1
|
Agrez M, Chandler C, Thurecht KJ, Fletcher NL, Liu F, Subramaniam G, Howard CB, Parker S, Turner D, Rzepecka J, Knox G, Nika A, Hall AM, Gooding H, Gallagher L. A novel immunomodulating peptide with potential to complement oligodeoxynucleotide-mediated adjuvanticity in vaccination strategies. Sci Rep 2024; 14:26737. [PMID: 39501043 PMCID: PMC11538426 DOI: 10.1038/s41598-024-78150-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024] Open
Abstract
The identification of adjuvants to improve vaccination efficacy is a major unmet need. One approach is to augment the functionality of dendritic cells (DCs) by using Toll-like receptor-9 (TLR9) agonists such as cytosine-phosphate-guanine oligodeoxynucleotides (CpG ODNs) as adjuvants. Another approach is adjuvant selection based on production of bioactive interleukin-12 (IL-12). We report a D-peptide isomer, designated D-15800, that induces monocyte differentiation to the DC phenotype in vitro and more effectively stimulates IL-12p70 production upon T cell receptor (TCR) activation than the L-isomer. In the absence of TCR activation and either IL-12p70 or interleukin-2 production, only D-15800 activates CD4+ T and natural killer cells. In the presence of CpG ODN, D-15800 synergistically enhances production of interferon-alpha (IFN-α). Taken together with its biostability in human serum and depot retention upon injection, co-delivery of D-15800 with TLR9 agonists could serve to improve vaccine efficacy.
Collapse
Affiliation(s)
- Michael Agrez
- InterK Peptide Therapeutics Limited, Lane Cove West, NSW, Australia.
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia.
| | | | - Kristofer J Thurecht
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Nicholas L Fletcher
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Feifei Liu
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Gayathri Subramaniam
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Christopher B Howard
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Stephen Parker
- InterK Peptide Therapeutics Limited, Lane Cove West, NSW, Australia
| | | | | | - Gavin Knox
- Concept Life Sciences, Edinburgh, Scotland
| | | | | | | | | |
Collapse
|
2
|
Camargo L, Gering I, Mastalipour M, Kraemer-Schulien V, Bujnicki T, Willbold D, Coronado MA, Eberle RJ. A Snake Venom Peptide and Its Derivatives Prevent Aβ 42 Aggregation and Eliminate Toxic Aβ 42 Aggregates In Vitro. ACS Chem Neurosci 2024; 15:2600-2611. [PMID: 38957957 PMCID: PMC11258689 DOI: 10.1021/acschemneuro.4c00089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/28/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024] Open
Abstract
Over a century has passed since Alois Alzheimer first described Alzheimer's disease (AD), and since then, researchers have made significant strides in understanding its pathology. One key feature of AD is the presence of amyloid-β (Aβ) peptides, which form amyloid plaques, and therefore, it is a primary target for treatment studies. Naturally occurring peptides have garnered attention for their potential pharmacological benefits, particularly in the central nervous system. In this study, nine peptide derivatives of Crotamine, a polypeptide from Crotalus durissus terrificus Rattlesnake venom, as well as one d-enantiomer, were evaluated for their ability to modulate Aβ42 aggregation through various assays such as ThT, QIAD, SPR, and sFIDA. All tested peptides were able to decrease Aβ42 aggregation and eliminate Aβ42 aggregates. Additionally, all of the peptides showed an affinity for Aβ42. This study is the first to describe the potential of crotamine derivative peptides against Aβ42 aggregation and to identify a promising d-peptide that could be used as an effective pharmacological tool against AD in the future.
Collapse
Affiliation(s)
- Luana
Cristina Camargo
- Institute
of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich 52428, Germany
- Faculty
of Mathematics and Natural Sciences, Institute of Physical Biology, Heinrich Heine University Düsseldorf, Düsseldorf 40225, Germany
| | - Ian Gering
- Institute
of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich 52428, Germany
| | - Mohammadamin Mastalipour
- Faculty
of Mathematics and Natural Sciences, Institute of Physical Biology, Heinrich Heine University Düsseldorf, Düsseldorf 40225, Germany
| | - Victoria Kraemer-Schulien
- Institute
of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich 52428, Germany
| | - Tuyen Bujnicki
- Institute
of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich 52428, Germany
| | - Dieter Willbold
- Institute
of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich 52428, Germany
- Faculty
of Mathematics and Natural Sciences, Institute of Physical Biology, Heinrich Heine University Düsseldorf, Düsseldorf 40225, Germany
| | - Mônika A. Coronado
- Institute
of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich 52428, Germany
- Faculty
of Mathematics and Natural Sciences, Institute of Physical Biology, Heinrich Heine University Düsseldorf, Düsseldorf 40225, Germany
| | - Raphael J. Eberle
- Institute
of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich 52428, Germany
- Faculty
of Mathematics and Natural Sciences, Institute of Physical Biology, Heinrich Heine University Düsseldorf, Düsseldorf 40225, Germany
| |
Collapse
|
3
|
Discovery of endosomalytic cell-penetrating peptides based on bacterial membrane-targeting sequences. Bioorg Chem 2023; 134:106424. [PMID: 36868126 DOI: 10.1016/j.bioorg.2023.106424] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 02/05/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023]
Abstract
Cell-penetrating peptides (CPPs) are prominent scaffolds for drug developments and related research, particularly the endocytic delivery of biomacromolecules. Effective cargo release from endosomes prior to lysosomal degradation is a crucial step, where the rational design and selection of CPPs remains a challenge and calls for deeper mechanistic understandings. Here, we have investigated a strategy of designing CPPs that selectively disrupt endosomal membranes based on bacterial membrane targeting sequences (MTSs). Six synthesized MTS peptides all exhibit cell-penetrating abilities, among which two d-peptides (d-EcMTS and d-TpMTS) are able to escape from endosomes and localize at ER after entering the cell. The utility of this strategy has been demonstrated by the intracellular delivery of green fluorescent protein (GFP). Together, these results suggest that the large pool of bacterial MTSs may be a rich source for the development of novel CPPs.
Collapse
|
4
|
Gueta O, Amiram M. Expanding the chemical repertoire of protein-based polymers for drug-delivery applications. Adv Drug Deliv Rev 2022; 190:114460. [PMID: 36030987 DOI: 10.1016/j.addr.2022.114460] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/12/2022] [Indexed: 01/24/2023]
Abstract
Expanding the chemical repertoire of natural and artificial protein-based polymers (PBPs) can enable the production of sequence-defined, yet chemically diverse, biopolymers with customized or new properties that cannot be accessed in PBPs composed of only natural amino acids. Various approaches can enable the expansion of the chemical repertoire of PBPs, including chemical and enzymatic treatments or the incorporation of unnatural amino acids. These techniques are employed to install a wide variety of chemical groups-such as bio-orthogonally reactive, cross-linkable, post-translation modifications, and environmentally responsive groups-which, in turn, can facilitate the design of customized PBP-based drug-delivery systems with modified, fine-tuned, or entirely new properties and functions. Here, we detail the existing and emerging technologies for expanding the chemical repertoire of PBPs and review several chemical groups that either demonstrate or are anticipated to show potential in the design of PBP-based drug delivery systems. Finally, we provide our perspective on the remaining challenges and future directions in this field.
Collapse
Affiliation(s)
- Osher Gueta
- The Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel
| | - Miriam Amiram
- The Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel.
| |
Collapse
|
5
|
Collett CF, Phillips HC, Fisher M, Smith S, Fenn C, Goodwin P, Morphew RM, Brophy PM. Fasciola hepatica Cathepsin L Zymogens: Immuno-Proteomic Evidence for Highly Immunogenic Zymogen-Specific Conformational Epitopes to Support Diagnostics Development. J Proteome Res 2022; 21:1997-2010. [PMID: 35849550 PMCID: PMC9361350 DOI: 10.1021/acs.jproteome.2c00299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Fasciola hepatica, the common liver fluke and causative agent of zoonotic fasciolosis, impacts on food security with global economic losses of over $3.2 BN per annum through deterioration of animal health, productivity losses, and livestock death and is also re-emerging as a foodborne human disease. Cathepsin proteases present a major vaccine and diagnostic target of the F. hepatica excretory/secretory (ES) proteome, but utilization in diagnostics of the highly antigenic zymogen stage of these proteins is surprisingly yet to be fully exploited. Following an immuno-proteomic investigation of recombinant and native procathepsins ((r)FhpCL1), including mass spectrometric analyses (DOI: 10.6019/PXD030293), and using counterpart polyclonal antibodies to a recombinant mutant procathepsin L (anti-rFhΔpCL1), we have confirmed recombinant and native cathepsin L zymogens contain conserved, highly antigenic epitopes that are conformationally dependent. Furthermore, using diagnostic platforms, including pilot serum and fecal antigen capture enzyme-linked immunosorbent assay (ELISA) tests, the diagnostic capacities of cathepsin L zymogens were assessed and validated, offering promising efficacy as markers of infection and for monitoring treatment efficacy.
Collapse
Affiliation(s)
- Clare F Collett
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, U.K
| | - Helen C Phillips
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, U.K
| | - Maggie Fisher
- Ridgeway Research Ltd., Park Farm Buildings, Park Lane, St. Briavels, Gloucestershire GL15 6QX, U.K
| | - Sian Smith
- Ridgeway Research Ltd., Park Farm Buildings, Park Lane, St. Briavels, Gloucestershire GL15 6QX, U.K
| | - Caroline Fenn
- Ridgeway Research Ltd., Park Farm Buildings, Park Lane, St. Briavels, Gloucestershire GL15 6QX, U.K
| | - Phil Goodwin
- Bio-Check UK, Spectrum House, Llys Edmund Prys, St. Asaph Business Park, St. Asaph, Denbighshire LL17 0LJ, U.K
| | - Russell M Morphew
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, U.K
| | - Peter M Brophy
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, U.K
| |
Collapse
|
6
|
Design of D-Amino Acids SARS-CoV-2 Main Protease Inhibitors Using the Cationic Peptide from Rattlesnake Venom as a Scaffold. Pharmaceuticals (Basel) 2022; 15:ph15050540. [PMID: 35631367 PMCID: PMC9146215 DOI: 10.3390/ph15050540] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/18/2022] [Accepted: 04/25/2022] [Indexed: 12/15/2022] Open
Abstract
The C30 endopeptidase (3C-like protease; 3CLpro) is essential for the life cycle of SARS-CoV-2 (severe acute respiratory syndrome-coronavirus-2) since it plays a pivotal role in viral replication and transcription and, hence, is a promising drug target. Molecules isolated from animals, insects, plants, or microorganisms can serve as a scaffold for the design of novel biopharmaceutical products. Crotamine, a small cationic peptide from the venom of the rattlesnake Crotalus durissus terrificus, has been the focus of many studies since it exhibits activities such as analgesic, in vitro antibacterial, and hemolytic activities. The crotamine derivative L-peptides (L-CDP) that inhibit the 3CL protease in the low µM range were examined since they are susceptible to proteolytic degradation; we explored the utility of their D-enantiomers form. Comparative uptake inhibition analysis showed D-CDP as a promising prototype for a D-peptide-based drug. We also found that the D-peptides can impair SARS-CoV-2 replication in vivo, probably targeting the viral protease 3CLpro.
Collapse
|
7
|
Rai R, Sureshan KM. Topochemical Synthesis of a Heterochiral Peptide Polymer in Different Polymorphic Forms from Crystals and Aerogels. Angew Chem Int Ed Engl 2022; 61:e202111623. [DOI: 10.1002/anie.202111623] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Indexed: 12/16/2022]
Affiliation(s)
- Rishika Rai
- School of Chemistry Indian Institute of Science Education and Research Thiruvananthapuram Kerala 695551 India
| | - Kana M. Sureshan
- School of Chemistry Indian Institute of Science Education and Research Thiruvananthapuram Kerala 695551 India
| |
Collapse
|
8
|
Sureshan KM, Rai R. Topochemical Synthesis of a Heterochiral Peptide Polymer in Different Polymorphic Forms from Crystals and Aerogels. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202111623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Kana M Sureshan
- Indian Institute of Science Education and Research Thiruvananthapuram School of Chemistry ThiruvananthapuramMaruthamalaVithura 695551 Thiruvananthapuram INDIA
| | - Rishika Rai
- IISER-TVM: Indian Institute of Science Education Research Thiruvananthapuram Chemistry 695551 Thiruvananthapuram INDIA
| |
Collapse
|
9
|
Preston GW. Different directions for retro-inverso peptides. J Pept Sci 2021; 28:e3384. [PMID: 34889485 DOI: 10.1002/psc.3384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/11/2021] [Accepted: 11/11/2021] [Indexed: 11/07/2022]
Abstract
Retro-inverso peptides are reversed sequences of mirror-image amino acid residues. Synthetic molecules of this type have long been considered potential mimics of functional peptides. This Peptide Highlights article examines some recent applications of the retro-inverso transformation at the interface of peptide chemistry and medicine.
Collapse
Affiliation(s)
- George W Preston
- Stoller Biomarker Discovery Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Stem Cell and Leukaemia Proteomics Laboratory, Manchester Cancer Research Centre, The University of Manchester, Manchester, UK
| |
Collapse
|
10
|
Zhang M, Wang Y, Li X, Meng G, Chen X, Wang L, Lin Z, Wang L. A Single L/D-Substitution at Q4 of the mInsA 2-10 Epitope Prevents Type 1 Diabetes in Humanized NOD Mice. Front Immunol 2021; 12:713276. [PMID: 34526989 PMCID: PMC8435724 DOI: 10.3389/fimmu.2021.713276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 08/09/2021] [Indexed: 11/13/2022] Open
Abstract
Autoreactive CD8+ T cells play an indispensable key role in the destruction of pancreatic islet β-cells and the initiation of type 1 diabetes (T1D). Insulin is an essential β-cell autoantigen in T1D. An HLA-A*0201-restricted epitope of insulin A chain (mInsA2-10) is an immunodominant ligand for autoreactive CD8+ T cells in NOD.β2mnull .HHD mice. Altered peptide ligands (APLs) carrying amino acid substitutions at T cell receptor (TCR) contact positions within an epitope are potential to modulate autoimmune responses via triggering altered TCR signaling. Here, we used a molecular simulation strategy to guide the generation of APL candidates by substitution of L-amino acids with D-amino acids at potential TCR contact residues (positions 4 and 6) of mInsA2-10, named mInsA2-10DQ4 and mInsA2-10DC6, respectively. We found that administration of mInsA2-10DQ4, but not DC6, significantly suppressed the development of T1D in NOD.β2mnull .HHD mice. Mechanistically, treatment with mInsA2-10DQ4 not only notably eliminated mInsA2-10 autoreactive CD8+ T cell responses but also prevented the infiltration of CD4+ T and CD8+ T cells, as well as the inflammatory responses in the pancreas of NOD.β2mnull.HHD mice. This study provides a new strategy for the development of APL vaccines for T1D prevention.
Collapse
Affiliation(s)
- Mengjun Zhang
- Department of Pharmaceutical Analysis, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, China.,Institute of Immunology People's Libration Army (PLA) & Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yuanqiang Wang
- Department of Pharmaceutical Engineering, School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Xiangqian Li
- Institute of Immunology People's Libration Army (PLA) & Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Gang Meng
- Department of Pathology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaoling Chen
- Institute of Immunology People's Libration Army (PLA) & Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lina Wang
- Department of Immunology, Weifang Medical University, Weifang, China
| | - Zhihua Lin
- Department of Pharmaceutical Engineering, School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Li Wang
- Institute of Immunology People's Libration Army (PLA) & Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
11
|
In Vitro and In Vivo Efficacies of the Linear and the Cyclic Version of an All-d-Enantiomeric Peptide Developed for the Treatment of Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22126553. [PMID: 34207233 PMCID: PMC8234218 DOI: 10.3390/ijms22126553] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 11/17/2022] Open
Abstract
Multiple sources of evidence suggest that soluble amyloid β (Aβ)-oligomers are responsible for the development and progression of Alzheimer’s disease (AD). In order to specifically eliminate these toxic Aβ-oligomers, our group has developed a variety of all-d-peptides over the past years. One of them, RD2, has been intensively studied and showed such convincing in vitro and in vivo properties that it is currently in clinical trials. In order to further optimize the compounds and to elucidate the characteristics of therapeutic d-peptides, several rational drug design approaches have been performed. Two of these d-peptides are the linear tandem (head-to-tail) d-peptide RD2D3 and its cyclized form cRD2D3. Tandemization and cyclization should result in an increased in vitro potency and increase pharmacokinetic properties, especially crossing the blood–brain-barrier. In comparison, cRD2D3 showed a superior pharmacokinetic profile to RD2D3. This fact suggests that higher efficacy can be achieved in vivo at equally administered concentrations. To prove this hypothesis, we first established the in vitro profile of both d-peptides here. Subsequently, we performed an intraperitoneal treatment study. This study failed to provide evidence that cRD2D3 is superior to RD2D3 in vivo as in some tests cRD2D3 failed to show equal or higher efficacy.
Collapse
|
12
|
Gibadullin R, Randall CJ, Sidney J, Sette A, Gellman SH. Backbone Modifications of HLA-A2-Restricted Antigens Induce Diverse Binding and T Cell Activation Outcomes. J Am Chem Soc 2021; 143:6470-6481. [PMID: 33881854 DOI: 10.1021/jacs.1c00016] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
CD8+ T cells express T cell receptors (TCRs) that recognize short peptide antigens in the context of major histocompatibility class I (MHC I) molecules. This recognition process produces an array of cytokine-mediated signals that help to govern immunological responses. Design of biostable MHC I peptide vaccines containing unnatural subunits is desirable, and synthetic antigens in which a native α-amino acid residue is replaced by a homologous β-amino acid residue (native side chain but extended backbone) might be useful in this regard. We have evaluated the impact of α-to-β backbone modification at a single site on T cell-mediated recognition of six clinically important viral and tumor-associated antigens bound to an MHC I. Effects of this modification on MHC I affinity and T cell activation were measured. Many of these modifications diminish or prevent T cell response. However, a number of α/β-peptide antigens were found to mimic the activity of natural antigens or to enhance maximal T cell response, as measured by interferon-γ release. Results from this broad exploratory study advance our understanding of immunological responses to antigens bearing unnatural modifications and suggest that α/β-peptides could be a source of potent and proteolytically stable variants of native antigens.
Collapse
Affiliation(s)
- Ruslan Gibadullin
- Department of Chemistry, University of Wisconsin, 1101 University Avenue, Madison, Wisconsin 53706, United States
| | - Caleb J Randall
- Department of Chemistry, University of Wisconsin, 1101 University Avenue, Madison, Wisconsin 53706, United States
| | - John Sidney
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, California 92037, United States
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, California 92037, United States.,Department of Medicine, University of California, San Diego, California 92093, United States
| | - Samuel H Gellman
- Department of Chemistry, University of Wisconsin, 1101 University Avenue, Madison, Wisconsin 53706, United States
| |
Collapse
|
13
|
Wesseling CMJ, Wood TM, Slingerland CJ, Bertheussen K, Lok S, Martin NI. Thrombin-Derived Peptides Potentiate the Activity of Gram-Positive-Specific Antibiotics against Gram-Negative Bacteria. Molecules 2021; 26:molecules26071954. [PMID: 33808488 PMCID: PMC8037310 DOI: 10.3390/molecules26071954] [Citation(s) in RCA: 3] [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: 03/14/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 12/13/2022] Open
Abstract
The continued rise of antibiotic resistance threatens to undermine the utility of the world’s current antibiotic arsenal. This problem is particularly troubling when it comes to Gram-negative pathogens for which there are inherently fewer antibiotics available. To address this challenge, recent attention has been focused on finding compounds capable of disrupting the Gram-negative outer membrane as a means of potentiating otherwise Gram-positive-specific antibiotics. In this regard, agents capable of binding to the lipopolysaccharide (LPS) present in the Gram-negative outer membrane are of particular interest as synergists. Recently, thrombin-derived C-terminal peptides (TCPs) were reported to exhibit unique LPS-binding properties. We here describe investigations establishing the capacity of TCPs to act as synergists with the antibiotics erythromycin, rifampicin, novobiocin, and vancomycin against multiple Gram-negative strains including polymyxin-resistant clinical isolates. We further assessed the structural features most important for the observed synergy and characterized the outer membrane permeabilizing activity of the most potent synergists. Our investigations highlight the potential for such peptides in expanding the therapeutic range of antibiotics typically only used to treat Gram-positive infections.
Collapse
Affiliation(s)
- Charlotte M. J. Wesseling
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2333 Leiden, The Netherlands; (C.M.J.W.); (T.M.W.); (C.J.S.); (K.B.); (S.L.)
| | - Thomas M. Wood
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2333 Leiden, The Netherlands; (C.M.J.W.); (T.M.W.); (C.J.S.); (K.B.); (S.L.)
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 Utrecht, The Netherlands
| | - Cornelis J. Slingerland
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2333 Leiden, The Netherlands; (C.M.J.W.); (T.M.W.); (C.J.S.); (K.B.); (S.L.)
| | - Kristine Bertheussen
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2333 Leiden, The Netherlands; (C.M.J.W.); (T.M.W.); (C.J.S.); (K.B.); (S.L.)
- Bio-Organic Synthesis Group, Leiden Institute of Chemistry, Leiden University, 2333 Leiden, The Netherlands
| | - Samantha Lok
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2333 Leiden, The Netherlands; (C.M.J.W.); (T.M.W.); (C.J.S.); (K.B.); (S.L.)
| | - Nathaniel I. Martin
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2333 Leiden, The Netherlands; (C.M.J.W.); (T.M.W.); (C.J.S.); (K.B.); (S.L.)
- Correspondence:
| |
Collapse
|
14
|
Dyakin VV, Wisniewski TM, Lajtha A. Racemization in Post-Translational Modifications Relevance to Protein Aging, Aggregation and Neurodegeneration: Tip of the Iceberg. Symmetry (Basel) 2021; 13:455. [PMID: 34350031 PMCID: PMC8330555 DOI: 10.3390/sym13030455] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Homochirality of DNA and prevalent chirality of free and protein-bound amino acids in a living organism represents the challenge for modern biochemistry and neuroscience. The idea of an association between age-related disease, neurodegeneration, and racemization originated from the studies of fossils and cataract disease. Under the pressure of new results, this concept has a broader significance linking protein folding, aggregation, and disfunction to an organism's cognitive and behavioral functions. The integrity of cognitive function is provided by a delicate balance between the evolutionarily imposed molecular homo-chirality and the epigenetic/developmental impact of spontaneous and enzymatic racemization. The chirality of amino acids is the crucial player in the modulation the structure and function of proteins, lipids, and DNA. The collapse of homochirality by racemization is the result of the conformational phase transition. The racemization of protein-bound amino acids (spontaneous and enzymatic) occurs through thermal activation over the energy barrier or by the tunnel transfer effect under the energy barrier. The phase transition is achieved through the intermediate state, where the chirality of alpha carbon vanished. From a thermodynamic consideration, the system in the homo-chiral (single enantiomeric) state is characterized by a decreased level of entropy. The oscillating protein chirality is suggesting its distinct significance in the neurotransmission and flow of perceptual information, adaptive associative learning, and cognitive laterality. The common pathological hallmarks of neurodegenerative disorders include protein misfolding, aging, and the deposition of protease-resistant protein aggregates. Each of the landmarks is influenced by racemization. The brain region, cell type, and age-dependent racemization critically influence the functions of many intracellular, membrane-bound, and extracellular proteins including amyloid precursor protein (APP), TAU, PrP, Huntingtin, α-synuclein, myelin basic protein (MBP), and collagen. The amyloid cascade hypothesis in Alzheimer's disease (AD) coexists with the failure of amyloid beta (Aβ) targeting drug therapy. According to our view, racemization should be considered as a critical factor of protein conformation with the potential for inducing order, disorder, misfolding, aggregation, toxicity, and malfunctions.
Collapse
Affiliation(s)
- Victor V. Dyakin
- Virtual Reality Perception Lab (VRPL), The Nathan S. Kline Institute for Psychiatric Research (NKI), Orangeburg, NY 10962, USA
| | - Thomas M. Wisniewski
- Departments of Neurology, Pathology and Psychiatry, Center for Cognitive Neurology, New York University School of Medicine, New York, NY 10016, USA
| | - Abel Lajtha
- Center for Neurochemistry, The Nathan S. Kline Institute for Psychiatric Research (NKI), Orangeburg, NY 10962, USA
| |
Collapse
|
15
|
Zhang X, Zhang X, Zhong M, Zhao P, Guo C, Li Y, Wang T, Gao H. Selection of a d-Enantiomeric Peptide Specifically Binding to PHF6 for Inhibiting Tau Aggregation in Transgenic Mice. ACS Chem Neurosci 2020; 11:4240-4253. [PMID: 33284003 DOI: 10.1021/acschemneuro.0c00518] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Tauopathies refer to a group of neurodegenerative disorders caused by the accumulation of insoluble hyperphosphorylated Tau protein in the brain. The inhibition and interruption of Tau aggregation are considered important strategies to ameliorate the neurodegenerative process. Previous work has shown that hexapeptide 306VQIVYK311 (PHF6) located in the repeat domain 3 of Tau protein drives Tau aggregation and itself forms a β-sheet structure similar to those of Tau-oligomers and neurofibrillary tangles (NFTs). In this study, a mirror image phage display technology was used to screen protease-resistant and low-immunogenic d-enantiomeric peptides for their capacity to inhibit Tau aggregation. Following the preparation of d-enantiomeric PHF6 fibrils and M13 phage peptide library biopanning, 7 sets of high specificity peptides were obtained. Through ELISA and competition inhibition assays, we chose a highly specific peptide p-NH with the sequence N-I-T-M-N-S-R-R-R-R-N-H. The molecular docking results showed that p-NH interacted with PHF6 fibrils mainly through van der Waals forces and hydrogen bonding and could inhibit PHF6 aggregation in a d-configuration and concentration-dependent manner. In vitro, p-NH prohibited the formation of PHF6 fibrils and was able to enter into mouse neuroblastoma N2a cells (N2a cells) to inhibit Tau hyperphosphorylation and aggregation. Intranasal administration of p-NH reduced NFTs and improved the cognitive ability of TauP301S transgenic mice. These findings represent a straightforward methodology to find therapeutic peptides with potential applications in tauopathies.
Collapse
Affiliation(s)
- Xiancheng Zhang
- College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Xiaoyu Zhang
- College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Manli Zhong
- College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Pu Zhao
- College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Chuang Guo
- College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - You Li
- College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Tao Wang
- College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Huiling Gao
- College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry, Northeastern University, Ministry of Education, Shenyang 110819, China
| |
Collapse
|
16
|
Robson B. Techniques assisting peptide vaccine and peptidomimetic design. Sidechain exposure in the SARS-CoV-2 spike glycoprotein. Comput Biol Med 2020; 128:104124. [PMID: 33276271 PMCID: PMC7679524 DOI: 10.1016/j.compbiomed.2020.104124] [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: 09/16/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 01/15/2023]
Abstract
The aim of the present study is to discuss the design of peptide vaccines and peptidomimetics against SARS-COV-2, to develop and apply a method of protein structure analysis that is particularly appropriate to applying and discussing such design, and also to use that method to summarize some important features of the SARS-COV-2 spike protein sequence. A tool for assessing sidechain exposure in the SARS-CoV-2 spike glycoprotein is described. It extends to assessing accessibility of sidechains by considering several different three-dimensional structure determinations of SARS-CoV-2 and SARS-CoV-1 spike protein. The method is designed to be insensitive to a distance limit for counting neighboring atoms and the results are in good agreement with the physical chemical properties and exposure trends of the 20 naturally occurring sidechains. The spike protein sequence is analyzed with comment regarding exposable character. It includes studies of complexes with antibody elements and ACE2. These indicate changes in exposure at sites remote to those at which the antibody binds. They are of interest concerning design of synthetic peptide vaccines, and for peptidomimetics as a basis of drug discovery. The method was also developed in order to provide linear (one-dimensional) information that can be used along with other bioinformatics data of this kind in data mining and machine learning, potentially as genomic data regarding protein polymorphisms to be combined with more traditional clinical data. Bioinformatics studies are carried out on SARS-CoV-2 spike, studying solvent exposure. The methods are particularly suited for synthetic vaccines and d-amino acid peptidomimetics. Methods of generating d-amino acid peptidomimetics are described and reviewed. The effect of antibody binding in stabilizing loop conformation and exposing remote sites is noted.
Collapse
Affiliation(s)
- B Robson
- Ingine Inc. Cleveland Ohio USA and the Dirac Foundation, Oxfordshire, UK.
| |
Collapse
|
17
|
Rai J. Peptide and protein mimetics by retro and retroinverso analogs. Chem Biol Drug Des 2019; 93:724-736. [PMID: 30582286 DOI: 10.1111/cbdd.13472] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/10/2018] [Accepted: 12/16/2018] [Indexed: 12/19/2022]
Abstract
Retroinverso analog of a natural polypeptide can sometimes mimic the structure and function of the natural peptide. The additional advantage of using retroinverso analog is that it is resistant to proteolysis. The retroinverso analogs have peptide sequence in reverse direction with respect to natural peptide and also have chirality of amino acid inverted from L to D. The D amino acids cannot be recognized by common proteases of the body; therefore, these peptides will not be degraded easily and have a longer-lasting effect as vaccine and inhibitor drugs. There have been many contested propositions about the geometric relationship between a peptide and its retro, inverso, or retroinverso analog. A retroinverso analog sometimes fails to adopt the structure that can mimic the function of the natural peptide. In such cases, partial retroinverso analog and other modifications can help in achieving the desired structure and function. Here, we review the theory, major experimental attempts, prediction methods, and alternative strategies related to retroinverso peptidomimetics.
Collapse
|
18
|
Rösener NS, Gremer L, Reinartz E, König A, Brener O, Heise H, Hoyer W, Neudecker P, Willbold D. A d-enantiomeric peptide interferes with heteroassociation of amyloid-β oligomers and prion protein. J Biol Chem 2018; 293:15748-15764. [PMID: 30131337 PMCID: PMC6187637 DOI: 10.1074/jbc.ra118.003116] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/17/2018] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects millions of people worldwide. One AD hallmark is the aggregation of β-amyloid (Aβ) into soluble oligomers and insoluble fibrils. Several studies have reported that oligomers rather than fibrils are the most toxic species in AD progression. Aβ oligomers bind with high affinity to membrane-associated prion protein (PrP), leading to toxic signaling across the cell membrane, which makes the Aβ-PrP interaction an attractive therapeutic target. Here, probing this interaction in more detail, we found that both full-length, soluble human (hu) PrP(23-230) and huPrP(23-144), lacking the globular C-terminal domain, bind to Aβ oligomers to form large complexes above the megadalton size range. Following purification by sucrose density-gradient ultracentrifugation, the Aβ and huPrP contents in these heteroassemblies were quantified by reversed-phase HPLC. The Aβ:PrP molar ratio in these assemblies exhibited some limited variation depending on the molar ratio of the initial mixture. Specifically, a molar ratio of about four Aβ to one huPrP in the presence of an excess of huPrP(23-230) or huPrP(23-144) suggested that four Aβ units are required to form one huPrP-binding site. Of note, an Aβ-binding all-d-enantiomeric peptide, RD2D3, competed with huPrP for Aβ oligomers and interfered with Aβ-PrP heteroassembly in a concentration-dependent manner. Our results highlight the importance of multivalent epitopes on Aβ oligomers for Aβ-PrP interactions and have yielded an all-d-peptide-based, therapeutically promising agent that competes with PrP for these interactions.
Collapse
Affiliation(s)
- Nadine S Rösener
- From the Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany and
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Lothar Gremer
- From the Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany and
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Elke Reinartz
- From the Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany and
| | - Anna König
- From the Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany and
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Oleksandr Brener
- From the Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany and
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Henrike Heise
- From the Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany and
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Wolfgang Hoyer
- From the Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany and
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Philipp Neudecker
- From the Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany and
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Dieter Willbold
- From the Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany and
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| |
Collapse
|
19
|
Miles JJ, Tan MP, Dolton G, Edwards ES, Galloway SA, Laugel B, Clement M, Makinde J, Ladell K, Matthews KK, Watkins TS, Tungatt K, Wong Y, Lee HS, Clark RJ, Pentier JM, Attaf M, Lissina A, Ager A, Gallimore A, Rizkallah PJ, Gras S, Rossjohn J, Burrows SR, Cole DK, Price DA, Sewell AK. Peptide mimic for influenza vaccination using nonnatural combinatorial chemistry. J Clin Invest 2018; 128:1569-1580. [PMID: 29528337 PMCID: PMC5873848 DOI: 10.1172/jci91512] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/18/2018] [Indexed: 01/11/2023] Open
Abstract
Polypeptide vaccines effectively activate human T cells but suffer from poor biological stability, which confines both transport logistics and in vivo therapeutic activity. Synthetic biology has the potential to address these limitations through the generation of highly stable antigenic "mimics" using subunits that do not exist in the natural world. We developed a platform based on D-amino acid combinatorial chemistry and used this platform to reverse engineer a fully artificial CD8+ T cell agonist that mirrored the immunogenicity profile of a native epitope blueprint from influenza virus. This nonnatural peptide was highly stable in human serum and gastric acid, reflecting an intrinsic resistance to physical and enzymatic degradation. In vitro, the synthetic agonist stimulated and expanded an archetypal repertoire of polyfunctional human influenza virus-specific CD8+ T cells. In vivo, specific responses were elicited in naive humanized mice by subcutaneous vaccination, conferring protection from subsequent lethal influenza challenge. Moreover, the synthetic agonist was immunogenic after oral administration. This proof-of-concept study highlights the power of synthetic biology to expand the horizons of vaccine design and therapeutic delivery.
Collapse
Affiliation(s)
- John J. Miles
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Griffith University, Brisbane, Queensland, Australia
| | - Mai Ping Tan
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Garry Dolton
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
- Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Emily S.J. Edwards
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Sarah A.E. Galloway
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Bruno Laugel
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Mathew Clement
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Julia Makinde
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Kristin Ladell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
- Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | | | - Thomas S. Watkins
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Katie Tungatt
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Yide Wong
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Han Siean Lee
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Richard J. Clark
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Johanne M. Pentier
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Meriem Attaf
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Anya Lissina
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Ann Ager
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Awen Gallimore
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Pierre J. Rizkallah
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Stephanie Gras
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, and
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia
| | - Jamie Rossjohn
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, and
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia
| | - Scott R. Burrows
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - David K. Cole
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - David A. Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
- Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Andrew K. Sewell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
- Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
20
|
Elfgen A, Santiago-Schübel B, Gremer L, Kutzsche J, Willbold D. Surprisingly high stability of the Aβ oligomer eliminating all-d-enantiomeric peptide D3 in media simulating the route of orally administered drugs. Eur J Pharm Sci 2017; 107:203-207. [PMID: 28711713 DOI: 10.1016/j.ejps.2017.07.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 11/27/2022]
Abstract
The aggregation of the amyloid β protein (Aβ) plays an important role in the pathology of Alzheimer's disease. Previously, we have developed the all-d-enantiomeric peptide D3, which is able to eliminate neurotoxic Aβ oligomers in vitro and improve cognition in a transgenic Alzheimer's disease mouse model in vivo even after oral administration. d-Peptides are expected to be more resistant against enzymatic proteolysis compared to their l-enantiomeric equivalents, and indeed, a pharmacokinetic study with tritiated D3 revealed the oral bioavailability to be about 58%. To further investigate the underlying properties, we examined the stability of D3 in comparison to its corresponding all-l-enantiomeric mirror image l-D3 in media simulating the gastrointestinal tract, blood and liver. Potential metabolization was followed by reversed-phase high-performance liquid chromatography. In simulated gastric fluid, D3 remained almost completely stable (89%) within 24h, while 70% of l-D3 was degraded within the same time period. Notably, in simulated intestinal fluid, D3 also remained stable (96%) for 24h, whereas l-D3 was completely metabolized within seconds. In human plasma and human liver microsomes, l-D3 was metabolized several hundred times faster than D3. The remarkably high stability may explain the high oral bioavailability seen in previous studies allowing oral administration of the drug candidate. Thus, all-d-enantiomeric peptides may represent a promising new compound class for drug development.
Collapse
Affiliation(s)
- Anne Elfgen
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Research Center Jülich, 52428 Jülich, Germany
| | - Beatrix Santiago-Schübel
- Central Institute for Engineering, Electronics and Analytics (ZEA-3), Research Center Jülich, 52428 Jülich, Germany
| | - Lothar Gremer
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Research Center Jülich, 52428 Jülich, Germany; Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Janine Kutzsche
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Research Center Jülich, 52428 Jülich, Germany
| | - Dieter Willbold
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Research Center Jülich, 52428 Jülich, Germany; Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany.
| |
Collapse
|
21
|
Uppalapati M, Lee DJ, Mandal K, Li H, Miranda LP, Lowitz J, Kenney J, Adams JJ, Ault-Riché D, Kent SBH, Sidhu SS. A Potent d-Protein Antagonist of VEGF-A is Nonimmunogenic, Metabolically Stable, and Longer-Circulating in Vivo. ACS Chem Biol 2016; 11:1058-65. [PMID: 26745345 DOI: 10.1021/acschembio.5b01006] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Polypeptides composed entirely of d-amino acids and the achiral amino acid glycine (d-proteins) inherently have in vivo properties that are proposed to be near-optimal for a large molecule therapeutic agent. Specifically, d-proteins are resistant to degradation by proteases and are anticipated to be nonimmunogenic. Furthermore, d-proteins are manufactured chemically and can be engineered to have other desirable properties, such as improved stability, affinity, and pharmacokinetics. Thus, a well-designed d-protein therapeutic would likely have significant advantages over l-protein drugs. Toward the goal of developing d-protein therapeutics, we previously generated RFX001.D, a d-protein antagonist of natural vascular endothelial growth factor A (VEGF-A) that inhibited binding to its receptor. However, RFX001.D is unstable at physiological temperatures (Tm = 33 °C). Here, we describe RFX037.D, a variant of RFX001.D with extreme thermal stability (Tm > 95 °C), high affinity for VEGF-A (Kd = 6 nM), and improved receptor blocking. Comparison of the two enantiomeric forms of RFX037 revealed that the d-protein is more stable in mouse, monkey, and human plasma and has a longer half-life in vivo in mice. Significantly, RFX037.D was nonimmunogenic in mice, whereas the l-enantiomer generated a strong immune response. These results confirm the potential utility of synthetic d-proteins as alternatives to therapeutic antibodies.
Collapse
Affiliation(s)
- Maruti Uppalapati
- Banting
and Best Department of Medical Research and Department of Molecular
Genetics, the Donnelly Centre, University of Toronto, Toronto, Ontario, Canada M5S 3E1
| | - Dong Jun Lee
- Department
of Chemistry; Department of Biochemistry and Molecular Biology, Institute
for Biophysical Dynamics, University of Chicago, Chicago, Illinois 60637, United States
| | - Kalyaneswar Mandal
- Department
of Chemistry; Department of Biochemistry and Molecular Biology, Institute
for Biophysical Dynamics, University of Chicago, Chicago, Illinois 60637, United States
| | - Hongyan Li
- Pharmacokinetics & Drug Metabolism, Amgen, Inc., Thousand Oaks, California 91320, United States
| | - Les P. Miranda
- Therapeutic Discovery, Amgen, Inc., Thousand
Oaks, California 91320, United States
| | - Joshua Lowitz
- Antibody Solutions, Sunnyvale, California 94089, United States
| | - John Kenney
- Antibody Solutions, Sunnyvale, California 94089, United States
| | - Jarrett J. Adams
- Banting
and Best Department of Medical Research and Department of Molecular
Genetics, the Donnelly Centre, University of Toronto, Toronto, Ontario, Canada M5S 3E1
| | - Dana Ault-Riché
- Reflexion Pharmaceuticals, Inc., San Francisco, California 94104, United States
| | - Stephen B. H. Kent
- Department
of Chemistry; Department of Biochemistry and Molecular Biology, Institute
for Biophysical Dynamics, University of Chicago, Chicago, Illinois 60637, United States
| | - Sachdev S. Sidhu
- Banting
and Best Department of Medical Research and Department of Molecular
Genetics, the Donnelly Centre, University of Toronto, Toronto, Ontario, Canada M5S 3E1
| |
Collapse
|
22
|
Leithold LHE, Jiang N, Post J, Niemietz N, Schartmann E, Ziehm T, Kutzsche J, Shah NJ, Breitkreutz J, Langen KJ, Willuweit A, Willbold D. Pharmacokinetic properties of tandem d-peptides designed for treatment of Alzheimer's disease. Eur J Pharm Sci 2016; 89:31-8. [PMID: 27086111 DOI: 10.1016/j.ejps.2016.04.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 04/06/2016] [Accepted: 04/12/2016] [Indexed: 11/15/2022]
Abstract
Peptides are more and more considered for the development of drug candidates. However, they frequently exhibit severe disadvantages such as instability and unfavourable pharmacokinetic properties. Many peptides are rapidly cleared from the organism and oral bioavailabilities as well as in vivo half-lives often remain low. In contrast, some peptides consisting solely of d-enantiomeric amino acid residues were shown to combine promising therapeutic properties with high proteolytic stability and enhanced pharmacokinetic parameters. Recently, we have shown that D3 and RD2 have highly advantageous pharmacokinetic properties. Especially D3 has already proven promising properties suitable for treatment of Alzheimer's disease. Here, we analyse the pharmacokinetic profiles of D3D3 and RD2D3, which are head-to-tail tandem d-peptides built of D3 and its derivative RD2. Both D3D3 and RD2D3 show proteolytic stability in mouse plasma and organ homogenates for at least 24h and in murine and human liver microsomes for 4h. Notwithstanding their high affinity to plasma proteins, both peptides are taken up into the brain following i.v. as well as i.p. administration. Although both peptides contain identical d-amino acid residues, they are arranged in a different sequence order and the peptides show differences in pharmacokinetic properties. After i.p. administration RD2D3 exhibits lower plasma clearance and higher bioavailability than D3D3. We therefore concluded that the amino acid sequence of RD2 leads to more favourable pharmacokinetic properties within the tandem peptide, which underlines the importance of particular sequence motifs, even in short peptides, for the design of further therapeutic d-peptides.
Collapse
Affiliation(s)
- Leonie H E Leithold
- Institute of Complex Systems, Structural Biochemistry, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.
| | - Nan Jiang
- Institute of Complex Systems, Structural Biochemistry, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.
| | - Julia Post
- Institute of Complex Systems, Structural Biochemistry, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.
| | - Nicole Niemietz
- Institute of Neuroscience and Medicine, Medical Imaging Physics, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.
| | - Elena Schartmann
- Institute of Complex Systems, Structural Biochemistry, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.
| | - Tamar Ziehm
- Institute of Complex Systems, Structural Biochemistry, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.
| | - Janine Kutzsche
- Institute of Complex Systems, Structural Biochemistry, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.
| | - N Jon Shah
- Institute of Neuroscience and Medicine, Medical Imaging Physics, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany; Department of Neurology, Universitätsklinikum der RWTH Aachen, 52074 Aachen, Germany.
| | - Jörg Breitkreutz
- Institute of Pharmaceutics and Biopharmaceutics, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany.
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine, Medical Imaging Physics, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany; Department of Nuclear Medicine, Universitätsklinikum der RWTH Aachen, 52074 Aachen, Germany.
| | - Antje Willuweit
- Institute of Neuroscience and Medicine, Medical Imaging Physics, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.
| | - Dieter Willbold
- Institute of Complex Systems, Structural Biochemistry, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany; Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany.
| |
Collapse
|
23
|
Babinska A, Clement CC, Swiatkowska M, Szymanski J, Shon A, Ehrlich YH, Kornecki E, Salifu MO. Development of new antiatherosclerotic and antithrombotic drugs utilizing F11 receptor (F11R/JAM-A) peptides. Biopolymers 2016; 102:322-34. [PMID: 24801754 DOI: 10.1002/bip.22503] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 04/01/2014] [Accepted: 05/02/2014] [Indexed: 12/29/2022]
Abstract
Peptides with enhanced resistance to proteolysis, based on the amino acid sequence of the F11 receptor molecule (F11R, aka JAM-A/Junctional adhesion molecule-A), were designed, prepared, and examined as potential candidates for the development of anti-atherosclerotic and anti-thrombotic therapeutic drugs. A sequence at the N-terminal of F11R together with another sequence located in the first Ig-loop of this protein, were identified to form a steric active-site operating in the F11R-dependent adhesion between cells that express F11R molecules on their external surface. In silico modeling of the complex between two polypeptide chains with the sequences positioned in the active-site was used to generate peptide-candidates designed to inhibit homophilic interactions between surface-located F11R molecules. The two lead F11R peptides were modified with D-Arg and D-Lys at selective sites, for attaining higher stability to proteolysis in vivo. Using molecular docking experiments we tested different conformational states and the putative binding affinity between two selected D-Arg and D-Lys-modified F11R peptides and the proposed binding pocket. The inhibitory effects of the F11R peptide 2HN-(dK)-SVT-(dR)-EDTGTYTC-CONH2 on antibody-induced platelet aggregation and on the adhesion of platelets to cytokine-inflammed endothelial cells are reported in detail, and the results point out the significant potential utilization of F11R peptides for the prevention and treatment of atherosclerotic plaques and associated thrombotic events.
Collapse
Affiliation(s)
- A Babinska
- Division of Nephrology, Department of Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, 11203; Department of Cell Biology and Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, 11203
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Torbeev V, Grogg M, Ruiz J, Boehringer R, Schirer A, Hellwig P, Jeschke G, Hilvert D. Chiral recognition in amyloid fiber growth. J Pept Sci 2016; 22:290-304. [DOI: 10.1002/psc.2861] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/05/2016] [Accepted: 01/11/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Vladimir Torbeev
- Institut de Science et d'Ingénierie Supramoléculaires; International Center for Frontier Research in Chemistry, UMR 7006, Université de Strasbourg; 8 allée Gaspard Monge 67000 Strasbourg France
| | - Marcel Grogg
- Laboratory of Organic Chemistry, ETH Zurich; Vladimir-Prelog-Weg 1-5/10; Zürich CH-8093 Switzerland
| | - Jérémy Ruiz
- Institut de Science et d'Ingénierie Supramoléculaires; International Center for Frontier Research in Chemistry, UMR 7006, Université de Strasbourg; 8 allée Gaspard Monge 67000 Strasbourg France
| | - Régis Boehringer
- Institut de Science et d'Ingénierie Supramoléculaires; International Center for Frontier Research in Chemistry, UMR 7006, Université de Strasbourg; 8 allée Gaspard Monge 67000 Strasbourg France
| | - Alicia Schirer
- Laboratoire de Bioélectrochimie et Spectroscopie; Chimie de la Matière Complexe, UMR 7140, Université de Strasbourg-CNRS; 1 rue Blaise Pascal 67070 Strasbourg France
| | - Petra Hellwig
- Laboratoire de Bioélectrochimie et Spectroscopie; Chimie de la Matière Complexe, UMR 7140, Université de Strasbourg-CNRS; 1 rue Blaise Pascal 67070 Strasbourg France
| | - Gunnar Jeschke
- Laboratory of Physical Chemistry; ETH Zurich, Vladimir-Prelog-Weg 1-5/10; Zürich CH-8093 Switzerland
| | - Donald Hilvert
- Laboratory of Organic Chemistry, ETH Zurich; Vladimir-Prelog-Weg 1-5/10; Zürich CH-8093 Switzerland
| |
Collapse
|
25
|
Rudolph S, Klein AN, Tusche M, Schlosser C, Elfgen A, Brener O, Teunissen C, Gremer L, Funke SA, Kutzsche J, Willbold D. Competitive Mirror Image Phage Display Derived Peptide Modulates Amyloid Beta Aggregation and Toxicity. PLoS One 2016; 11:e0147470. [PMID: 26840229 PMCID: PMC4740492 DOI: 10.1371/journal.pone.0147470] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 01/04/2016] [Indexed: 11/29/2022] Open
Abstract
Alzheimer´s disease is the most prominent type of dementia and currently no causative treatment is available. According to recent studies, oligomeric species of the amyloid beta (Aβ) peptide appear to be the most toxic Aβ assemblies. Aβ monomers, however, may be not toxic per se and may even have a neuroprotective role. Here we describe a competitive mirror image phage display procedure that allowed us to identify preferentially Aβ1–42 monomer binding and thereby stabilizing peptides, which destabilize and thereby eliminate toxic oligomer species. One of the peptides, called Mosd1 (monomer specific d-peptide 1), was characterized in more detail. Mosd1 abolished oligomers from a mixture of Aβ1–42 species, reduced Aβ1–42 toxicity in cell culture, and restored the physiological phenotype in neuronal cells stably transfected with the gene coding for human amyloid precursor protein.
Collapse
Affiliation(s)
- Stephan Rudolph
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Research Centre Jülich, 52425 Jülich, Germany
| | - Antonia Nicole Klein
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Research Centre Jülich, 52425 Jülich, Germany
| | - Markus Tusche
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Research Centre Jülich, 52425 Jülich, Germany
| | - Christine Schlosser
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Research Centre Jülich, 52425 Jülich, Germany
| | - Anne Elfgen
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Research Centre Jülich, 52425 Jülich, Germany
| | - Oleksandr Brener
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Research Centre Jülich, 52425 Jülich, Germany
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Charlotte Teunissen
- Neurochemistry Laboratory and Biobank, VU University Medical Center Amsterdam, The Netherlands
| | - Lothar Gremer
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Research Centre Jülich, 52425 Jülich, Germany
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Susanne Aileen Funke
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Research Centre Jülich, 52425 Jülich, Germany
- Fakultät Angewandte Naturwissenschaften, Hochschule für angewandte Wissenschaften Coburg, 96450 Coburg, Germany
| | - Janine Kutzsche
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Research Centre Jülich, 52425 Jülich, Germany
| | - Dieter Willbold
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Research Centre Jülich, 52425 Jülich, Germany
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
- * E-mail:
| |
Collapse
|
26
|
|
27
|
Pharmacokinetic Properties of a Novel d-Peptide Developed to be Therapeutically Active Against Toxic β-Amyloid Oligomers. Pharm Res 2015; 33:328-36. [DOI: 10.1007/s11095-015-1791-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 09/08/2015] [Indexed: 10/23/2022]
|
28
|
VanCompernolle S, Smith PB, Bowie JH, Tyler MJ, Unutmaz D, Rollins-Smith LA. Inhibition of HIV infection by caerin 1 antimicrobial peptides. Peptides 2015; 71:296-303. [PMID: 26026377 DOI: 10.1016/j.peptides.2015.05.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Revised: 05/14/2015] [Accepted: 05/16/2015] [Indexed: 10/23/2022]
Abstract
The major mode of transmission of the human immunodeficiency virus (HIV) is by sexual intercourse. In the effort to halt the spread of HIV, one measure that holds great promise is the development of effective microbicides that can prevent transmission. Previously we showed that several amphibian antimicrobial peptides (AMPs) completely inhibit HIV infection of T cells while maintaining good viability of the T cell targets. These peptides also inhibited the transfer of HIV by dendritic cells (DCs) to T cells when added up to 8h after virus exposure. Here we report on the anti-HIV activity of 18 additional structurally related caerin 1 family peptides in comparison with our previous best candidate caerin 1.9. Nine peptides were equally effective or more effective in the inhibition of T cell infection and disruption of the HIV envelope as caerin 1.9. Of those nine peptides, three peptides (caerin 1.2, caerin 1.10, and caerin 1.20) exhibited excellent inhibition of HIV infectivity at low concentrations (12-25μM) and limited toxicity against target T cells and endocervical epithelial cells. There was a direct correlation between the effectiveness of the peptides in disruption of the viral envelope and their capacity to inhibit infection. Thus, several additional caerin 1 family peptides inhibit HIV infection have limited toxicity for vaginal epithelial cells, and would be good candidates for inclusion in microbicide formulations.
Collapse
Affiliation(s)
- Scott VanCompernolle
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States
| | - Patricia B Smith
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States
| | - John H Bowie
- Department of Chemistry, The University of Adelaide, Australia
| | - Michael J Tyler
- Department of Environmental Biology, The University of Adelaide, Australia
| | - Derya Unutmaz
- Department of Microbiology, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, United States
| | - Louise A Rollins-Smith
- Departments of Pathology, Microbiology and Immunology, and of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232, United States; Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, United States.
| |
Collapse
|
29
|
Retroinverso analogs of spadin display increased antidepressant effects. Psychopharmacology (Berl) 2015; 232:561-74. [PMID: 25080852 PMCID: PMC4302242 DOI: 10.1007/s00213-014-3683-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 07/07/2014] [Indexed: 01/22/2023]
Abstract
RATIONALE Although depression is the most common mood disorder, only one third of patients are treated with success. Finding new targets, new drugs, and also new drug intake way are the main challenges in the depression field. Several years ago, we identified a new target with the TWIK-related potassium channel-1 (TREK-1) potassium channel, and more recently, we have discovered a peptide of 17 amino acids with antidepressant properties. This peptide, that we called spadin, can be considered as a new concept in antidepressant drug design. Spadin derives from a larger peptide resulting to a posttranslational maturation of sortilin; consequently, spadin can be considered as a natural molecule. Moreover, spadin acts more rapidly than classical antidepressants and does not induce side effects. OBJECTIVES In this work, we sought analogs of spadin displaying a better affinity on TREK-1 channels and an increased action duration. METHODS Analogs were characterized by electrophysiology measurements, by behavioral tests, and by their ability to induce neurogenesis. RESULTS We identified two retro-inverso peptides that have kept the antidepressant properties of spadin; particularly, they increased the hippocampal neurogenesis after a 4-day treatment. As spadin, these analogs did not induce side effects on either pain, epilepsy processes, or at the cardiac level. CONCLUSIONS Together, our results indicated that spadin retro-inverso peptides could represent new potent antidepressant drugs. As exemplified by spadin in the field of depression, retro-inverso strategies could represent a useful technique for developing new classes of drugs in a number of pathologies.
Collapse
|
30
|
Fujiwara T, Yasuda H, Nishimura Y, Nambu H, Yakura T. Synthesis of 10b-fluorinated analogues of protubonine A and its 11a-epimer via fluorocyclisation of tryptophan-containing dipeptides. RSC Adv 2015. [DOI: 10.1039/c4ra08741k] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The 10b-fluorinated analogues of protubonine A and its 11a-epimer were synthesisedviafluorocyclisation of tryptophan-containing dipeptides withN-fluoro-2,4,6-trimethylpyridinium triflate.
Collapse
Affiliation(s)
- Tomoya Fujiwara
- Graduate School of Medicine and Pharmaceutical Sciences
- University of Toyama
- Toyama 930-0194
- Japan
| | - Hiroko Yasuda
- Graduate School of Medicine and Pharmaceutical Sciences
- University of Toyama
- Toyama 930-0194
- Japan
| | - Yushi Nishimura
- Graduate School of Medicine and Pharmaceutical Sciences
- University of Toyama
- Toyama 930-0194
- Japan
| | - Hisanori Nambu
- Graduate School of Medicine and Pharmaceutical Sciences
- University of Toyama
- Toyama 930-0194
- Japan
| | - Takayuki Yakura
- Graduate School of Medicine and Pharmaceutical Sciences
- University of Toyama
- Toyama 930-0194
- Japan
| |
Collapse
|
31
|
Baek KH, Park SK, Lee MH, Kim SI, Cho SH, Choi CB. Development of Rapid Diagnostic Kit for Identification of Hanwoo (Korean Native Cattle) Brand Meat by Detecting BIO-TAG. Korean J Food Sci Anim Resour 2014; 34:339-45. [PMID: 26761175 PMCID: PMC4597872 DOI: 10.5851/kosfa.2014.34.3.339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 04/14/2014] [Accepted: 05/12/2014] [Indexed: 11/06/2022] Open
Abstract
This study was performed to develop a rapid immuno-assay kit, by using a specific antigen to detect Hanwoo brand meat. We selected a synthetic antigen specific to our target antibody, named BIO-TAG (Tyr-D-Ala-Phe), by utilizing a computer-based analysis and literature review. BIO-TAG tagged with adjuvant was subcutaneously injected in sheep and Hanwoo. The serum and meat juice of the immunized or non-immunized animal were then analyzed, to measure the titer of antibody by ELISA and Western blot. The amount of antibodies against the BIO-TAG increased (p<0.05) in serum by vaccination. Furthermore, meat juice from the immunized Hanwoo showed greater (p<0.05) antibody titer, compared with those from non-immunized groups. To optimze the dilution factor, we performed dot-ELISA, with various combination levels of BIO-TAG. Results from dot-ELISA showed that 2 mg/mL BIO-TAG was sufficient to distinguish the immunized meat from non-immunized groups. These results support our hypothesis that simple immunization of Hanwoo generates a sufficient amount of antibodies to be detectable in the meat juice by means of the immune-assay. Therefore, specific Hanwoo brand meat can be more precisely identified by our rapid diagnostic kit. This technology can deter possible fraud of counterfeit meat brands in the Korean domestic market with ease and rapidity; and offers a new tool that guarantees consumers high quality Hanwoo brand beef.
Collapse
Affiliation(s)
| | - Sung Kwon Park
- National Institute of Animal Resource, RDA, Suwon 441-706, Korea
| | | | | | - Soo Hyun Cho
- National Institute of Animal Resource, RDA, Suwon 441-706, Korea
| | - Chang Bon Choi
- Corresponding author: Chang Bon Choi, School of Biotechnology, Yeungnam University, Gyeongsan 712-749, Korea. Tel: +82-53-810-3023, Fax: +82-53-810-4769, E-mail:
| |
Collapse
|
32
|
Chen X, Fan Z, Chen Y, Fang X, Sha X. Retro-inverso carbohydrate mimetic peptides with annexin1-binding selectivity, are stable in vivo, and target tumor vasculature. PLoS One 2013; 8:e80390. [PMID: 24312470 PMCID: PMC3846562 DOI: 10.1371/journal.pone.0080390] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 10/02/2013] [Indexed: 11/30/2022] Open
Abstract
Previous research suggests that carbohydrate mimetic peptide IF7 (IFLLWQR) has an excellent targeting property to annexin1 (Anxa1), a specific marker on the tumor endothelium. However, IF7 is susceptible to proteolysis and has a poor stability in vivo. We prepared a D-amino acid, reverse sequence peptide of IF7, designated RIF7, to confer protease resistance while retaining bioactivity. Experimental results indicate that RIF7 had significantly increased stability and an increased receptor binding affinity than IF7, and this new moiety may represent a clinically relevant vehicle for anticancer drugs.
Collapse
Affiliation(s)
- Xinyi Chen
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
| | - Zhuoyang Fan
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
| | - Yanzuo Chen
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
| | - Xiaoling Fang
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
| | - Xianyi Sha
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
- * E-mail:
| |
Collapse
|
33
|
Chen S, Gfeller D, Buth SA, Michielin O, Leiman PG, Heinis C. Improving binding affinity and stability of peptide ligands by substituting glycines with D-amino acids. Chembiochem 2013; 14:1316-22. [PMID: 23828687 DOI: 10.1002/cbic.201300228] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Indexed: 11/07/2022]
Abstract
Improving the binding affinity and/or stability of peptide ligands often requires testing of large numbers of variants to identify beneficial mutations. Herein we propose a type of mutation that promises a high success rate. In a bicyclic peptide inhibitor of the cancer-related protease urokinase-type plasminogen activator (uPA), we observed a glycine residue that has a positive ϕ dihedral angle when bound to the target. We hypothesized that replacing it with a D-amino acid, which favors positive ϕ angles, could enhance the binding affinity and/or proteolytic resistance. Mutation of this specific glycine to D-serine in the bicyclic peptide indeed improved inhibitory activity (1.75-fold) and stability (fourfold). X-ray-structure analysis of the inhibitors in complex with uPA showed that the peptide backbone conformation was conserved. Analysis of known cyclic peptide ligands showed that glycine is one of the most frequent amino acids, and that glycines with positive ϕ angles are found in many protein-bound peptides. These results suggest that the glycine-to-D-amino acid mutagenesis strategy could be broadly applied.
Collapse
Affiliation(s)
- Shiyu Chen
- Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, BCH 5305 (Batochime), Avenue Forel 2, 1015 Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
34
|
Li J, Gao Y, Kuang Y, Shi J, Du X, Zhou J, Wang H, Yang Z, Xu B. Dephosphorylation of D-peptide derivatives to form biofunctional, supramolecular nanofibers/hydrogels and their potential applications for intracellular imaging and intratumoral chemotherapy. J Am Chem Soc 2013; 135:9907-14. [PMID: 23742714 PMCID: PMC3730259 DOI: 10.1021/ja404215g] [Citation(s) in RCA: 203] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
D-Peptides, as the enantiomers of the naturally occurring L-peptides, usually resist endogenous proteases and are presumably insensitive to most enzymes. But, it is unclear whether or how a phosphatase catalyzes the dephosphorylation from D-peptides. In this work, we examine the formation of the nanofibers of D-peptides via enzymatic dephosphorylation. By comparing the enzymatic hydrogelation of L-peptide and D-peptide based hydrogelators, we find that the chirality of the precursors of the hydrogelators affects little on the enzymatic hydrogelation resulted from the removal of the phosphate group from a tyrosine phosphate residue. The attachment of a therapeutic agent (e.g., taxol) or a fluorophore (e.g., 4-nitro-2,1,3-benzoxadiazole) to the D-peptide based hydrogelators affords a new type of biostable or biocompatible hydrogelators, which may find applications in intratumoral chemotherapy or intracellular imaging, respectively. This work, as the first comprehensive and systematic study of the unexpected enzymatic dephosphorylation of D-peptides, illustrates a useful approach to generate supramolecular hydrogels that have both biostability and other desired functions.
Collapse
Affiliation(s)
- Jiayang Li
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Yuan Gao
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Yi Kuang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Junfeng Shi
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Xuewen Du
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Jie Zhou
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Huaimin Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Zhimou Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| |
Collapse
|
35
|
Li C, Zhan C, Zhao L, Chen X, Lu WY, Lu W. Functional consequences of retro-inverso isomerization of a miniature protein inhibitor of the p53-MDM2 interaction. Bioorg Med Chem 2013; 21:4045-50. [PMID: 23660015 DOI: 10.1016/j.bmc.2013.04.039] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 04/08/2013] [Accepted: 04/10/2013] [Indexed: 12/13/2022]
Abstract
Peptide retro-inverso isomerization is thought to be functionally neutral and has been widely used as a tool for designing proteolytically stable d-isomers to recapitulate biological activities of their parent l-peptides. Despite success in a wide range of applications, exceptions amply exist that clearly defy this rule of thumb when parent l-peptides adopt an α-helical conformation in their bound state. The detrimental energetic effect of retro-inverso isomerization of an α-helical l-peptide on its target protein binding has been estimated to be 3.0-3.4kcal/mol. To better understand how the retro-inverso isomer of a structured protein works at the molecular level, we chemically synthesized and functionally characterized the retro-inverso isomer of a rationally designed miniature protein termed stingin of 18 amino acid residues, which adopts an N-terminal loop and a C-terminal α-helix stabilized by two intra-molecular disulfide bridges. Stingin emulated the transactivation peptide of the p53 tumor suppressor protein and bound with high affinity and via its C-terminal α-helix to MDM2 and MDMX-the two negative regulators of p53. We also prepared the retro isomer and d-enantiomer of stingin for comparative functional studies using fluorescence polarization and surface plasmon resonance techniques. We found that retro-inverso isomerization of l-stingin weakened its MDM2 binding by 720 fold (3.9kcal/mol); while enantiomerization of l-stingin drastically reduced its binding to MDM2 by three orders of magnitude, sequence reversal completely abolished it. Our findings demonstrate the limitation of peptide retro-inverso isomerization in molecular mimicry and reinforce the notion that the strategy works poorly with biologically active α-helical peptides due to inherent differences at the secondary and tertiary structural levels between an l-peptide and its retro-inverso isomer despite their similar side chain topologies at the primary structural level.(1.)
Collapse
Affiliation(s)
- Chong Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | | | | | | | | | | |
Collapse
|
36
|
Falconar AKI. Epitope reactions can be gauged by relative antibody discriminating specificity (RADS) values supported by deletion, substitution and cysteine bridge formation analyses: potential uses in pathogenesis studies. BMC Res Notes 2012; 5:208. [PMID: 22546090 PMCID: PMC3392722 DOI: 10.1186/1756-0500-5-208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 04/30/2012] [Indexed: 01/05/2023] Open
Abstract
Background Epitope-mapping of infectious agents is essential for pathogenesis studies. Since polyclonal antibodies (PAbs) and monoclonal antibodies (MAbs) are always polyspecific and can react with multiple epitopes, it is important to distinguish between specific and non-specific reactions. Relative antibody discriminating specificity (RADS) values, obtained from their relative ELISA reactions with L-amino acid peptides prepared in the natural versus reverse orientations (x-fold absorbance natural/absorbance reverse = RADS value) may be valuable for this purpose. PAbs generated against the dengue type-2 virus (DENV-2) nonstructural-1 (NS1) glycoprotein candidate vaccine also reacted with both DENV envelope (E) glycoproteins and blood-clotting proteins. New xKGSx/xSGKx amino acid motifs were identified on DENV-2 glycoproteins, HIV-1 gp41 and factor IXa. Their potential roles in DENV and HIV-1 antibody-enhanced replication (AER) and auto-immunity were assessed. In this study, a) RADS values were determined for MAbs and PAbs, generated in congeneic (H2: class II) mice against DENV NS1 glycoprotein epitopes, to account for their cross-reaction patterns, and b) MAb 1G5.3 reactions with xKGSx/xSGKx motifs present in the DENV-4 NS1, E and HIV-1 glycoproteins and factor IXa were assessed after the introduction of amino acid substitutions, deletions, or intra-/inter-cysteine (C-C) bridges. Results MAbs 1H7.4, 5H4.3, 3D1.4 and 1G5.3 had high (4.23- to 16.83-fold) RADS values against single epitopes on the DENV-2 NS1 glycoprotein, and MAb 3D1.4 defined the DENV complex-conserved LX1 epitope. In contrast, MAbs 1G5.4-A1-C3 and 1C6.3 had low (0.47- to 1.67-fold) RADS values against multiple epitopes. PAb DENV complex-reactions occurred through moderately-high (2.77- and 3.11-fold) RADS values against the LX1 epitope. MAb 1G5.3 reacted with xSGKx motifs present in DENV-4 NS1 and E glycoproteins, HIV-1 gp41 and factor IXa, while natural C-C bridge formations or certain amino acid substitutions increased its binding activity. Conclusions These results: i) were readily obtained using a standard 96-well ELISA format, ii) showed the LX1 epitope to be the immuno-dominant DENV complex determinant in the NS1 glycoprotein, iii) supported an antigenic co-evolution of the DENV NS1 and E glycoproteins, and iv) identified methods that made it possible to determine the role of anti-DENV PAb reactions in viral pathogenesis.
Collapse
Affiliation(s)
- Andrew K I Falconar
- Laboratorio de Investigaciones en Enfermedades Tropicales, Departamento de Medicina, Universidad del Norte, Km5 Antigua via Puerto Colombia, Barranquilla, Colombia.
| |
Collapse
|
37
|
The insect peptide coprisin prevents Clostridium difficile-mediated acute inflammation and mucosal damage through selective antimicrobial activity. Antimicrob Agents Chemother 2011; 55:4850-7. [PMID: 21807975 DOI: 10.1128/aac.00177-11] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Clostridium difficile-associated diarrhea and pseudomembranous colitis are typically treated with vancomycin or metronidazole, but recent increases in relapse incidence and the emergence of drug-resistant strains of C. difficile indicate the need for new antibiotics. We previously isolated coprisin, an antibacterial peptide from Copris tripartitus, a Korean dung beetle, and identified a nine-amino-acid peptide in the α-helical region of it (LLCIALRKK) that had antimicrobial activity (J.-S. Hwang et al., Int. J. Pept., 2009, doi:10.1155/2009/136284). Here, we examined whether treatment with a coprisin analogue (a disulfide dimer of the nine peptides) prevented inflammation and mucosal damage in a mouse model of acute gut inflammation established by administration of antibiotics followed by C. difficile infection. In this model, coprisin treatment significantly ameliorated body weight decreases, improved the survival rate, and decreased mucosal damage and proinflammatory cytokine production. In contrast, the coprisin analogue had no apparent antibiotic activity against commensal bacteria, including Lactobacillus and Bifidobacterium, which are known to inhibit the colonization of C. difficile. The exposure of C. difficile to the coprisin analogue caused a marked increase in nuclear propidium iodide (PI) staining, indicating membrane damage; the staining levels were similar to those seen with bacteria treated with a positive control for membrane disruption (EDTA). In contrast, coprisin analogue treatment did not trigger increases in the nuclear PI staining of Bifidobacterium thermophilum. This observation suggests that the antibiotic activity of the coprisin analogue may occur through specific membrane disruption of C. difficile. Thus, these results indicate that the coprisin analogue may prove useful as a therapeutic agent for C. difficile infection-associated inflammatory diarrhea and pseudomembranous colitis.
Collapse
|
38
|
Lee SL, Yu LX, Cai B, Johnsons GR, Rosenberg AS, Cherney BW, Guo W, Raw AS. Scientific considerations for generic synthetic salmon calcitonin nasal spray products. AAPS J 2011; 13:14-9. [PMID: 21052882 PMCID: PMC3032093 DOI: 10.1208/s12248-010-9242-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 10/19/2010] [Indexed: 11/30/2022] Open
Abstract
Under the Abbreviated New Drug Application pathway, a proposed generic salmon calcitonin nasal spray is required to demonstrate pharmaceutical equivalence and bioequivalence to the brand-name counterpart or the reference listed drug. This review discusses two important aspects of pharmaceutical equivalence for this synthetic peptide nasal spray product. The first aspect is drug substance sameness, in which a proposed generic salmon calcitonin product is required to demonstrate that it contains the same active ingredient as that in the brand-name counterpart. The second aspect is comparability in product- and process-related factors that may influence immunogenicity (i.e., peptide-related impurities, aggregates, formulation, and leachates from the container/closure system). The comparability of these factors helps to ensure the product safety, particularly with respect to immunogenicity. This review also highlights the key features of in vitro and/or in vivo studies for establishing bioequivalence for a solution nasal spray containing a systemically acting salmon calcitonin.
Collapse
Affiliation(s)
- Sau L. Lee
- />Office of Generic Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, 7519 Standish Place, Rockville, Maryland 20855 USA
| | - Lawrence X. Yu
- />Office of Generic Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, 7519 Standish Place, Rockville, Maryland 20855 USA
| | - Bing Cai
- />Office of Generic Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, 7519 Standish Place, Rockville, Maryland 20855 USA
| | - Gibbes R. Johnsons
- />Office of Biotechnology Products, Center for Drug Evaluation and Research, US Food and Drug Administration, 9000 Rockville Pike, Bethesda, Maryland 20892 USA
| | - Amy S. Rosenberg
- />Office of Biotechnology Products, Center for Drug Evaluation and Research, US Food and Drug Administration, 9000 Rockville Pike, Bethesda, Maryland 20892 USA
| | - Barry W. Cherney
- />Office of Biotechnology Products, Center for Drug Evaluation and Research, US Food and Drug Administration, 9000 Rockville Pike, Bethesda, Maryland 20892 USA
| | - Wei Guo
- />Office of Biotechnology Products, Center for Drug Evaluation and Research, US Food and Drug Administration, 9000 Rockville Pike, Bethesda, Maryland 20892 USA
| | - Andre S. Raw
- />Office of Generic Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, 7519 Standish Place, Rockville, Maryland 20855 USA
| |
Collapse
|
39
|
Brun A, Bárcena J, Blanco E, Borrego B, Dory D, Escribano JM, Le Gall-Reculé G, Ortego J, Dixon LK. Current strategies for subunit and genetic viral veterinary vaccine development. Virus Res 2011; 157:1-12. [PMID: 21316403 DOI: 10.1016/j.virusres.2011.02.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Revised: 02/03/2011] [Accepted: 02/04/2011] [Indexed: 12/24/2022]
Abstract
Developing vaccines for livestock provides researchers with the opportunity to perform efficacy testing in the natural hosts. This enables the evaluation of different strategies, including definition of effective antigens or antigen combinations, and improvement in delivery systems for target antigens so that protective immune responses can be modulated or potentiated. An impressive amount of knowledge has been generated in recent years on vaccine strategies and consequently a wide variety of antigen delivery systems is now available for vaccine research. This paper reviews several antigen production and delivery strategies other than those based on the use of live viral vectors. Genetic and protein subunit vaccines as well as alternative production systems are considered in this review.
Collapse
Affiliation(s)
- Alejandro Brun
- Centro de Investigación en Sanidad Animal (CISA-INIA), Valdeolmos, 28130 Madrid, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Funke SA, van Groen T, Kadish I, Bartnik D, Nagel-Steger L, Brener O, Sehl T, Batra-Safferling R, Moriscot C, Schoehn G, Horn AHC, Müller-Schiffmann A, Korth C, Sticht H, Willbold D. Oral treatment with the d-enantiomeric peptide D3 improves the pathology and behavior of Alzheimer's Disease transgenic mice. ACS Chem Neurosci 2010; 1:639-48. [PMID: 22778851 DOI: 10.1021/cn100057j] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Accepted: 07/21/2010] [Indexed: 11/28/2022] Open
Abstract
Several lines of evidence suggest that the amyloid-β-peptide (Aβ) plays a central role in the pathogenesis of Alzheimer's disease (AD). Not only Aβ fibrils but also small soluble Aβ oligomers in particular are suspected to be the major toxic species responsible for disease development and progression. The present study reports on in vitro and in vivo properties of the Aβ targeting d-enantiomeric amino acid peptide D3. We show that next to plaque load and inflammation reduction, oral application of the peptide improved the cognitive performance of AD transgenic mice. In addition, we provide in vitro data elucidating the potential mechanism underlying the observed in vivo activity of D3. These data suggest that D3 precipitates toxic Aβ species and converts them into nonamyloidogenic, nonfibrillar, and nontoxic aggregates without increasing the concentration of monomeric Aβ. Thus, D3 exerts an interesting and novel mechanism of action that abolishes toxic Aβ oligomers and thereby supports their decisive role in AD development and progression.
Collapse
Affiliation(s)
| | - Thomas van Groen
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Inga Kadish
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Dirk Bartnik
- Heinrich-Heine-Universität Düsseldorf, Institut für Physikalische Biologie and BMFZ, 40225 Düsseldorf, Germany
| | - Luitgard Nagel-Steger
- Heinrich-Heine-Universität Düsseldorf, Institut für Physikalische Biologie and BMFZ, 40225 Düsseldorf, Germany
| | - Oleksandr Brener
- Heinrich-Heine-Universität Düsseldorf, Institut für Physikalische Biologie and BMFZ, 40225 Düsseldorf, Germany
| | - Torsten Sehl
- Heinrich-Heine-Universität Düsseldorf, Institut für Physikalische Biologie and BMFZ, 40225 Düsseldorf, Germany
| | | | - Christine Moriscot
- CEA
- CNRS
- Université Joseph Fourier
- Unit for Virus Host Cell Interactions, 6, rue Jules Horowitz BP 181, F38042 Grenoble, France
| | - Guy Schoehn
- CEA
- CNRS
- Université Joseph Fourier
- Unit for Virus Host Cell Interactions, 6, rue Jules Horowitz BP 181, F38042 Grenoble, France
| | - Anselm H. C. Horn
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Institut für Biochemie, 91054 Erlangen, Germany
| | | | - Carsten Korth
- Heinrich-Heine-Universität Düsseldorf, Institut für Neuropathologie, 40225 Düsseldorf, Germany
| | - Heinrich Sticht
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Institut für Biochemie, 91054 Erlangen, Germany
| | - Dieter Willbold
- Forschungszentrum Jülich, ISB-3, 52425 Jülich, Germany
- Heinrich-Heine-Universität Düsseldorf, Institut für Physikalische Biologie and BMFZ, 40225 Düsseldorf, Germany
- CEA
| |
Collapse
|
41
|
Li C, Pazgier M, Li J, Li C, Liu M, Zou G, Li Z, Chen J, Tarasov SG, Lu WY, Lu W. Limitations of peptide retro-inverso isomerization in molecular mimicry. J Biol Chem 2010; 285:19572-81. [PMID: 20382735 PMCID: PMC2885236 DOI: 10.1074/jbc.m110.116814] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Revised: 04/09/2010] [Indexed: 12/13/2022] Open
Abstract
A retro-inverso peptide is made up of d-amino acids in a reversed sequence and, when extended, assumes a side chain topology similar to that of its parent molecule but with inverted amide peptide bonds. Despite their limited success as antigenic mimicry, retro-inverso isomers generally fail to emulate the protein-binding activities of their parent peptides of an alpha-helical nature. In studying the interaction between the tumor suppressor protein p53 and its negative regulator MDM2, Sakurai et al. (Sakurai, K., Chung, H. S., and Kahne, D. (2004) J. Am. Chem. Soc. 126, 16288-16289) made a surprising finding that the retro-inverso isomer of p53(15-29) retained the same binding activity as the wild type peptide as determined by inhibition enzyme-linked immunosorbent assay. The authors attributed the unusual outcome to the ability of the D-peptide to adopt a right-handed helical conformation upon MDM2 binding. Using a battery of biochemical and biophysical tools, we found that retro-inverso isomerization diminished p53 (15-29) binding to MDM2 or MDMX by 3.2-3.3 kcal/mol. Similar results were replicated with the C-terminal domain of HIV-1 capsid protein (3.0 kcal/mol) and the Src homology 3 domain of Abl tyrosine kinase (3.4 kcal/mol). CD and NMR spectroscopic as well as x-ray crystallographic studies showed that D-peptide ligands of MDM2 invariably adopted left-handed helical conformations in both free and bound states. Our findings reinforce that the retro-inverso strategy works poorly in molecular mimicry of biologically active helical peptides, due to inherent differences at the secondary and tertiary structure levels between an l-peptide and its retro-inverso isomer despite their similar side chain topologies at the primary structure level.
Collapse
Affiliation(s)
- Chong Li
- From the School of Pharmacy, Fudan University, Shanghai 201203, China
- the Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Marzena Pazgier
- the Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Jing Li
- the Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Changqing Li
- the Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Min Liu
- the Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Guozhang Zou
- the Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Zhenyu Li
- the Molecular Oncology Program, H. Lee Moffitt Cancer Center, Tampa, Florida 33612, and
| | - Jiandong Chen
- the Molecular Oncology Program, H. Lee Moffitt Cancer Center, Tampa, Florida 33612, and
| | - Sergey G. Tarasov
- the Structural Biophysics Laboratory, NCI-Frederick, National Institutes of Health, Frederick, Maryland 21702
| | - Wei-Yue Lu
- From the School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Wuyuan Lu
- the Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| |
Collapse
|
42
|
Bartnik D, Funke SA, Andrei-Selmer LC, Bacher M, Dodel R, Willbold D. Differently Selected d-Enantiomeric Peptides Act on Different Aβ Species. Rejuvenation Res 2010; 13:202-5. [DOI: 10.1089/rej.2009.0924] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Dirk Bartnik
- Institut für Physikalische Biologie, Heinrich-Heine-Universität, 40225 Düsseldorf, Germany
- These authors contributed equally to the paper
| | - Susanne Aileen Funke
- Forschungszentrum Jülich, ISB-3, 52425 Jülich, Germany
- These authors contributed equally to the paper
| | - Luminita-Cornelia Andrei-Selmer
- Biomedizinisches Forschungszentrum, Department of Neurology, Philipps University Marburg, 35037 Marburg, Germany
- These authors contributed equally to the paper
| | - Michael Bacher
- Biomedizinisches Forschungszentrum, Department of Neurology, Philipps University Marburg, 35037 Marburg, Germany
| | - Richard Dodel
- Biomedizinisches Forschungszentrum, Department of Neurology, Philipps University Marburg, 35037 Marburg, Germany
| | - Dieter Willbold
- Institut für Physikalische Biologie, Heinrich-Heine-Universität, 40225 Düsseldorf, Germany
- Forschungszentrum Jülich, ISB-3, 52425 Jülich, Germany
| |
Collapse
|
43
|
Moiani D, Salvalaglio M, Cavallotti C, Bujacz A, Redzynia I, Bujacz G, Dinon F, Pengo P, Fassina G. Structural Characterization of a Protein A Mimetic Peptide Dendrimer Bound to Human IgG. J Phys Chem B 2009; 113:16268-75. [DOI: 10.1021/jp909405b] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- D. Moiani
- Department of Chimica, Materiali e Ingegneria Chimica, Politecnico di Milano, 20131 Milano, Italy, Institute of Technical Biochemistry, Technical University of Lodz, Stefanowskiego 4/10, Lodz, Poland, and Xeptagen S.p.A., VEGA Science Park-Building Auriga Via delle Industrie 9, I-30175 Marghera (VE), Italy
| | - M. Salvalaglio
- Department of Chimica, Materiali e Ingegneria Chimica, Politecnico di Milano, 20131 Milano, Italy, Institute of Technical Biochemistry, Technical University of Lodz, Stefanowskiego 4/10, Lodz, Poland, and Xeptagen S.p.A., VEGA Science Park-Building Auriga Via delle Industrie 9, I-30175 Marghera (VE), Italy
| | - C. Cavallotti
- Department of Chimica, Materiali e Ingegneria Chimica, Politecnico di Milano, 20131 Milano, Italy, Institute of Technical Biochemistry, Technical University of Lodz, Stefanowskiego 4/10, Lodz, Poland, and Xeptagen S.p.A., VEGA Science Park-Building Auriga Via delle Industrie 9, I-30175 Marghera (VE), Italy
| | - A. Bujacz
- Department of Chimica, Materiali e Ingegneria Chimica, Politecnico di Milano, 20131 Milano, Italy, Institute of Technical Biochemistry, Technical University of Lodz, Stefanowskiego 4/10, Lodz, Poland, and Xeptagen S.p.A., VEGA Science Park-Building Auriga Via delle Industrie 9, I-30175 Marghera (VE), Italy
| | - I. Redzynia
- Department of Chimica, Materiali e Ingegneria Chimica, Politecnico di Milano, 20131 Milano, Italy, Institute of Technical Biochemistry, Technical University of Lodz, Stefanowskiego 4/10, Lodz, Poland, and Xeptagen S.p.A., VEGA Science Park-Building Auriga Via delle Industrie 9, I-30175 Marghera (VE), Italy
| | - G. Bujacz
- Department of Chimica, Materiali e Ingegneria Chimica, Politecnico di Milano, 20131 Milano, Italy, Institute of Technical Biochemistry, Technical University of Lodz, Stefanowskiego 4/10, Lodz, Poland, and Xeptagen S.p.A., VEGA Science Park-Building Auriga Via delle Industrie 9, I-30175 Marghera (VE), Italy
| | - F. Dinon
- Department of Chimica, Materiali e Ingegneria Chimica, Politecnico di Milano, 20131 Milano, Italy, Institute of Technical Biochemistry, Technical University of Lodz, Stefanowskiego 4/10, Lodz, Poland, and Xeptagen S.p.A., VEGA Science Park-Building Auriga Via delle Industrie 9, I-30175 Marghera (VE), Italy
| | - P. Pengo
- Department of Chimica, Materiali e Ingegneria Chimica, Politecnico di Milano, 20131 Milano, Italy, Institute of Technical Biochemistry, Technical University of Lodz, Stefanowskiego 4/10, Lodz, Poland, and Xeptagen S.p.A., VEGA Science Park-Building Auriga Via delle Industrie 9, I-30175 Marghera (VE), Italy
| | - G. Fassina
- Department of Chimica, Materiali e Ingegneria Chimica, Politecnico di Milano, 20131 Milano, Italy, Institute of Technical Biochemistry, Technical University of Lodz, Stefanowskiego 4/10, Lodz, Poland, and Xeptagen S.p.A., VEGA Science Park-Building Auriga Via delle Industrie 9, I-30175 Marghera (VE), Italy
| |
Collapse
|
44
|
Funke SA, Willbold D. Mirror image phage display--a method to generate D-peptide ligands for use in diagnostic or therapeutical applications. MOLECULAR BIOSYSTEMS 2009; 5:783-6. [PMID: 19603110 DOI: 10.1039/b904138a] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mirror image phage display is a straightforward approach to identify new potentially therapeutically active D-enantiomeric peptides. Such D-peptides are more resistant to proteolytic degradation compared to L-peptides. In this review, several examples of mirror image phage display derived D-peptides with therapeutical potential are introduced and discussed.
Collapse
|
45
|
van Groen T, Kadish I, Wiesehan K, Funke S, Willbold D. In vitro and in vivo Staining Characteristics of Small, Fluorescent, Aβ42-Binding D-Enantiomeric Peptides in Transgenic AD Mouse Models. ChemMedChem 2009; 4:276-82. [DOI: 10.1002/cmdc.200800289] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
46
|
van Groen T, Wiesehan K, Funke SA, Kadish I, Nagel-Steger L, Willbold D. Reduction of Alzheimer's disease amyloid plaque load in transgenic mice by D3, A D-enantiomeric peptide identified by mirror image phage display. ChemMedChem 2009; 3:1848-52. [PMID: 19016284 DOI: 10.1002/cmdc.200800273] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Thomas van Groen
- Dept. Cell Biology, University of Alabama at Birmingham, AL 35294, USA
| | | | | | | | | | | |
Collapse
|
47
|
Müller-Schiffmann A, Petsch B, Leliveld SR, Muyrers J, Salwierz A, Mangels C, Schwarzinger S, Riesner D, Stitz L, Korth C. Complementarity determining regions of an anti-prion protein scFv fragment orchestrate conformation specificity and antiprion activity. Mol Immunol 2008; 46:532-40. [PMID: 18973947 DOI: 10.1016/j.molimm.2008.07.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2008] [Revised: 07/16/2008] [Accepted: 07/17/2008] [Indexed: 11/16/2022]
Abstract
The prion protein, PrP, exists in several stable conformations, with the presence of one conformation, PrP(Sc), associated with transmissible neurodegenerative diseases. Targeting PrP by high-affinity ligands has been proven to be an effective way of preventing peripheral prion infections. Here, we have generated bacterially expressed single chain fragments of the variable domains (scFv) of a monoclonal antibody in Escherichia coli, originally raised against purified PrP(Sc) that recognizes both PrP(C) and PrP(Sc). This scFv fragment had a dissociation constant (K(D)) with recombinant PrP of 2 nM and cleared prions in ScN2a cells at 4 nM, as demonstrated by a mouse prion bioassay. A peptide corresponding to the complementarity determining region 3 of the heavy chain (CDR3H) selectively bound PrP(Sc) but had lost antiprion activity. However, synthesis and application of an improved peptide mimicking side chain topology of CDR3H while exhibiting increased protease resistance, a retro-inverso d-peptide of CDR3H, still bound PrP(Sc) and reinstated antiprion activity. We conclude that (1) scFvW226 is so far the smallest polypeptide with bioassay confirmed antiprion activity, and (2) differential conformation specificity and bioactivity can be regulated by orchestrating the participation of different CDRs.
Collapse
Affiliation(s)
- Andreas Müller-Schiffmann
- Department Neuropathology, Heinrich Heine University Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Peacock AFA, Hemmingsen L, Pecoraro VL. Using diastereopeptides to control metal ion coordination in proteins. Proc Natl Acad Sci U S A 2008; 105:16566-71. [PMID: 18940928 PMCID: PMC2575460 DOI: 10.1073/pnas.0806792105] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2008] [Indexed: 11/18/2022] Open
Abstract
Here, we report a previously undescribed approach for controlling metal ion coordination geometry in biomolecules by reorientating amino acid side chains through substitution of L- to D-amino acids. These diastereopeptides allow us to manipulate the spatial orientation of amino acid side chains to alter the sterics of metal binding pockets. We have used this approach to design the de novo metallopeptide, Cd(TRIL12L(D)L16C)(3)(-), which is an example of Cd(II) bound to 3 L-Cys as exclusively trigonal CdS(3), as characterized by a combination of (113)Cd NMR and (111m)Cd PAC spectroscopy. We subsequently show that the physical properties of such a site, such as the high pK(a2) for Cd(II) binding of 15.1, is due to the nature of the coordination number and not the ligating group. Further more this approach allowed for the design of a construct, GRANDL12L(D)L16CL26AL30C, capable of independently binding 2 equivalents of Cd(II) to 2 very similar Cys sites as exclusively 3- and 4-, CdS(3) and CdS(3)O, respectively. Demonstrating that we are capable of controlling the Cd(II) coordination number in these 2 sites solely by varying the nature of a noncoordinating second coordination sphere amino acid, with D-leucine and L-alanine resulting in exclusively 3- and 4-coordinate structures, respectively. Cd(II) was found to selectively bind to the 4-coordinate CdS(3)O site, demonstrating that a protein can be designed that displays metal-binding selectivity based solely on coordination number control and not on the chemical identity of coordinating ligands.
Collapse
Affiliation(s)
- Anna F. A. Peacock
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109-1055; and
| | - Lars Hemmingsen
- Department of Natural Sciences, Faculty of Life Sciences, University of Copenhagen, Thorvaldsensvej 40, 1871 Frederiksberg C, Denmark
| | - Vincent L. Pecoraro
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109-1055; and
| |
Collapse
|
49
|
Abstract
The S peptide from ribonuclease S was used as a model system to explore the relationship between the native peptide and its retroinverso (RI) analog. As probed by circular dichroism, the conformations of S peptide and retroinverso S peptide (RIS peptide) are each right-handed helical conformation. The helical propensity of retro S peptide is greater than S peptide, in trifluoroethanol (TFE). In 70% TFE, the S peptide possesses greater helicity at pH 4 than at pH 7, whereas RIS peptide possesses greater helicity at pH 7 than at pH 4. The RIS peptide does not mimic the S peptide in binding to S protein. Specifically, the RIS peptide does not mimic the S peptide to effect RNase activity with S protein and it also does not inhibit the RNase activity of S peptide with S protein. The biological mimicry between the S peptide and its RIS analog depends on the conformation and relatedness of both the side chain and backbone substructures. The backbones in the S peptide and its RIS analog are reverted with respect to each other; however, the side chain patterns are predicted to be similar. Importantly, if the molecular interactions of backbone atoms of the S peptide and its binding to S protein, then the RIS analog would be unlikely to mimic this parent peptide.
Collapse
|
50
|
Mileusnic R, Lancashire C, Clark J, Rose SPR. Protection against Aβ-induced memory loss by tripeptide D-Arg-L-Glu-L-Arg. Behav Pharmacol 2007; 18:231-8. [PMID: 17426487 DOI: 10.1097/fbp.0b013e32814fcde9] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The molecular and cellular mechanisms underlying the memory deficits in Alzheimer's disease are increasingly thought to be associated with faulty processing of amyloid precursor protein. Following our earlier findings that it is possible to use the tripeptide RER (NH2-D-Arg-L-Glu-L-Arg-COOH, derived from the external domain of amyloid precursor protein) to rescue memory in animal models, we report here that the diasteromeric (D/L) form of the acetylated tripeptide RER protects against Abeta-induced memory loss for a passive avoidance task in young chicks and enhances retention for a weak version of the task when injected peripherally up to 12 h before training. The tripeptide readily crosses the blood-brain barrier, binds to membrane receptor sites in the brain and is without adverse effects on general behaviour. We discuss this finding in the context of other studies of the importance of peptides containing D-amino acids, and conclude that these RER-related peptides may form the basis for a potential therapeutic agent in the early stages of Alzheimer's disease.
Collapse
Affiliation(s)
- Radmila Mileusnic
- Department of Biological Sciences, the Open University, Milton Keynes, UK.
| | | | | | | |
Collapse
|