sequence engineering, the replacements that have been commonly applied with a higher chance of success are highlighted at the beginning, while the rarely used and more complicated replacements are discussed later. Although a one-size-fits-all approach does not exist to apply to different projects, this review provides a 3-step strategy for effectively and efficiently conducting the proteolytic stability experiments to achieve the eventual goal of improving the stability by engineering the molecule itself.Expert opinion: Improving the proteolytic stability is a spiraling up process sequenced by testing and engineering. There are many ways to engineer amino acids, but the choice must consider the cost and properties affected by the changes of the amino acids.
Collapse
Affiliation(s)
- Xianyin Lai
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Jason Tang
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Mohamed E H ElSayed
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
311
|
Stanzione F, Giangreco I, Cole JC. Use of molecular docking computational tools in drug discovery. PROGRESS IN MEDICINAL CHEMISTRY 2021; 60:273-343. [PMID: 34147204 DOI: 10.1016/bs.pmch.2021.01.004] [Citation(s) in RCA: 204] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Molecular docking has become an important component of the drug discovery process. Since first being developed in the 1980s, advancements in the power of computer hardware and the increasing number of and ease of access to small molecule and protein structures have contributed to the development of improved methods, making docking more popular in both industrial and academic settings. Over the years, the modalities by which docking is used to assist the different tasks of drug discovery have changed. Although initially developed and used as a standalone method, docking is now mostly employed in combination with other computational approaches within integrated workflows. Despite its invaluable contribution to the drug discovery process, molecular docking is still far from perfect. In this chapter we will provide an introduction to molecular docking and to the different docking procedures with a focus on several considerations and protocols, including protonation states, active site waters and consensus, that can greatly improve the docking results.
Collapse
Affiliation(s)
| | - Ilenia Giangreco
- Cambridge Crystallographic Data Centre, Cambridge, United Kingdom
| | - Jason C Cole
- Cambridge Crystallographic Data Centre, Cambridge, United Kingdom
| |
Collapse
|
312
|
Zolotarev YA, Mitkevich VA, Shram SI, Adzhubei AA, Tolstova AP, Talibov OB, Dadayan AK, Myasoyedov NF, Makarov AA, Kozin SA. Pharmacokinetics and Molecular Modeling Indicate nAChRα4-Derived Peptide HAEE Goes through the Blood-Brain Barrier. Biomolecules 2021; 11:biom11060909. [PMID: 34207317 PMCID: PMC8234734 DOI: 10.3390/biom11060909] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 12/24/2022] Open
Abstract
One of the treatment strategies for Alzheimer's disease (AD) is based on the use of pharmacological agents capable of binding to beta-amyloid (Aβ) and blocking its aggregation in the brain. Previously, we found that intravenous administration of the synthetic tetrapeptide Acetyl-His-Ala-Glu-Glu-Amide (HAEE), which is an analogue of the 35-38 region of the α4 subunit of α4β2 nicotinic acetylcholine receptor and specifically binds to the 11-14 site of Aβ, reduced the development of cerebral amyloidogenesis in a mouse model of AD. In the current study on three types of laboratory animals, we determined the biodistribution and tissue localization patterns of HAEE peptide after single intravenous bolus administration. The pharmacokinetic parameters of HAEE were established using uniformly tritium-labeled HAEE. Pharmacokinetic data provided evidence that HAEE goes through the blood-brain barrier. Based on molecular modeling, a role of LRP1 in receptor-mediated transcytosis of HAEE was proposed. Altogether, the results obtained indicate that the anti-amyloid effect of HAEE, previously found in a mouse model of AD, most likely occurs due to its interaction with Aβ species directly in the brain.
Collapse
Affiliation(s)
- Yurii A. Zolotarev
- Laboratory of Protein Conformational Polymorphism in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.A.Z.); (V.A.M.); (A.A.A.); (A.P.T.); (A.A.M.)
- Department of Physiologically Active Substances Chemistry, Institute of Molecular Genetics of National Research Center «Kurchatov Institute», 123182 Moscow, Russia; (S.I.S.); (A.K.D.); (N.F.M.)
| | - Vladimir A. Mitkevich
- Laboratory of Protein Conformational Polymorphism in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.A.Z.); (V.A.M.); (A.A.A.); (A.P.T.); (A.A.M.)
| | - Stanislav I. Shram
- Department of Physiologically Active Substances Chemistry, Institute of Molecular Genetics of National Research Center «Kurchatov Institute», 123182 Moscow, Russia; (S.I.S.); (A.K.D.); (N.F.M.)
| | - Alexei A. Adzhubei
- Laboratory of Protein Conformational Polymorphism in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.A.Z.); (V.A.M.); (A.A.A.); (A.P.T.); (A.A.M.)
| | - Anna P. Tolstova
- Laboratory of Protein Conformational Polymorphism in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.A.Z.); (V.A.M.); (A.A.A.); (A.P.T.); (A.A.M.)
| | - Oleg B. Talibov
- Department of Clinical Pharmacology, Faculty of Common Medicine, Evdokimov Moscow State University of Medicine and Dentistry, 127473 Moscow, Russia;
| | - Alexander K. Dadayan
- Department of Physiologically Active Substances Chemistry, Institute of Molecular Genetics of National Research Center «Kurchatov Institute», 123182 Moscow, Russia; (S.I.S.); (A.K.D.); (N.F.M.)
| | - Nikolai F. Myasoyedov
- Department of Physiologically Active Substances Chemistry, Institute of Molecular Genetics of National Research Center «Kurchatov Institute», 123182 Moscow, Russia; (S.I.S.); (A.K.D.); (N.F.M.)
| | - Alexander A. Makarov
- Laboratory of Protein Conformational Polymorphism in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.A.Z.); (V.A.M.); (A.A.A.); (A.P.T.); (A.A.M.)
| | - Sergey A. Kozin
- Laboratory of Protein Conformational Polymorphism in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.A.Z.); (V.A.M.); (A.A.A.); (A.P.T.); (A.A.M.)
- Correspondence: ; Tel.: +7-499-135-98-24
| |
Collapse
|
313
|
Sokullu E, Gauthier MS, Coulombe B. Discovery of Antivirals Using Phage Display. Viruses 2021; 13:v13061120. [PMID: 34200959 PMCID: PMC8230593 DOI: 10.3390/v13061120] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023] Open
Abstract
The latest coronavirus disease outbreak, COVID-19, has brought attention to viral infections which have posed serious health threats to humankind throughout history. The rapid global spread of COVID-19 is attributed to the increased human mobility of today's world, yet the threat of viral infections to global public health is expected to increase continuously in part due to increasing human-animal interface. Development of antiviral agents is crucial to combat both existing and novel viral infections. Recently, there is a growing interest in peptide/protein-based drug molecules. Antibodies are becoming especially predominant in the drug market. Indeed, in a remarkably short period, four antibody therapeutics were authorized for emergency use in COVID-19 treatment in the US, Russia, and India as of November 2020. Phage display has been one of the most widely used screening methods for peptide/antibody drug discovery. Several phage display-derived biologics are already in the market, and the expiration of intellectual property rights of phage-display antibody discovery platforms suggests an increment in antibody drugs in the near future. This review summarizes the most common phage display libraries used in antiviral discovery, highlights the approaches employed to enhance the antiviral potency of selected peptides/antibody fragments, and finally provides a discussion about the present status of the developed antivirals in clinic.
Collapse
Affiliation(s)
- Esen Sokullu
- Department of Translational Proteomics, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada;
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada
- Correspondence: (E.S.); (B.C.)
| | - Marie-Soleil Gauthier
- Department of Translational Proteomics, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada;
| | - Benoit Coulombe
- Department of Translational Proteomics, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada;
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada
- Correspondence: (E.S.); (B.C.)
| |
Collapse
|
314
|
Sahoo A, Swain SS, Behera A, Sahoo G, Mahapatra PK, Panda SK. Antimicrobial Peptides Derived From Insects Offer a Novel Therapeutic Option to Combat Biofilm: A Review. Front Microbiol 2021; 12:661195. [PMID: 34248873 PMCID: PMC8265172 DOI: 10.3389/fmicb.2021.661195] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/12/2021] [Indexed: 12/20/2022] Open
Abstract
Biofilms form a complex layer with defined structures, that attach on biotic or abiotic surfaces, are tough to eradicate and tend to cause some resistance against most antibiotics. Several studies confirmed that biofilm-producing bacteria exhibit higher resistance compared to the planktonic form of the same species. Antibiotic resistance factors are well understood in planktonic bacteria which is not so in case of biofilm producing forms. This may be due to the lack of available drugs with known resistance mechanisms for biofilms. Existing antibiotics cannot eradicate most biofilms, especially of ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species). Insects produce complex and diverse set of chemicals for survival and defense. Antimicrobial peptides (AMPs), produced by most insects, generally have a broad spectrum of activity and the potential to bypass the resistance mechanisms of classical antibiotics. Besides, AMPs may well act synergistically with classical antibiotics for a double-pronged attack on infections. Thus, AMPs could be promising alternatives to overcome medically important biofilms, decrease the possibility of acquired resistance and treatment of multidrug-resistant pathogens including ESKAPE. The present review focuses on insect-derived AMPs with special reference to anti-biofilm-based strategies. It covers the AMP composition, pathways and mechanisms of action, the formation of biofilms, impact of biofilms on human diseases, current strategies as well as therapeutic options to combat biofilm with antimicrobial peptides from insects. In addition, the review also illustrates the importance of bioinformatics tools and molecular docking studies to boost the importance of select bioactive peptides those can be developed as drugs, as well as suggestions for further basic and clinical research.
Collapse
Affiliation(s)
- Alaka Sahoo
- Department of Skin & VD, Institute of Medical Sciences, SUM Hospital, Siksha O Anusandhan University, Bhubaneswar, India
| | - Shasank Sekhar Swain
- Division of Microbiology & NCDs, ICMR-Regional Medical Research Centre, Bhubaneswar, India
| | - Ayusman Behera
- Department of Zoology, Maharaja Sriram Chandra Bhanja Deo University, Baripada, India
| | - Gunanidhi Sahoo
- Department of Zoology, Utkal University, Vani Vihar, Bhubaneswar, India
| | | | - Sujogya Kumar Panda
- Centre of Environment, Climate Change and Public Health, RUSA 2.0, Utkal University, Vani Vihar, Bhubaneswar, India
| |
Collapse
|
315
|
Kerry RG, Mahapatra GP, Maurya GK, Patra S, Mahari S, Das G, Patra JK, Sahoo S. Molecular prospect of type-2 diabetes: Nanotechnology based diagnostics and therapeutic intervention. Rev Endocr Metab Disord 2021; 22:421-451. [PMID: 33052523 DOI: 10.1007/s11154-020-09606-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/08/2020] [Indexed: 02/08/2023]
Abstract
About ninety percent of all diabetic conditions account for T2D caused due to abnormal insulin secretion/ action or increased hepatic glucose production. Factors that contribute towards the aetiology of T2D could be well explained through biochemical, molecular, and cellular aspects. In this review, we attempt to explain the recent evolving molecular and cellular advancement associated with T2D pathophysiology. Current progress fabricated in T2D research concerning intracellular signaling cascade, inflammasome, autophagy, genetic and epigenetics changes is discretely explained in simple terms. Present available anti-diabetic therapeutic strategies commercialized and their limitations which are needed to be acknowledged are addressed in the current review. In particular, the pre-eminence of nanotechnology-based approaches to nullify the inadequacy of conventional anti-diabetic therapeutics and heterogeneous nanoparticulated systems exploited in diabetic researches are also discretely mentioned and are also listed in a tabular format in the review. Additionally, as a future prospect of nanotechnology, the review presents several strategic hypotheses to ameliorate the austerity of T2D by an engineered smart targeted nano-delivery system. In detail, an effort has been made to hypothesize novel nanotechnological based therapeutic strategies, which exploits previously described inflammasome, autophagic target points. Utilizing graphical description it is explained how a smart targeted nano-delivery system could promote β-cell growth and development by inducing the Wnt signaling pathway (inhibiting Gsk3β), inhibiting inflammasome (inhibiting NLRP3), and activating autophagic target points (protecting Atg3/Atg7 complex from oxidative stress) thereby might ameliorate the severity of T2D. Additionally, several targeting molecules associated with autophagic and epigenetic factors are also highlighted, which can be exploited in future diabetic research.
Collapse
Affiliation(s)
- Rout George Kerry
- Department of Biotechnology, Utkal University, Vani Vihar, Bhubaneswar, Odisha, 751004, India
| | | | - Ganesh Kumar Maurya
- Zoology Section, Mahila MahaVidyalya, Banaras Hindu University, Varanasi, 221005, India
| | - Sushmita Patra
- Department of Biotechnology, North Odissa University, Takatpur, Baripada, Odisha, 757003, India
| | - Subhasis Mahari
- DBT- National Institute of Animal Biotechnology, Hyderabad, 500032, India
| | - Gitishree Das
- Research Institute of Biotechnology & Medical Converged Science, Dongguk University-Seoul, Goyangsi, 10326, Republic of Korea
| | - Jayanta Kumar Patra
- Research Institute of Biotechnology & Medical Converged Science, Dongguk University-Seoul, Goyangsi, 10326, Republic of Korea.
| | - Sabuj Sahoo
- Department of Biotechnology, Utkal University, Vani Vihar, Bhubaneswar, Odisha, 751004, India.
| |
Collapse
|
316
|
Fioravanti G, Hua PQ, Tomlinson RE. The TrkA agonist gambogic amide augments skeletal adaptation to mechanical loading. Bone 2021; 147:115908. [PMID: 33713848 PMCID: PMC8097708 DOI: 10.1016/j.bone.2021.115908] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 10/21/2022]
Abstract
The periosteal and endosteal surfaces of mature bone are densely innervated by sensory nerves expressing TrkA, the high-affinity receptor for nerve growth factor (NGF). In previous work, we demonstrated that administration of exogenous NGF significantly increased load-induced bone formation through the activation of Wnt signaling. However, the translational potential of NGF is limited by the induction of substantial mechanical and thermal hyperalgesia in mice and humans. Here, we tested the effect of gambogic amide (GA), a recently identified robust small molecule agonist for TrkA, on hyperalgesia and load-induced bone formation. Behavioral analysis was used to assess pain up to one week after axial forelimb compression. Contrary to our expectations, GA treatment was not associated with diminished use of the loaded forelimb or sensitivity to thermal stimulus. Furthermore, dynamic histomorphometry revealed a significant increase in relative periosteal bone formation rate as compared to vehicle treatment. Additionally, we found that GA treatment was associated with an increase in the number of osteoblasts per bone surface in loaded limbs as well as a significant increase in the fold change of Ngf, Wnt7b, and Axin2 mRNA expression as compared to vehicle (control). To test the effect of GA on osteoblasts directly, we cultured MC3T3-E1 cells for up to 21 days in osteogenic differentiation media containing NGF, GA, or vehicle (control). Media containing GA induced the significant upregulation of the osteoblastic differentiation markers Runx2, Bglap2, and Sp7 in a dose-dependent manner, whereas treatment with NGF was not associated with any significant increases in these markers. Furthermore, consistent with our in vivo findings, we observed that administration of 50 nM of GA upregulated expression of Ngf at both Day 3 and Day 7. However, cells treated with the highest dose of GA (500 nM) had significantly increased apoptosis and impaired cell proliferation. In conclusion, our study indicates GA may be useful for augmenting skeletal adaptation to mechanical forces without inducing hyperalgesia.
Collapse
Affiliation(s)
- Gabriella Fioravanti
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Phuong Q Hua
- Department of Biomedical Engineering, Drexel University, Philadelphia, PA, United States of America
| | - Ryan E Tomlinson
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA, United States of America.
| |
Collapse
|
317
|
Rodríguez-Carlos A, Jacobo-Delgado YM, Santos-Mena AO, Rivas-Santiago B. Modulation of cathelicidin and defensins by histone deacetylase inhibitors: A potential treatment for multi-drug resistant infectious diseases. Peptides 2021; 140:170527. [PMID: 33744370 DOI: 10.1016/j.peptides.2021.170527] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/26/2021] [Accepted: 03/09/2021] [Indexed: 12/12/2022]
Abstract
Infectious diseases are an important growing public health problem, which perspective has worsened due to the increasing number of drug-resistant strains in the last few years. Although diverse solutions have been proposed, one viable solution could be the use of immune system modulators. The induction of the immune response can be increased by histone deacetylase inhibitors (iHDAC), which in turn modulate the chromatin and increase the activation of different cellular pathways and nuclear factors such as STAT3, HIF-1α NF-kB, C/EBPα and, AP-1. These pathways are capable to promote several immune response-related molecules including those with antimicrobial properties such as antimicrobial peptides (AMPs) that lead to the elimination of pathogens including multi drug-resistant strains.
Collapse
Affiliation(s)
- Adrián Rodríguez-Carlos
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico
| | | | - Alan O Santos-Mena
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico
| | - Bruno Rivas-Santiago
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico.
| |
Collapse
|
318
|
Dahlgren D, Olander T, Sjöblom M, Hedeland M, Lennernäs H. Effect of paracellular permeation enhancers on intestinal permeability of two peptide drugs, enalaprilat and hexarelin, in rats. Acta Pharm Sin B 2021; 11:1667-1675. [PMID: 34221875 PMCID: PMC8245904 DOI: 10.1016/j.apsb.2020.12.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/05/2020] [Accepted: 10/27/2020] [Indexed: 12/29/2022] Open
Abstract
Transcellular permeation enhancers are known to increase the intestinal permeability of enalaprilat, a 349 Da peptide, but not hexarelin (887 Da). The primary aim of this paper was to investigate if paracellular permeability enhancers affected the intestinal permeation of the two peptides. This was investigated using the rat single-pass intestinal perfusion model with concomitant blood sampling. These luminal compositions included two paracellular permeation enhancers, chitosan (5 mg/mL) and ethylenediaminetetraacetate (EDTA, 1 and 5 mg/mL), as well as low luminal tonicity (100 mOsm) with or without lidocaine. Effects were evaluated by the change in lumen-to-blood permeability of hexarelin and enalaprilat, and the blood-to-lumen clearance of 51chromium-labeled EDTA (CLCr-EDTA), a clinical marker for mucosal barrier integrity. The two paracellular permeation enhancers increased the mucosal permeability of both peptide drugs to a similar extent. The data in this study suggests that the potential for paracellular permeability enhancers to increase intestinal absorption of hydrophilic peptides with low molecular mass is greater than for those with transcellular mechanism-of-action. Further, the mucosal blood-to-lumen flux of 51Cr-EDTA was increased by the two paracellular permeation enhancers and by luminal hypotonicity. In contrast, luminal hypotonicity did not affect the lumen-to-blood transport of enalaprilat and hexarelin. This suggests that hypotonicity affects paracellular solute transport primarily in the mucosal crypt region, as this area is protected from luminal contents by a constant water flow from the crypts.
Collapse
Affiliation(s)
- David Dahlgren
- Department of Pharmaceutical Biosciences, Translational Drug Discovery and Development, Uppsala University, Uppsala 752 36, Sweden
| | - Tobias Olander
- Department of Pharmaceutical Biosciences, Translational Drug Discovery and Development, Uppsala University, Uppsala 752 36, Sweden
| | - Markus Sjöblom
- Department of Neuroscience, Division of Physiology, Uppsala University, Uppsala 752 36, Sweden
| | - Mikael Hedeland
- Department of Medicinal Chemistry, Analytical Pharmaceutical Chemistry, Uppsala University, Uppsala 752 36, Sweden
- Department of Chemistry, Environment and Feed Hygiene, National Veterinary Institute (SVA), Uppsala 751 89, Sweden
| | - Hans Lennernäs
- Department of Pharmaceutical Biosciences, Translational Drug Discovery and Development, Uppsala University, Uppsala 752 36, Sweden
| |
Collapse
|
319
|
Improvement on Permeability of Cyclic Peptide/Peptidomimetic: Backbone N-Methylation as A Useful Tool. Mar Drugs 2021; 19:md19060311. [PMID: 34072121 PMCID: PMC8229464 DOI: 10.3390/md19060311] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 12/14/2022] Open
Abstract
Peptides have a three-dimensional configuration that can adopt particular conformations for binding to proteins, which are well suited to interact with larger contact surface areas on target proteins. However, low cell permeability is a major challenge in the development of peptide-related drugs. In recent years, backbone N-methylation has been a useful tool for manipulating the permeability of cyclic peptides/peptidomimetics. Backbone N-methylation permits the adjustment of molecule’s conformational space. Several pathways are involved in the drug absorption pathway; the relative importance of each N-methylation to total permeation is likely to differ with intrinsic properties of cyclic peptide/peptidomimetic. Recent studies on the permeability of cyclic peptides/peptidomimetics using the backbone N-methylation strategy and synthetic methodologies will be presented in this review.
Collapse
|
320
|
Wilson CJ, Chang M, Karttunen M, Choy WY. KEAP1 Cancer Mutants: A Large-Scale Molecular Dynamics Study of Protein Stability. Int J Mol Sci 2021; 22:5408. [PMID: 34065616 PMCID: PMC8161161 DOI: 10.3390/ijms22105408] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/30/2022] Open
Abstract
We have performed 280 μs of unbiased molecular dynamics (MD) simulations to investigate the effects of 12 different cancer mutations on Kelch-like ECH-associated protein 1 (KEAP1) (G333C, G350S, G364C, G379D, R413L, R415G, A427V, G430C, R470C, R470H, R470S and G476R), one of the frequently mutated proteins in lung cancer. The aim was to provide structural insight into the effects of these mutants, including a new class of ANCHOR (additionally NRF2-complexed hypomorph) mutant variants. Our work provides additional insight into the structural dynamics of mutants that could not be analyzed experimentally, painting a more complete picture of their mutagenic effects. Notably, blade-wise analysis of the Kelch domain points to stability as a possible target of cancer in KEAP1. Interestingly, structural analysis of the R470C ANCHOR mutant, the most prevalent missense mutation in KEAP1, revealed no significant change in structural stability or NRF2 binding site dynamics, possibly indicating an covalent modification as this mutant's mode of action.
Collapse
Affiliation(s)
- Carter J. Wilson
- Department of Biochemistry, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 5C1, Canada; (C.J.W.); (M.C.)
- Department of Applied Mathematics, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 5B7, Canada
| | - Megan Chang
- Department of Biochemistry, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 5C1, Canada; (C.J.W.); (M.C.)
| | - Mikko Karttunen
- Department of Applied Mathematics, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 5B7, Canada
- Department of Chemistry, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 3K7, Canada
- Centre for Advanced Materials and Biomaterials Research, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 5B7, Canada
| | - Wing-Yiu Choy
- Department of Biochemistry, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 5C1, Canada; (C.J.W.); (M.C.)
| |
Collapse
|
321
|
Lukinović V, Biggar KK. Deconvoluting complex protein interaction networks through reductionist strategies in peptide biochemistry: Modern approaches and research questions. Comp Biochem Physiol B Biochem Mol Biol 2021; 256:110616. [PMID: 34000427 DOI: 10.1016/j.cbpb.2021.110616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/06/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022]
Abstract
Following the decoding of the first human genome, researchers have vastly improved their understanding of cell biology and its regulation. As a result, it has become clear that it is not merely genetic information, but the aberrant changes in the functionality and connectivity of its encoded proteins that drive cell response to periods of stress and external cues. Therefore, proper utilization of refined methods that help to describe protein signalling or regulatory networks (i.e., functional connectivity), can help us understand how change in the signalling landscape effects the cell. However, given the vast complexity in 'how and when' proteins communicate or interact with each other, it is extremely difficult to define, characterize, and understand these interaction networks in a tangible manner. Herein lies the challenge of tackling the functional proteome; its regulation is encoded in multiple layers of interaction, chemical modification and cell compartmentalization. To address and refine simple research questions, modern reductionist strategies in protein biochemistry have successfully used peptide-based experiments; their summation helping to simplify the overall complexity of these protein interaction networks. In this way, peptides are powerful tools used in fundamental research that can be readily applied to comparative biochemical research. Understanding and defining how proteins interact is one of the key aspects towards understanding how the proteome functions. To date, reductionist peptide-based research has helped to address a wide range of proteome-related research questions, including the prediction of enzymes substrates, identification of posttranslational modifications, and the annotation of protein interaction partners. Peptide arrays have been used to identify the binding specificity of reader domains, which are able to recognise the posttranslational modifications; forming dynamic protein interactions that are dependent on modification state. Finally, representing one of the fastest growing classes of inhibitor molecules, peptides are now begin explored as "disruptors" of protein-protein interactions or enzyme activity. Collectively, this review will discuss the use of peptides, peptide arrays, peptide-oriented computational biochemistry as modern reductionist strategies in deconvoluting the functional proteome.
Collapse
Affiliation(s)
- Valentina Lukinović
- Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| | - Kyle K Biggar
- Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada.
| |
Collapse
|
322
|
Wang J, Miao Y. Peptide Gaussian accelerated molecular dynamics (Pep-GaMD): Enhanced sampling and free energy and kinetics calculations of peptide binding. J Chem Phys 2021; 153:154109. [PMID: 33092378 DOI: 10.1063/5.0021399] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Peptides mediate up to 40% of known protein-protein interactions in higher eukaryotes and play an important role in cellular signaling. However, it is challenging to simulate both binding and unbinding of peptides and calculate peptide binding free energies through conventional molecular dynamics, due to long biological timescales and extremely high flexibility of the peptides. Based on the Gaussian accelerated molecular dynamics (GaMD) enhanced sampling technique, we have developed a new computational method "Pep-GaMD," which selectively boosts essential potential energy of the peptide in order to effectively model its high flexibility. In addition, another boost potential is applied to the remaining potential energy of the entire system in a dual-boost algorithm. Pep-GaMD has been demonstrated on binding of three model peptides to the SH3 domains. Independent 1 µs dual-boost Pep-GaMD simulations have captured repetitive peptide dissociation and binding events, which enable us to calculate peptide binding thermodynamics and kinetics. The calculated binding free energies and kinetic rate constants agreed very well with available experimental data. Furthermore, the all-atom Pep-GaMD simulations have provided important insights into the mechanism of peptide binding to proteins that involves long-range electrostatic interactions and mainly conformational selection. In summary, Pep-GaMD provides a highly efficient, easy-to-use approach for unconstrained enhanced sampling and calculations of peptide binding free energies and kinetics.
Collapse
Affiliation(s)
- Jinan Wang
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66047, USA
| | - Yinglong Miao
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66047, USA
| |
Collapse
|
323
|
Cao X, Tian P. "Dividing and Conquering" and "Caching" in Molecular Modeling. Int J Mol Sci 2021; 22:5053. [PMID: 34068835 PMCID: PMC8126232 DOI: 10.3390/ijms22095053] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 11/17/2022] Open
Abstract
Molecular modeling is widely utilized in subjects including but not limited to physics, chemistry, biology, materials science and engineering. Impressive progress has been made in development of theories, algorithms and software packages. To divide and conquer, and to cache intermediate results have been long standing principles in development of algorithms. Not surprisingly, most important methodological advancements in more than half century of molecular modeling are various implementations of these two fundamental principles. In the mainstream classical computational molecular science, tremendous efforts have been invested on two lines of algorithm development. The first is coarse graining, which is to represent multiple basic particles in higher resolution modeling as a single larger and softer particle in lower resolution counterpart, with resulting force fields of partial transferability at the expense of some information loss. The second is enhanced sampling, which realizes "dividing and conquering" and/or "caching" in configurational space with focus either on reaction coordinates and collective variables as in metadynamics and related algorithms, or on the transition matrix and state discretization as in Markov state models. For this line of algorithms, spatial resolution is maintained but results are not transferable. Deep learning has been utilized to realize more efficient and accurate ways of "dividing and conquering" and "caching" along these two lines of algorithmic research. We proposed and demonstrated the local free energy landscape approach, a new framework for classical computational molecular science. This framework is based on a third class of algorithm that facilitates molecular modeling through partially transferable in resolution "caching" of distributions for local clusters of molecular degrees of freedom. Differences, connections and potential interactions among these three algorithmic directions are discussed, with the hope to stimulate development of more elegant, efficient and reliable formulations and algorithms for "dividing and conquering" and "caching" in complex molecular systems.
Collapse
Affiliation(s)
- Xiaoyong Cao
- School of Life Sciences, Jilin University, Changchun 130012, China;
| | - Pu Tian
- School of Life Sciences, Jilin University, Changchun 130012, China;
- School of Artificial Intelligence, Jilin University, Changchun 130012, China
| |
Collapse
|
324
|
Marsh SR, Williams ZJ, Pridham KJ, Gourdie RG. Peptidic Connexin43 Therapeutics in Cardiac Reparative Medicine. J Cardiovasc Dev Dis 2021; 8:52. [PMID: 34063001 PMCID: PMC8147937 DOI: 10.3390/jcdd8050052] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/19/2021] [Accepted: 05/01/2021] [Indexed: 12/12/2022] Open
Abstract
Connexin (Cx43)-formed channels have been linked to cardiac arrhythmias and diseases of the heart associated with myocardial tissue loss and fibrosis. These pathologies include ischemic heart disease, ischemia-reperfusion injury, heart failure, hypertrophic cardiomyopathy, arrhythmogenic right ventricular cardiomyopathy, and Duchenne muscular dystrophy. A number of Cx43 mimetic peptides have been reported as therapeutic candidates for targeting disease processes linked to Cx43, including some that have advanced to clinical testing in humans. These peptides include Cx43 sequences based on the extracellular loop domains (e.g., Gap26, Gap 27, and Peptide5), cytoplasmic-loop domain (Gap19 and L2), and cytoplasmic carboxyl-terminal domain (e.g., JM2, Cx43tat, CycliCX, and the alphaCT family of peptides) of this transmembrane protein. Additionally, RYYN peptides binding to the Cx43 carboxyl-terminus have been described. In this review, we survey preclinical and clinical data available on short mimetic peptides based on, or directly targeting, Cx43, with focus on their potential for treating heart disease. We also discuss problems that have caused reluctance within the pharmaceutical industry to translate peptidic therapeutics to the clinic, even when supporting preclinical data is strong. These issues include those associated with the administration, stability in vivo, and tissue penetration of peptide-based therapeutics. Finally, we discuss novel drug delivery technologies including nanoparticles, exosomes, and other nanovesicular carriers that could transform the clinical and commercial viability of Cx43-targeting peptides in treatment of heart disease, stroke, cancer, and other indications requiring oral or parenteral administration. Some of these newly emerging approaches to drug delivery may provide a path to overcoming pitfalls associated with the drugging of peptide therapeutics.
Collapse
Affiliation(s)
- Spencer R. Marsh
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA 24016, USA; (S.R.M.); (Z.J.W.); (K.J.P.)
- Center for Heart and Reparative Medicine Research, Virginia Tech, Roanoke, VA 24016, USA
| | - Zachary J. Williams
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA 24016, USA; (S.R.M.); (Z.J.W.); (K.J.P.)
- Center for Heart and Reparative Medicine Research, Virginia Tech, Roanoke, VA 24016, USA
- Translational Biology Medicine and Health Graduate Program, Virginia Tech, Roanoke, VA 24016, USA
| | - Kevin J. Pridham
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA 24016, USA; (S.R.M.); (Z.J.W.); (K.J.P.)
- Center for Heart and Reparative Medicine Research, Virginia Tech, Roanoke, VA 24016, USA
| | - Robert G. Gourdie
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA 24016, USA; (S.R.M.); (Z.J.W.); (K.J.P.)
- Center for Heart and Reparative Medicine Research, Virginia Tech, Roanoke, VA 24016, USA
- Translational Biology Medicine and Health Graduate Program, Virginia Tech, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA
- Department of Emergency Medicine, Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, VA 24016, USA
| |
Collapse
|
325
|
Wang X, Ni D, Liu Y, Lu S. Rational Design of Peptide-Based Inhibitors Disrupting Protein-Protein Interactions. Front Chem 2021; 9:682675. [PMID: 34017824 PMCID: PMC8128998 DOI: 10.3389/fchem.2021.682675] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 04/09/2021] [Indexed: 12/12/2022] Open
Abstract
Protein-protein interactions (PPIs) are well-established as a class of promising drug targets for their implications in a wide range of biological processes. However, drug development toward PPIs is inevitably hampered by their flat and wide interfaces, which generally lack suitable pockets for ligand binding, rendering most PPI systems "undruggable." Here, we summarized drug design strategies for developing peptide-based PPI inhibitors. Importantly, several quintessential examples toward well-established PPI targets such as Bcl-2 family members, p53-MDM2, as well as APC-Asef are presented to illustrate the detailed schemes for peptide-based PPI inhibitor development and optimizations. This review supplies a comprehensive overview of recent progresses in drug discovery targeting PPIs through peptides or peptidomimetics, and will shed light on future therapeutic agent development toward the historically "intractable" PPI systems.
Collapse
Affiliation(s)
- Xuefei Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Duan Ni
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Yaqin Liu
- Medicinal Bioinformatics Center, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Shaoyong Lu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
- Medicinal Bioinformatics Center, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| |
Collapse
|
326
|
Arsenyan P, Lapcinska S. Straightforward Functionalization of Sulfur-Containing Peptides via 5- and 6-endo-dig Cyclization Reactions. SYNTHESIS-STUTTGART 2021. [DOI: 10.1055/a-1343-5607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
AbstractWe present a simple and convenient method for the generation of sulfenyl electrophiles from peptides containing S–S or S–H bonds by employing N-chlorosuccinimide. The corresponding sulfenyl electrophiles are further utilized in 5- and 6-endo-dig cyclization reactions yielding indolizinium salts, indoles, benzo[b]furans, polyaromatic hydrocarbons (PAHs) and isocoumarins, as well as quinolinones bearing a glutathione moiety. PAH derivatives can be used as selective fluorescent dyes for the visualization of lipid droplets in living cells.
Collapse
|
327
|
Hashemi ZS, Zarei M, Fath MK, Ganji M, Farahani MS, Afsharnouri F, Pourzardosht N, Khalesi B, Jahangiri A, Rahbar MR, Khalili S. In silico Approaches for the Design and Optimization of Interfering Peptides Against Protein-Protein Interactions. Front Mol Biosci 2021; 8:669431. [PMID: 33996914 PMCID: PMC8113820 DOI: 10.3389/fmolb.2021.669431] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/06/2021] [Indexed: 01/01/2023] Open
Abstract
Large contact surfaces of protein-protein interactions (PPIs) remain to be an ongoing issue in the discovery and design of small molecule modulators. Peptides are intrinsically capable of exploring larger surfaces, stable, and bioavailable, and therefore bear a high therapeutic value in the treatment of various diseases, including cancer, infectious diseases, and neurodegenerative diseases. Given these promising properties, a long way has been covered in the field of targeting PPIs via peptide design strategies. In silico tools have recently become an inevitable approach for the design and optimization of these interfering peptides. Various algorithms have been developed to scrutinize the PPI interfaces. Moreover, different databases and software tools have been created to predict the peptide structures and their interactions with target protein complexes. High-throughput screening of large peptide libraries against PPIs; "hotspot" identification; structure-based and off-structure approaches of peptide design; 3D peptide modeling; peptide optimization strategies like cyclization; and peptide binding energy evaluation are among the capabilities of in silico tools. In the present study, the most recent advances in the field of in silico approaches for the design of interfering peptides against PPIs will be reviewed. The future perspective of the field and its advantages and limitations will also be pinpointed.
Collapse
Affiliation(s)
- Zahra Sadat Hashemi
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research, Tehran, Iran
| | - Mahboubeh Zarei
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Mahmoud Ganji
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahboube Shahrabi Farahani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Afsharnouri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Navid Pourzardosht
- Cellular and Molecular Research Center, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Biochemistry, Guilan University of Medical Sciences, Rasht, Iran
| | - Bahman Khalesi
- Department of Research and Production of Poultry Viral Vaccine, Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization, Karaj, Iran
| | - Abolfazl Jahangiri
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Rahbar
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| |
Collapse
|
328
|
Cheung CHP, Xu J, Lee CL, Zhang Y, Wei R, Bierer D, Huang X, Li X. Construction of diverse peptide structural architectures via chemoselective peptide ligation. Chem Sci 2021; 12:7091-7097. [PMID: 34123337 PMCID: PMC8153220 DOI: 10.1039/d1sc01174j] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/13/2021] [Indexed: 12/22/2022] Open
Abstract
Herein, we report the development of a facile synthetic strategy for constructing diverse peptide structural architectures via chemoselective peptide ligation. The key advancement involved is to utilize the benzofuran moiety as the peptide salicylaldehyde ester surrogate, and Dap-Ser/Lys-Ser dipeptide as the hydroxyl amino functionality, which could be successfully introduced at the side chain of peptides enabling peptide ligation. With this method, the side chain-to-side chain cyclic peptide, branched/bridged peptides, tailed cyclic peptides and multi-cyclic peptides have been designed and successfully synthesized with native peptidic linkages at the ligation sites. This strategy has provided an alternative strategic opportunity for synthetic peptide development. It also serves as an inspiration for the structural design of PPI inhibitors with new modalities.
Collapse
Affiliation(s)
- Carina Hey Pui Cheung
- Department of Chemistry, State Key Lab of Synthetic Chemistry, The University of Hong Kong Hong Kong
| | - Jianchao Xu
- Department of Chemistry, State Key Lab of Synthetic Chemistry, The University of Hong Kong Hong Kong
| | - Chi Lung Lee
- Department of Chemistry, State Key Lab of Synthetic Chemistry, The University of Hong Kong Hong Kong
| | - Yanfeng Zhang
- Department of Chemistry, State Key Lab of Synthetic Chemistry, The University of Hong Kong Hong Kong
| | - Ruohan Wei
- Department of Chemistry, State Key Lab of Synthetic Chemistry, The University of Hong Kong Hong Kong
| | - Donald Bierer
- Department of Medicinal Chemistry, Bayer AG Aprather Weg 18A 42096 Wuppertal Germany
| | - Xuhui Huang
- Department of Biological and Chemical Engineering, The Hong Kong University of Science and Technology Clear Water Bay Kowloon Hong Kong
| | - Xuechen Li
- Department of Chemistry, State Key Lab of Synthetic Chemistry, The University of Hong Kong Hong Kong
| |
Collapse
|
329
|
Abstract
Cancer is the second leading cause of death worldwide, and the search for specialised therapy options has been a challenge for decades. The emergence of active targeted therapies provides the opportunity to treat cancerous tissues without harming healthy ones due to peculiar physiological changes. Herein, peptides and peptide analogs have been gaining a lot of attention over the last decade, especially for the on-site delivery of therapeutics to target tissues in order to achieve efficient and reliable cancer treatment. Combining peptides with highly efficient drug delivery platforms could potentially eliminate off-target adverse effects encountered during active targeting of conventional chemotherapeutics. Small size, ease of production and characterisation, low immunogenicity and satisfactory binding affinity of peptides offer some advantages over other complex targeting moiety, no wonder the market of peptide-based drugs continues to expand expeditiously. It is estimated that the global peptide drug market will be worth around USD 48.04 billion by 2025, with a compound annual growth rate of 9.4%. In this review, the current state of art of peptide-based therapeutics with special interest on tumour targeting peptides has been discussed. Moreover, various active targeting strategies such as the use functionalised peptides or peptide analogs are also elaborated.
Collapse
Affiliation(s)
- Selin Seda Timur
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - R Neslihan Gürsoy
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
330
|
Lukasevics L, Cizikovs A, Grigorjeva L. Cobalt-Catalyzed C(sp 2)-H Carbonylation of Amino Acids Using Picolinamide as a Traceless Directing Group. Org Lett 2021; 23:2748-2753. [PMID: 33724856 DOI: 10.1021/acs.orglett.1c00660] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Herein we report an efficient protocol for the C(sp2)-H carbonylation of amino acid derivatives based on an inexpensive cobalt(II) salt catalyst. Carbonylation was accomplished using picolinamide as a traceless directing group, CO (1 atm) as the carbonyl source, and Co(dpm)2 as the catalyst. A broad range of phenylalanine derivatives bearing diverse functional groups were tolerated. Moreover, the method can be successfully applied for the C(sp2)-H carbonylation of short peptides thereby allowing access for peptide late-stage carbonylation.
Collapse
Affiliation(s)
- Lukass Lukasevics
- Latvian Institute of Organic Synthesis, Aizkraukles Street 21, Riga LV-1006, Latvia
| | - Aleksandrs Cizikovs
- Latvian Institute of Organic Synthesis, Aizkraukles Street 21, Riga LV-1006, Latvia
| | - Liene Grigorjeva
- Latvian Institute of Organic Synthesis, Aizkraukles Street 21, Riga LV-1006, Latvia
| |
Collapse
|
331
|
Midde S, Vytla D, Velayuthaperumal R, Kaliyaperumal K, Reddy CA, Jarugu LB, Gupta A, Roy A, Mathur A. Solid-phase synthesis of JBIR-126 (Tambromycin), JBIR-35 and their analogs. Tetrahedron Lett 2021. [DOI: 10.1016/j.tetlet.2021.152970] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
332
|
Structural Characterization of Receptor-Receptor Interactions in the Allosteric Modulation of G Protein-Coupled Receptor (GPCR) Dimers. Int J Mol Sci 2021; 22:ijms22063241. [PMID: 33810175 PMCID: PMC8005122 DOI: 10.3390/ijms22063241] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/17/2021] [Accepted: 03/20/2021] [Indexed: 01/07/2023] Open
Abstract
G protein-coupled receptor (GPCR) oligomerization, while contentious, continues to attract the attention of researchers. Numerous experimental investigations have validated the presence of GPCR dimers, and the relevance of dimerization in the effectuation of physiological functions intensifies the attractiveness of this concept as a potential therapeutic target. GPCRs, as a single entity, have been the main source of scrutiny for drug design objectives for multiple diseases such as cancer, inflammation, cardiac, and respiratory diseases. The existence of dimers broadens the research scope of GPCR functions, revealing new signaling pathways that can be targeted for disease pathogenesis that have not previously been reported when GPCRs were only viewed in their monomeric form. This review will highlight several aspects of GPCR dimerization, which include a summary of the structural elucidation of the allosteric modulation of class C GPCR activation offered through recent solutions to the three-dimensional, full-length structures of metabotropic glutamate receptor and γ-aminobutyric acid B receptor as well as the role of dimerization in the modification of GPCR function and allostery. With the growing influence of computational methods in the study of GPCRs, we will also be reviewing recent computational tools that have been utilized to map protein-protein interactions (PPI).
Collapse
|
333
|
Zhou P, Liu Q, Wu T, Miao Q, Shang S, Wang H, Chen Z, Wang S, Wang H. Systematic Comparison and Comprehensive Evaluation of 80 Amino Acid Descriptors in Peptide QSAR Modeling. J Chem Inf Model 2021; 61:1718-1731. [DOI: 10.1021/acs.jcim.0c01370] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Peng Zhou
- Center for Informational Biology, University of Electronic Science and Technology of China (UESTC) at Qingshuihe Campus, Chengdu 611731, China
- School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC) at Shahe Campus, Chengdu 610054, China
| | - Qian Liu
- Center for Informational Biology, University of Electronic Science and Technology of China (UESTC) at Qingshuihe Campus, Chengdu 611731, China
- School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC) at Shahe Campus, Chengdu 610054, China
| | - Ting Wu
- School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC) at Shahe Campus, Chengdu 610054, China
| | - Qingqing Miao
- Center for Informational Biology, University of Electronic Science and Technology of China (UESTC) at Qingshuihe Campus, Chengdu 611731, China
- School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC) at Shahe Campus, Chengdu 610054, China
| | - Shuyong Shang
- College of Chemistry and Life Science, Chengdu Normal University, Chengdu 611130, China
| | - Heyi Wang
- Center for Informational Biology, University of Electronic Science and Technology of China (UESTC) at Qingshuihe Campus, Chengdu 611731, China
- School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC) at Shahe Campus, Chengdu 610054, China
| | - Zheng Chen
- Center for Informational Biology, University of Electronic Science and Technology of China (UESTC) at Qingshuihe Campus, Chengdu 611731, China
- School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC) at Shahe Campus, Chengdu 610054, China
| | - Shaozhou Wang
- School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC) at Shahe Campus, Chengdu 610054, China
| | - Heyan Wang
- School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC) at Shahe Campus, Chengdu 610054, China
| |
Collapse
|
334
|
Casey R, Adelfio A, Connolly M, Wall A, Holyer I, Khaldi N. Discovery through Machine Learning and Preclinical Validation of Novel Anti-Diabetic Peptides. Biomedicines 2021; 9:276. [PMID: 33803471 PMCID: PMC8000967 DOI: 10.3390/biomedicines9030276] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/05/2021] [Accepted: 03/07/2021] [Indexed: 12/19/2022] Open
Abstract
While there have been significant advances in drug discovery for diabetes mellitus over the past couple of decades, there is an opportunity and need for improved therapies. While type 2 diabetic patients better manage their illness, many of the therapeutics in this area are peptide hormones with lengthy sequences and a molecular structure that makes them challenging and expensive to produce. Using machine learning, we present novel anti-diabetic peptides which are less than 16 amino acids in length, distinct from human signalling peptides. We validate the capacity of these peptides to stimulate glucose uptake and Glucose transporter type 4 (GLUT4) translocation in vitro. In obese insulin-resistant mice, predicted peptides significantly lower plasma glucose, reduce glycated haemoglobin and even improve hepatic steatosis when compared to treatments currently in use in a clinical setting. These unoptimised, linear peptides represent promising candidates for blood glucose regulation which require further evaluation. Further, this indicates that perhaps we have overlooked the class of natural short linear peptides, which usually come with an excellent safety profile, as therapeutic modalities.
Collapse
Affiliation(s)
| | | | | | - Audrey Wall
- Nuritas Ltd., Joshua Dawson House, D02 RY95 Dublin, Ireland; (R.C.); (A.A.); (M.C.); (I.H.); (N.K.)
| | | | | |
Collapse
|
335
|
Makhlynets OV, Caputo GA. Characteristics and therapeutic applications of antimicrobial peptides. BIOPHYSICS REVIEWS 2021; 2:011301. [PMID: 38505398 PMCID: PMC10903410 DOI: 10.1063/5.0035731] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/31/2020] [Indexed: 12/20/2022]
Abstract
The demand for novel antimicrobial compounds is rapidly growing due to the phenomenon of antibiotic resistance in bacteria. In response, numerous alternative approaches are being taken including use of polymers, metals, combinatorial approaches, and antimicrobial peptides (AMPs). AMPs are a naturally occurring part of the immune system of all higher organisms and display remarkable broad-spectrum activity and high selectivity for bacterial cells over host cells. However, despite good activity and safety profiles, AMPs have struggled to find success in the clinic. In this review, we outline the fundamental properties of AMPs that make them effective antimicrobials and extend this into three main approaches being used to help AMPs become viable clinical options. These three approaches are the incorporation of non-natural amino acids into the AMP sequence to impart better pharmacological properties, the incorporation of AMPs in hydrogels, and the chemical modification of surfaces with AMPs for device applications. These approaches are being developed to enhance the biocompatibility, stability, and/or bioavailability of AMPs as clinical options.
Collapse
Affiliation(s)
- Olga V. Makhlynets
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, New York 13244, USA
| | | |
Collapse
|
336
|
|
337
|
MCH-R1 Antagonist GPS18169, a Pseudopeptide, Is a Peripheral Anti-Obesity Agent in Mice. Molecules 2021; 26:molecules26051291. [PMID: 33673598 PMCID: PMC7957705 DOI: 10.3390/molecules26051291] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/17/2021] [Accepted: 02/22/2021] [Indexed: 01/01/2023] Open
Abstract
Melanin-concentrating hormone (MCH) is a 19 amino acid long peptide found in the brain of animals, including fishes, batrachians, and mammals. MCH is implicated in appetite and/or energy homeostasis. Antagonists at its receptor (MCH-R1) could be major tools (or ultimately drugs) to understand the mechanism of MCH action and to fight the obesity syndrome that is a worldwide societal health problem. Ever since the deorphanisation of the MCH receptor, we cloned, expressed, and characterized the receptor MCH-R1 and started a vast medicinal chemistry program aiming at the discovery of such usable compounds. In the present final work, we describe GPS18169, a pseudopeptide antagonist at the MCH-R1 receptor with an affinity in the nanomolar range and a Ki for its antagonistic effect in the 20 picomolar range. Its metabolic stability is rather ameliorated compared to its initial parent compound, the antagonist S38151. We tested it in an in vivo experiment using high diet mice. GPS18169 was found to be active in limiting the accumulation of adipose tissues and, correlatively, we observed a normalization of the insulin level in the treated animals, while no change in food or water consumption was observed.
Collapse
|
338
|
Philippe GJB, Mittermeier A, Lawrence N, Huang YH, Condon ND, Loewer A, Craik DJ, Henriques ST. Angler Peptides: Macrocyclic Conjugates Inhibit p53:MDM2/X Interactions and Activate Apoptosis in Cancer Cells. ACS Chem Biol 2021; 16:414-428. [PMID: 33533253 DOI: 10.1021/acschembio.0c00988] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Peptides are being developed as targeted anticancer drugs to modulate cytosolic protein-protein interactions involved in cancer progression. However, their use as therapeutics is often limited by their low cell membrane permeation and/or inability to reach cytosolic targets. Conjugation to cell penetrating peptides has been successfully used to improve the cytosolic delivery of high affinity binder peptides, but cellular uptake does not always result in modulation of the targeted pathway. To overcome this limitation, we developed "angler peptides" by conjugating KD3, a noncell permeable but potent and specific peptide inhibitor of p53:MDM2 and p53:MDMX interactions, with a set of cyclic cell-penetrating peptides. We examined their binding affinity for MDM2 and MDMX, the cell entry mechanism, and role in reactivation of the p53 pathway. We identified two angler peptides, cTAT-KD3 and cR10-KD3, able to activate the p53 pathway in cancer cells. cTAT-KD3 entered cells via endocytic pathways, escaped endosomes, and activated the p53 pathway in breast (MCF7), lung (A549), and colon (HCT116) cancer cell lines at concentrations in the range of 1-12 μM. cR10-KD3 reached the cytosol via direct membrane translocation and activated the p53 pathway at 1 μM in all the tested cell lines. Our work demonstrates that nonpermeable anticancer peptides can be delivered into the cytosol and inhibit intracellular cancer pathways when they are conjugated with stable cell penetrating peptides. The mechanistic studies suggest that direct translocation leads to less toxicity, higher cytosol delivery at lower concentrations, and lower dependencies on the membrane of the tested cell line than occurs for an endocytic pathway with endosomal escape. The angler strategy can rescue high affinity peptide binders identified from high throughput screening and convert them into targeted anticancer therapeutics, but investigation of their cellular uptake and cell death mechanisms is essential to confirming modulation of the targeted cancer pathways.
Collapse
Affiliation(s)
- Grégoire J.-B. Philippe
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| | | | - Nicole Lawrence
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Yen-Hua Huang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Nicholas D. Condon
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | | | - David J. Craik
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Sónia T. Henriques
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
- Queensland University of Technology, School of Biomedical Sciences, Institute of Health & Biomedical Innovation and Translational Research Institute, Brisbane, Queensland 4102, Australia
| |
Collapse
|
339
|
Tuning of a Membrane-Perforating Antimicrobial Peptide to Selectively Target Membranes of Different Lipid Composition. J Membr Biol 2021; 254:75-96. [PMID: 33564914 DOI: 10.1007/s00232-021-00174-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 01/21/2021] [Indexed: 12/16/2022]
Abstract
The use of designed antimicrobial peptides as drugs has been impeded by the absence of simple sequence-structure-function relationships and design rules. The likely cause is that many of these peptides permeabilize membranes via highly disordered, heterogeneous mechanisms, forming aggregates without well-defined tertiary or secondary structure. We suggest that the combination of high-throughput library screening with atomistic computer simulations can successfully address this challenge by tuning a previously developed general pore-forming peptide into a selective pore-former for different lipid types. A library of 2916 peptides was designed based on the LDKA template. The library peptides were synthesized and screened using a high-throughput orthogonal vesicle leakage assay. Dyes of different sizes were entrapped inside vesicles with varying lipid composition to simultaneously screen for both pore size and affinity for negatively charged and neutral lipid membranes. From this screen, nine different LDKA variants that have unique activity were selected, sequenced, synthesized, and characterized. Despite the minor sequence changes, each of these peptides has unique functional properties, forming either small or large pores and being selective for either neutral or anionic lipid bilayers. Long-scale, unbiased atomistic molecular dynamics (MD) simulations directly reveal that rather than rigid, well-defined pores, these peptides can form a large repertoire of functional dynamic and heterogeneous aggregates, strongly affected by single mutations. Predicting the propensity to aggregate and assemble in a given environment from sequence alone holds the key to functional prediction of membrane permeabilization.
Collapse
|
340
|
Macpherson A, Laabei M, Ahdash Z, Graewert MA, Birtley JR, Schulze MSE, Crennell S, Robinson SA, Holmes B, Oleinikovas V, Nilsson PH, Snowden J, Ellis V, Mollnes TE, Deane CM, Svergun D, Lawson AD, van den Elsen JM. The allosteric modulation of complement C5 by knob domain peptides. eLife 2021; 10:63586. [PMID: 33570492 PMCID: PMC7972453 DOI: 10.7554/elife.63586] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/11/2021] [Indexed: 12/22/2022] Open
Abstract
Bovines have evolved a subset of antibodies with ultra-long heavy chain complementarity determining regions that harbour cysteine-rich knob domains. To produce high-affinity peptides, we previously isolated autonomous 3–6 kDa knob domains from bovine antibodies. Here, we show that binding of four knob domain peptides elicits a range of effects on the clinically validated drug target complement C5. Allosteric mechanisms predominated, with one peptide selectively inhibiting C5 cleavage by the alternative pathway C5 convertase, revealing a targetable mechanistic difference between the classical and alternative pathway C5 convertases. Taking a hybrid biophysical approach, we present C5-knob domain co-crystal structures and, by solution methods, observed allosteric effects propagating >50 Å from the binding sites. This study expands the therapeutic scope of C5, presents new inhibitors, and introduces knob domains as new, low molecular weight antibody fragments, with therapeutic potential. Antibodies are proteins produced by the immune system that can selectively bind to other molecules and modify their behaviour. Cows are highly equipped at fighting-off disease-causing microbes due to the unique shape of some of their antibodies. Unlike other jawed vertebrates, cows’ antibodies contain an ultra-long loop region that contains a ‘knob domain’ which sticks out from the rest of the antibody. Recent research has shown that when detached, the knob domain behaves like an antibody fragment, and can independently bind to a range of different proteins. Antibody fragments are commonly developed in the laboratory to target proteins associated with certain diseases, such as arthritis and cancer. But it was unclear whether the knob domains from cows’ antibodies could also have therapeutic potential. To investigate this, Macpherson et al. studied how knob domains attach to complement C5, a protein in the inflammatory pathway which is a drug target for various diseases, including severe COVID-19. The experiments identified various knob domains that bind to complement C5 and inhibits its activity by altering its structure or movement. Further tests studying the structure of these interactions, led to the discovery of a common mechanism by which inhibitors can modify the behaviour of this inflammatory protein. Complement C5 is involved in numerous molecular pathways in the immune system, which means many of the drugs developed to inhibit its activity can also leave patients vulnerable to infection. However, one of the knob domains identified by Macpherson et al. was found to reduce the activity of complement C5 in some pathways, whilst leaving other pathways intact. This could potentially reduce the risk of bacterial infections which sometimes arise following treatment with these types of inhibitors. These findings highlight a new approach for developing drug inhibitors for complement C5. Furthermore, the ability of knob domains to bind to multiple sites of complement C5 suggests that this fragment could be used to target proteins associated with other diseases.
Collapse
Affiliation(s)
- Alex Macpherson
- UCB, Slough, United Kingdom.,Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Maisem Laabei
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | | | | | | | | | - Susan Crennell
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Sarah A Robinson
- Department of Statistics, University of Oxford, Oxford, United Kingdom
| | | | | | - Per H Nilsson
- UCB, Slough, United Kingdom.,Department of Chemistry and Biomedicine, Linnaeus University, Kalmar, Sweden.,Department of Immunology, Oslo University Hospital, University of Oslo, Oslo, Norway
| | | | | | - Tom Eirik Mollnes
- Department of Immunology, Oslo University Hospital, University of Oslo, Oslo, Norway.,Research Laboratory, Bodø Hospital, K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway.,Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Charlotte M Deane
- Department of Statistics, University of Oxford, Oxford, United Kingdom
| | - Dmitri Svergun
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany
| | | | - Jean Mh van den Elsen
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom.,Centre for Therapeutic Innovation, University of Bath, Bath, United Kingdom
| |
Collapse
|
341
|
Toplak A, Teixeira de Oliveira EF, Schmidt M, Rozeboom HJ, Wijma HJ, Meekels LKM, de Visser R, Janssen DB, Nuijens T. From thiol-subtilisin to omniligase: Design and structure of a broadly applicable peptide ligase. Comput Struct Biotechnol J 2021; 19:1277-1287. [PMID: 33717424 PMCID: PMC7921005 DOI: 10.1016/j.csbj.2021.02.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 02/01/2021] [Accepted: 02/01/2021] [Indexed: 11/05/2022] Open
Abstract
Omniligase-1 is a broadly applicable enzyme for peptide bond formation between an activated acyl donor peptide and a non-protected acyl acceptor peptide. The enzyme is derived from an earlier subtilisin variant called peptiligase by several rounds of protein engineering aimed at increasing synthetic yields and substrate range. To examine the contribution of individual mutations on S/H ratio and substrate scope in peptide synthesis, we selected peptiligase variant M222P/L217H as a starting enzyme and introduced successive mutations. Mutation A225N in the S1′ pocket and F189W of the S2′ pocket increased the synthesis to hydrolysis (S/H) ratio and overall coupling efficiency, whereas the I107V mutation was added to S4 pocket to increase the reaction rate. The final omniligase variants appeared to have a very broad substrate range, coupling more than 250 peptides in a 400-member library of acyl acceptors, as indicated by a high-throughput FRET assay. Crystal structures and computational modelling could rationalize the exceptional properties of omniligase-1 in peptide synthesis
Collapse
Affiliation(s)
- Ana Toplak
- EnzyPep B.V., Brightlands Campus Urmonderbaan 22, 6167 RD Geleen, The Netherlands
| | - Eduardo F Teixeira de Oliveira
- Biotransformation and Biocatalysis, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Marcel Schmidt
- EnzyPep B.V., Brightlands Campus Urmonderbaan 22, 6167 RD Geleen, The Netherlands
| | - Henriëtte J Rozeboom
- Biotransformation and Biocatalysis, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Hein J Wijma
- Biotransformation and Biocatalysis, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Linda K M Meekels
- EnzyPep B.V., Brightlands Campus Urmonderbaan 22, 6167 RD Geleen, The Netherlands
| | - Rowin de Visser
- EnzyPep B.V., Brightlands Campus Urmonderbaan 22, 6167 RD Geleen, The Netherlands
| | - Dick B Janssen
- Biotransformation and Biocatalysis, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Timo Nuijens
- EnzyPep B.V., Brightlands Campus Urmonderbaan 22, 6167 RD Geleen, The Netherlands
| |
Collapse
|
342
|
Zitterbart R, Berger N, Reimann O, Noble GT, Lüdtke S, Sarma D, Seitz O. Traceless parallel peptide purification by a first-in-class reductively cleavable linker system featuring a safety-release. Chem Sci 2021; 12:2389-2396. [PMID: 34164003 PMCID: PMC8179278 DOI: 10.1039/d0sc06285e] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/07/2021] [Indexed: 12/26/2022] Open
Abstract
Hundreds of peptides can be synthesized by automated parallel synthesizers in a single run. In contrast, the most widely used peptide purification method - high-pressure liquid chromatography (HPLC) - only allows one-by-one processing of each sample. The chromatographic purification of many peptides, therefore, remains a time-consuming and costly effort. Catch-and-release methods can be processed in parallel and potentially provide a remedy. However, no such system has yet provided a true alternative to HPLC. Herein we present the development of a side-reaction free, reductively cleavable linker. The linker is added to the target peptide as the last building block during peptide synthesis. After acidic cleavage from synthetic resin, the linker-tagged full-length peptide is caught onto an aldehyde-modified solid support by rapid oxime ligation, allowing removal of all impurities lacking the linker by washing. Reducing the aryl azide to an aniline sensitizes the linker for cleavage. However, scission does not occur at non-acidic pH enabling wash out of reducing agent. Final acidic treatment safely liberates the peptide by an acid-catalysed 1,6-elimination. We showcase this first-in-class reductively cleavable linker system in the parallel purification of a personalized neoantigen cocktail, containing 20 peptides for cancer immunotherapy within six hours.
Collapse
Affiliation(s)
| | - Nadja Berger
- Belyntic GmbH Richard-Willstätter-Str. 11 12489 Berlin Germany
| | - Oliver Reimann
- Belyntic GmbH Richard-Willstätter-Str. 11 12489 Berlin Germany
| | - Gavin T Noble
- Bachem (UK) Ltd. Delph Court, Sullivans Way, St. Helens Merseyside WA9 5GL UK
| | - Stephan Lüdtke
- Belyntic GmbH Richard-Willstätter-Str. 11 12489 Berlin Germany
| | - Dominik Sarma
- Belyntic GmbH Richard-Willstätter-Str. 11 12489 Berlin Germany
| | - Oliver Seitz
- Humboldt-Universität zu Berlin Brook-Taylor-Str. 2 12489 Berlin Germany
| |
Collapse
|
343
|
Zhang K, Zhou L, Zhang T, Fan Z, Xie M, Ding Y, Li H. Peptide based Biosensing of Protein Functional Control Indicates Novel Mechanism of Cancerous Development under Oxidative Stress. SENSORS AND ACTUATORS B: CHEMICAL 2021; 329:129121. [DOI: 10.1016/j.snb.2020.129121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
344
|
Cavaco M, Andreu D, Castanho MARB. The Challenge of Peptide Proteolytic Stability Studies: Scarce Data, Difficult Readability, and the Need for Harmonization. Angew Chem Int Ed Engl 2021; 60:1686-1688. [PMID: 33200441 DOI: 10.1002/anie.202006372] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/28/2020] [Indexed: 11/06/2022]
Abstract
Proteolytic stability assessment is increasingly viewed as a fundamental component of peptide characterization, arguably of comparable importance as efficacy and toxicity data. A literature survey over the last decade reveals steady growth in the stability information available. However, it also uncovers two significant problems that hinder proper data comparison: 1) the use of different stability assays, and 2) the differences in how stability information is reported. In this Viewpoint, we present results from a database meta-analysis as well as concerns about the stability assessments published so far. We also suggest guidelines for a proper discussion between experts in the field on how to improve data readability so that peptide stability, an often-missing parameter in older literature, is adequately reported to take maximum advantage of it.
Collapse
Affiliation(s)
- Marco Cavaco
- Department of Biochemistry, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028, Lisboa, Portugal.,Proteomics and Protein Chemistry Unit, Department of Experimental and Health Sciences, Pompeu Fabra University, Dr Aiguader 88, Barcelona Biomedical Research Park, 08003, Barcelona, Spain
| | - David Andreu
- Proteomics and Protein Chemistry Unit, Department of Experimental and Health Sciences, Pompeu Fabra University, Dr Aiguader 88, Barcelona Biomedical Research Park, 08003, Barcelona, Spain
| | - Miguel A R B Castanho
- Department of Biochemistry, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028, Lisboa, Portugal
| |
Collapse
|
345
|
Mylonis I, Chachami G, Simos G. Specific Inhibition of HIF Activity: Can Peptides Lead the Way? Cancers (Basel) 2021; 13:cancers13030410. [PMID: 33499237 PMCID: PMC7865418 DOI: 10.3390/cancers13030410] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/15/2021] [Accepted: 01/16/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Cancer cells in solid tumors often experience lack of oxygen (hypoxia), which they overcome with the help of hypoxia inducible transcription factors (HIFs). When HIFs are activated, they stimulate the expression of many genes and cause the production of proteins that help cancer cells grow and migrate even in the presence of very little oxygen. Many experiments have shown that agents that block the activity of HIFs (HIF inhibitors) can prevent growth of cancer cells under hypoxia and, subsequently, hinder formation of malignant tumors or metastases. Most small chemical HIF inhibitors lack the selectivity required for development of safe anticancer drugs. On the other hand, peptides derived from HIFs themselves can be very selective HIF inhibitors by disrupting specific associations of HIFs with cellular components that are essential for HIF activation. This review discusses the nature of available peptide HIF inhibitors and their prospects as effective pharmaceuticals against cancer. Abstract Reduced oxygen availability (hypoxia) is a characteristic of many disorders including cancer. Central components of the systemic and cellular response to hypoxia are the Hypoxia Inducible Factors (HIFs), a small family of heterodimeric transcription factors that directly or indirectly regulate the expression of hundreds of genes, the products of which mediate adaptive changes in processes that include metabolism, erythropoiesis, and angiogenesis. The overexpression of HIFs has been linked to the pathogenesis and progression of cancer. Moreover, evidence from cellular and animal models have convincingly shown that targeting HIFs represents a valid approach to treat hypoxia-related disorders. However, targeting transcription factors with small molecules is a very demanding task and development of HIF inhibitors with specificity and therapeutic potential has largely remained an unattainable challenge. Another promising approach to inhibit HIFs is to use peptides modelled after HIF subunit domains known to be involved in protein–protein interactions that are critical for HIF function. Introduction of these peptides into cells can inhibit, through competition, the activity of endogenous HIFs in a sequence and, therefore also isoform, specific manner. This review summarizes the involvement of HIFs in cancer and the approaches for targeting them, with a special focus on the development of peptide HIF inhibitors and their prospects as highly-specific pharmacological agents.
Collapse
Affiliation(s)
- Ilias Mylonis
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece;
- Correspondence: (I.M.); (G.S.)
| | - Georgia Chachami
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece;
| | - George Simos
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece;
- Gerald Bronfman Department of Oncology, Faculty of Medicine, McGill University, Montreal, QC H4A 3T2, Canada
- Correspondence: (I.M.); (G.S.)
| |
Collapse
|
346
|
Lee IK, Kim G, Kim DH, Kim BB. PEG-BHD1028 Peptide Regulates Insulin Resistance and Fatty Acid β-Oxidation, and Mitochondrial Biogenesis by Binding to Two Heterogeneous Binding Sites of Adiponectin Receptors, AdipoR1 and AdipoR2. Int J Mol Sci 2021; 22:ijms22020884. [PMID: 33477324 PMCID: PMC7830917 DOI: 10.3390/ijms22020884] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 12/17/2022] Open
Abstract
Adiponectin plays multiple critical roles in modulating various physiological processes by binding to its receptors. The functions of PEG-BHD1028, a potent novel peptide agonist to AdipoRs, was evaluated using in vitro and in vivo models based on the reported action spectrum of adiponectin. To confirm the design concept of PEG-BHD1028, the binding sites and their affinities were analyzed using the SPR (Surface Plasmon Resonance) assay. The results revealed that PEG-BHD1028 was bound to two heterogeneous binding sites of AdipoR1 and AdipoR2 with a relatively high affinity. In C2C12 cells, PEG-BHD1028 significantly activated AMPK and subsequent pathways and enhanced fatty acid β-oxidation and mitochondrial biogenesis. Furthermore, it also facilitated glucose uptake by lowering insulin resistance in insulin-resistant C2C12 cells. PEG-BHD1028 significantly reduced the fasting plasma glucose level in db/db mice following a single s.c. injection of 50, 100, and 200 μg/Kg and glucose tolerance at a dose of 50 μg/Kg with significantly decreased insulin production. The animals received 5, 25, and 50 μg/Kg of PEG-BHD1028 for 21 days significantly lost their weight after 18 days in a range of 5-7%. These results imply the development of PEG-BHD1028 as a potential adiponectin replacement therapeutic agent.
Collapse
Affiliation(s)
| | | | | | - Brian B. Kim
- Correspondence: ; Tel.: +82-31-360-3132; Fax: +82-31-360-3133
| |
Collapse
|
347
|
Sala V, Cnudde SJ, Murabito A, Massarotti A, Hirsch E, Ghigo A. Therapeutic peptides for the treatment of cystic fibrosis: Challenges and perspectives. Eur J Med Chem 2021; 213:113191. [PMID: 33493828 DOI: 10.1016/j.ejmech.2021.113191] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/21/2020] [Accepted: 01/08/2021] [Indexed: 02/07/2023]
Abstract
Cystic fibrosis (CF) is the most common amongst rare genetic diseases, affecting more than 70.000 people worldwide. CF is characterized by a dysfunctional chloride channel, termed cystic fibrosis conductance regulator (CFTR), which leads to the production of a thick and viscous mucus layer that clogs the lungs of CF patients and traps pathogens, leading to chronic infections and inflammation and, ultimately, lung damage. In recent years, the use of peptides for the treatment of respiratory diseases, including CF, has gained growing interest. Therapeutic peptides for CF include antimicrobial peptides, inhibitors of proteases, and modulators of ion channels, among others. Peptides display unique features that make them appealing candidates for clinical translation, like specificity of action, high efficacy, and low toxicity. Nevertheless, the intrinsic properties of peptides, together with the need of delivering these compounds locally, e.g. by inhalation, raise a number of concerns in the development of peptide therapeutics for CF lung disease. In this review, we discuss the challenges related to the use of peptides for the treatment of CF lung disease through inhalation, which include retention within mucus, proteolysis, immunogenicity and aggregation. Strategies for overcoming major shortcomings of peptide therapeutics will be presented, together with recent developments in peptide design and optimization, including computational analysis and high-throughput screening.
Collapse
Affiliation(s)
- Valentina Sala
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Sophie Julie Cnudde
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Alessandra Murabito
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Alberto Massarotti
- Department of Pharmaceutical Science, University of Piemonte Orientale "A. Avogadro", Largo Donegani 2, 28100, Novara, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy; Kither Biotech S.r.l., Via Nizza 52, 10126, Torino, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy; Kither Biotech S.r.l., Via Nizza 52, 10126, Torino, Italy.
| |
Collapse
|
348
|
Bennett R, Pirrone GF, Nowak T, Mukherjee D, Shchurik V, Mapelli C, Hickey JL, Regalado EL, Mangion I, Makarov AA. Ultra-high-throughput SPE-MALDI workflow: Blueprint for efficient purification and screening of peptide libraries. Anal Chim Acta 2021; 1142:10-18. [PMID: 33280687 DOI: 10.1016/j.aca.2020.10.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/19/2020] [Accepted: 10/21/2020] [Indexed: 12/30/2022]
Abstract
At the forefront of synthetic endeavors in the pharmaceutical industry, including drug discovery and high-throughput screening, timelines are tight and large quantities of pure chemical targets are rarely available. In this regard, the development of novel and increasingly challenging chemistries requires a commensurate level of innovation to develop reliable analytical assays and purification workflows with rapid turnaround that enables accelerated pharmacological evaluation. A small-scale automation platform enabling high-throughput analysis and purification to streamline the selection of candidate leads would be a transformative advance. Herein, we introduce an automation-friendly solid-phase extraction-matrix-assisted laser desorption/ionization (SPE-MALDI) platform applied to the high-throughput purification and analysis of peptide libraries. This advance enabled us to purify peptides from microgram levels in less than a day with results comparable to traditional high-performance liquid chromatography-diode array detection-mass spectrometry (HPLC-DAD-MS).
Collapse
Affiliation(s)
- Raffeal Bennett
- Merck & Co., Inc., MRL, Analytical Research & Development, Rahway, NJ, 07065, USA.
| | - Gregory F Pirrone
- Merck & Co., Inc., MRL, Analytical Research & Development, Rahway, NJ, 07065, USA.
| | - Timothy Nowak
- Merck & Co., Inc., MRL, Analytical Research & Development, Rahway, NJ, 07065, USA
| | - Debopreeti Mukherjee
- Merck & Co., Inc., MRL, Analytical Research & Development, Rahway, NJ, 07065, USA
| | - Vladimir Shchurik
- Merck & Co., Inc., MRL, Analytical Research & Development, Rahway, NJ, 07065, USA
| | - Claudio Mapelli
- Merck & Co., Inc., MRL, Chemistry Capabilities for Accelerating Therapeutics, Discovery Chemistry, Kenilworth, NJ, 07033, USA
| | - Jennifer L Hickey
- Merck & Co., Inc., MRL, Chemistry Capabilities for Accelerating Therapeutics, Discovery Chemistry, Kenilworth, NJ, 07033, USA
| | - Erik L Regalado
- Merck & Co., Inc., MRL, Analytical Research & Development, Rahway, NJ, 07065, USA.
| | - Ian Mangion
- Merck & Co., Inc., MRL, Analytical Research & Development, Rahway, NJ, 07065, USA
| | - Alexey A Makarov
- Merck & Co., Inc., MRL, Analytical Research & Development, Rahway, NJ, 07065, USA
| |
Collapse
|
349
|
Apostolopoulos V, Bojarska J, Chai TT, Elnagdy S, Kaczmarek K, Matsoukas J, New R, Parang K, Lopez OP, Parhiz H, Perera CO, Pickholz M, Remko M, Saviano M, Skwarczynski M, Tang Y, Wolf WM, Yoshiya T, Zabrocki J, Zielenkiewicz P, AlKhazindar M, Barriga V, Kelaidonis K, Sarasia EM, Toth I. A Global Review on Short Peptides: Frontiers and Perspectives. Molecules 2021; 26:430. [PMID: 33467522 PMCID: PMC7830668 DOI: 10.3390/molecules26020430] [Citation(s) in RCA: 216] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/23/2020] [Accepted: 01/09/2021] [Indexed: 12/13/2022] Open
Abstract
Peptides are fragments of proteins that carry out biological functions. They act as signaling entities via all domains of life and interfere with protein-protein interactions, which are indispensable in bio-processes. Short peptides include fundamental molecular information for a prelude to the symphony of life. They have aroused considerable interest due to their unique features and great promise in innovative bio-therapies. This work focusing on the current state-of-the-art short peptide-based therapeutical developments is the first global review written by researchers from all continents, as a celebration of 100 years of peptide therapeutics since the commencement of insulin therapy in the 1920s. Peptide "drugs" initially played only the role of hormone analogs to balance disorders. Nowadays, they achieve numerous biomedical tasks, can cross membranes, or reach intracellular targets. The role of peptides in bio-processes can hardly be mimicked by other chemical substances. The article is divided into independent sections, which are related to either the progress in short peptide-based theranostics or the problems posing challenge to bio-medicine. In particular, the SWOT analysis of short peptides, their relevance in therapies of diverse diseases, improvements in (bio)synthesis platforms, advanced nano-supramolecular technologies, aptamers, altered peptide ligands and in silico methodologies to overcome peptide limitations, modern smart bio-functional materials, vaccines, and drug/gene-targeted delivery systems are discussed.
Collapse
Affiliation(s)
- Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (V.A.); (J.M.); (V.B.)
| | - Joanna Bojarska
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, Żeromskiego 116, 90-924 Lodz, Poland
| | - Tsun-Thai Chai
- Department of Chemical Science, Faculty of Science, Universiti Tunku Abdul Rahman, Kampar 31900, Malaysia;
| | - Sherif Elnagdy
- Botany and Microbiology Department, Faculty of Science, Cairo University, Gamaa St., Giza 12613, Egypt; (S.E.); (M.A.)
| | - Krzysztof Kaczmarek
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Żeromskiego 116, 90-924 Lodz, Poland; (K.K.); (J.Z.)
| | - John Matsoukas
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (V.A.); (J.M.); (V.B.)
- NewDrug, Patras Science Park, 26500 Patras, Greece;
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Roger New
- Vaxcine (UK) Ltd., c/o London Bioscience Innovation Centre, London NW1 0NH, UK;
- Faculty of Science & Technology, Middlesex University, The Burroughs, London NW4 4BT, UK;
| | - Keykavous Parang
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA;
| | - Octavio Paredes Lopez
- Centro de Investigación y de Estudios Avanzados del IPN, Departamento de Biotecnología y Bioquímica, Irapuato 36824, Guanajuato, Mexico;
| | - Hamideh Parhiz
- Infectious Disease Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6073, USA;
| | - Conrad O. Perera
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand;
| | - Monica Pickholz
- Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires 1428, Argentina;
- Instituto de Física de Buenos Aires (IFIBA, UBA-CONICET), Argentina, Buenos Aires 1428, Argentina
| | - Milan Remko
- Remedika, Luzna 9, 85104 Bratislava, Slovakia;
| | - Michele Saviano
- Institute of Crystallography (CNR), Via Amendola 122/o, 70126 Bari, Italy;
| | - Mariusz Skwarczynski
- School of Chemistry & Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (I.T.)
| | - Yefeng Tang
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (MOE), School of Pharma Ceutical Sciences, Tsinghua University, Beijing 100084, China;
| | - Wojciech M. Wolf
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, Żeromskiego 116, 90-924 Lodz, Poland
| | | | - Janusz Zabrocki
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Żeromskiego 116, 90-924 Lodz, Poland; (K.K.); (J.Z.)
| | - Piotr Zielenkiewicz
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106 Warsaw, Poland;
- Department of Systems Biology, Institute of Experimental Plant Biology and Biotechnology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Maha AlKhazindar
- Botany and Microbiology Department, Faculty of Science, Cairo University, Gamaa St., Giza 12613, Egypt; (S.E.); (M.A.)
| | - Vanessa Barriga
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (V.A.); (J.M.); (V.B.)
| | | | | | - Istvan Toth
- School of Chemistry & Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (I.T.)
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia
| |
Collapse
|
350
|
Raynal L, Rose NC, Donald JR, Spicer CD. Photochemical Methods for Peptide Macrocyclisation. Chemistry 2021; 27:69-88. [PMID: 32914455 PMCID: PMC7821122 DOI: 10.1002/chem.202003779] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/10/2020] [Indexed: 12/19/2022]
Abstract
Photochemical reactions have been the subject of renewed interest over the last two decades, leading to the development of many new, diverse and powerful chemical transformations. More recently, these developments have been expanded to enable the photochemical macrocyclisation of peptides and small proteins. These constructs benefit from increased stability, structural rigidity and biological potency over their linear counterparts, providing opportunities for improved therapeutic agents. In this review, an overview of both the established and emerging methods for photochemical peptide macrocyclisation is presented, highlighting both the limitations and opportunities for further innovation in the field.
Collapse
Affiliation(s)
- Laetitia Raynal
- Department of ChemistryUniversity of YorkHeslingtonYorkYO10 5DDUK
| | - Nicholas C. Rose
- Department of ChemistryUniversity of YorkHeslingtonYorkYO10 5DDUK
| | - James R. Donald
- Department of ChemistryUniversity of YorkHeslingtonYorkYO10 5DDUK
- York Biomedical Research InstituteUniversity of YorkHeslingtonYorkYO10 5DDUK
| | - Christopher D. Spicer
- Department of ChemistryUniversity of YorkHeslingtonYorkYO10 5DDUK
- York Biomedical Research InstituteUniversity of YorkHeslingtonYorkYO10 5DDUK
| |
Collapse
|