1
|
Boll LB, Kreft LW, Wennemers H. Fluorophores Nucleate and Stabilize Collagen Triple Helices - En Route to Better Diagnostic Tools for Fibrosis and Tissue Repair. Angew Chem Int Ed Engl 2025; 64:e202500856. [PMID: 40079675 DOI: 10.1002/anie.202500856] [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: 01/10/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/15/2025]
Abstract
Fluorescently labeled collagen model peptides (CMPs) are useful tools for monitoring fibrotic processes and tissue repair. Fine-tuning the propensity of the CMP to form a collagen triple helix is crucial since only single-stranded but not triple-helical CMPs integrate efficiently into damaged or remodeling tissue. Yet the impact of fluorophores on the folding behavior of CMPs and triple helix stability has so far been underappreciated. Here, we show that N-terminal fluorophores enhance triple helix stability and nucleation. Thermal denaturation and temperature jump experiments showed that triple helix stability and nucleation are proportionally affected by the accessible surface area of the fluorophore. For biological applications, fluorophores with a small surface area are therefore beneficial. Furthermore, we show that fluorophores bearing ionizable groups with an appropriate pKa value, such as commonly used fluorescein, can be used to create pH-responsive assemblies. Our findings highlight the importance of carefully selecting appropriate fluorophores and spacers when developing fluorescent probes for biological applications.
Collapse
Affiliation(s)
- Linus B Boll
- Laboratory of Organic Chemistry, ETH Zürich, Vladimir-Prelog-Weg 3, 8093, Zürich, Switzerland
| | - Leonard W Kreft
- Laboratory of Organic Chemistry, ETH Zürich, Vladimir-Prelog-Weg 3, 8093, Zürich, Switzerland
| | - Helma Wennemers
- Laboratory of Organic Chemistry, ETH Zürich, Vladimir-Prelog-Weg 3, 8093, Zürich, Switzerland
| |
Collapse
|
2
|
Kishko M, Stuebler A, Sasmal S, Chan Y, Huang D, Reyes C, Lin J, Price O, Kume A, Zong K, Bricault C, Alamares-Sapuay J, Zhang L. A Computationally Designed Prefusion Stabilized Human Metapneumovirus Fusion Protein Vaccine Antigen Elicited a Potent Neutralization Response. Vaccines (Basel) 2025; 13:523. [PMID: 40432132 PMCID: PMC12115362 DOI: 10.3390/vaccines13050523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 05/04/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Human metapneumovirus (hMPV) is a leading cause of respiratory infections in the elderly, with high morbidity and mortality and with no vaccines or specific therapies available. The primary protective antigen of hMPV is the fusion protein, and its prefusion conformation (pre-F) is considered the most promising target for vaccine development. Methods: Utilizing computational design strategies focused on intraprotomer interface stabilization, we designed hMPV pre-F recombinant subunit vaccine candidates based on the most prevalent A2 subtype and characterized them in vitro and in vivo, benchmarking to the prototypical hMPV pre-F stabilized by an introduction of a proline at site 185. Results: The top candidate (N46V_T160F) yielded 14.4 mg/L with a melting temperature of 79.3 °C as compared to 5.7 mg/L and 70.4 °C for the benchmark. By employing monoclonal antibody binding to all six antigenic sites of hMPV pre-F, we confirmed this construct retained all pre-F specific antigenic sites and that the key sites Ø and V were stable at 4 °C for up to 6 months. When immunogenicity of N46V_T160F was evaluated in mice, it induced higher binding and neutralizing antibody titers than the benchmark, which stemmed in part from increased levels of site Ø and site II targeting Abs. Further, this A2 based construct induced cross-neutralizing Abs against all four hMPV subtypes. Lastly, our construct exhibited similar immunogenicity as the recently published next-generation hMPV pre-F constructs, DS-CavEs2 and v3B_Δ12_D454C-V458C. Conclusions: N46V_T160F is a promising hMPV vaccine candidate paving the way for further development and optimization.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Linong Zhang
- Sanofi, 200 West Street, Waltham, MA 02451, USA; (M.K.); (A.S.); (S.S.); (Y.C.); (D.H.); (C.R.); (J.L.); (O.P.); (A.K.); (K.Z.); (C.B.); (J.A.-S.)
| |
Collapse
|
3
|
Fiala T, Bittner P, Heeb R, Islami V, Söll C, Pruška A, Zenobi R, Wennemers H. Hyperstable, Minimal-Length, and Blunt-Ended Collagen Heterotrimers. Angew Chem Int Ed Engl 2025:e202503353. [PMID: 40344344 DOI: 10.1002/anie.202503353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/11/2025]
Abstract
Most natural collagens are heterotrimers-triple helices formed from three non-identical peptide strands. The design of synthetic heterotrimeric collagen is challenging since a mixture of three different peptides can form as many as 27 unique triple helices. Here, we present a general method for the assembly of collagen heterotrimers with a wide range of lengths, thermal stabilities, and strand arrangements driven by complementary interstrand salt bridges between (2S,4S)-4-aminoproline and aspartate residues. We show how kinetic trapping of undesired trimers can be overcome by adjusting the annealing conditions to obtain the target heterotrimeric helix selectively under thermodynamic control. The design rules and annealing methods allowed the creation of the most stable supramolecular heterotrimer (32 residues, Tm = 76 °C) and the shortest stable heterotrimer (17 residues, Tm = 19 °C) to date. Furthermore, frame-shifting enabled, for the first time, the creation of a collagen triple helix with blunt ends.
Collapse
Affiliation(s)
- Tomas Fiala
- Laboratory of Organic Chemistry, ETH Zürich, Vladimir-Prelog-Weg 3, Zürich, 8093, Switzerland
- Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Philipp Bittner
- Laboratory of Organic Chemistry, ETH Zürich, Vladimir-Prelog-Weg 3, Zürich, 8093, Switzerland
| | - Rahel Heeb
- Laboratory of Organic Chemistry, ETH Zürich, Vladimir-Prelog-Weg 3, Zürich, 8093, Switzerland
| | - Valdrin Islami
- Laboratory of Organic Chemistry, ETH Zürich, Vladimir-Prelog-Weg 3, Zürich, 8093, Switzerland
| | - Carolina Söll
- Laboratory of Organic Chemistry, ETH Zürich, Vladimir-Prelog-Weg 3, Zürich, 8093, Switzerland
| | - Adam Pruška
- Laboratory of Organic Chemistry, ETH Zürich, Vladimir-Prelog-Weg 3, Zürich, 8093, Switzerland
| | - Renato Zenobi
- Laboratory of Organic Chemistry, ETH Zürich, Vladimir-Prelog-Weg 3, Zürich, 8093, Switzerland
| | - Helma Wennemers
- Laboratory of Organic Chemistry, ETH Zürich, Vladimir-Prelog-Weg 3, Zürich, 8093, Switzerland
| |
Collapse
|
4
|
Morano NC, Guo Y, Becker JE, Li Z, Yu J, Ho DD, Shapiro L, Kwong PD. Structure of a zoonotic H5N1 hemagglutinin reveals a receptor-binding site occupied by an auto-glycan. Structure 2025; 33:228-233.e3. [PMID: 39884273 DOI: 10.1016/j.str.2025.01.001] [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: 12/04/2024] [Revised: 12/23/2024] [Accepted: 01/03/2025] [Indexed: 02/01/2025]
Abstract
Highly pathogenic avian influenza has spilled into many mammals, most notably cows and poultry, with several dozen human breakthrough infections. Zoonotic crossovers, with hemagglutinins mutated to enhance viral ability to use human α2-6-linked sialic acid receptors versus avian α2-3-linked ones, highlight the pandemic risk. To gain insight into these crossovers, we determined the cryoelectron microscopy (cryo-EM) structure of the hemagglutinin from the zoonotic H5N1 A/Texas/37/2024 strain (clade 2.3.4.4b) in complex with a previously reported neutralizing antibody. Surprisingly, we found that the receptor-binding site of this H5N1 hemagglutinin was already occupied by an α2-3-linked sialic acid and that this glycan emanated from asparagine N169 of a neighboring protomer on hemagglutinin itself. This structure thus highlights recognition by influenza hemagglutinin of an "auto"-α2-3-linked sialic acid from N169, an N-linked glycan conserved in 95% of H5 strains, and adds "auto-glycan recognition," which may play a role in viral dispersal, to the complexities surrounding H5N1 zoonosis.
Collapse
MESH Headings
- Influenza A Virus, H5N1 Subtype/chemistry
- Influenza A Virus, H5N1 Subtype/metabolism
- Hemagglutinin Glycoproteins, Influenza Virus/chemistry
- Hemagglutinin Glycoproteins, Influenza Virus/metabolism
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Binding Sites
- Humans
- Animals
- Polysaccharides/chemistry
- Polysaccharides/metabolism
- Cryoelectron Microscopy
- Protein Binding
- Models, Molecular
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/metabolism
- N-Acetylneuraminic Acid/metabolism
- N-Acetylneuraminic Acid/chemistry
- Receptors, Virus/metabolism
- Receptors, Virus/chemistry
- Influenza, Human/virology
Collapse
Affiliation(s)
- Nicholas C Morano
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10027, USA
| | - Yicheng Guo
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA.
| | - Jordan E Becker
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10027, USA
| | - Zhiteng Li
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Jian Yu
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - David D Ho
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Lawrence Shapiro
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10027, USA.
| | - Peter D Kwong
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
5
|
Yang Q, Xue B, Liu F, Lu Y, Tang J, Yan M, Wu Q, Chen R, Zhou A, Liu L, Liu J, Qu C, Wu Q, Fu M, Zhong J, Dong J, Chen S, Wang F, Zhou Y, Zheng J, Peng W, Shang J, Chen X. Farnesyltransferase inhibitor lonafarnib suppresses respiratory syncytial virus infection by blocking conformational change of fusion glycoprotein. Signal Transduct Target Ther 2024; 9:144. [PMID: 38853183 PMCID: PMC11163014 DOI: 10.1038/s41392-024-01858-5] [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: 12/07/2023] [Revised: 04/28/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024] Open
Abstract
Respiratory syncytial virus (RSV) is the major cause of bronchiolitis and pneumonia in young children and the elderly. There are currently no approved RSV-specific therapeutic small molecules available. Using high-throughput antiviral screening, we identified an oral drug, the prenylation inhibitor lonafarnib, which showed potent inhibition of the RSV fusion process. Lonafarnib exhibited antiviral activity against both the RSV A and B genotypes and showed low cytotoxicity in HEp-2 and human primary bronchial epithelial cells (HBEC). Time-of-addition and pseudovirus assays demonstrated that lonafarnib inhibits RSV entry, but has farnesyltransferase-independent antiviral efficacy. Cryo-electron microscopy revealed that lonafarnib binds to a triple-symmetric pocket within the central cavity of the RSV F metastable pre-fusion conformation. Mutants at the RSV F sites interacting with lonafarnib showed resistance to lonafarnib but remained fully sensitive to the neutralizing monoclonal antibody palivizumab. Furthermore, lonafarnib dose-dependently reduced the replication of RSV in BALB/c mice. Collectively, lonafarnib could be a potential fusion inhibitor for RSV infection.
Collapse
Affiliation(s)
- Qi Yang
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Bao Xue
- Guangzhou National Laboratory, Guangzhou, 510005, China
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Fengjiang Liu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yongzhi Lu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jielin Tang
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Mengrong Yan
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qiong Wu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ruyi Chen
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Anqi Zhou
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Lijie Liu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Junjun Liu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Changbin Qu
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Qingxin Wu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Muqing Fu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jiayi Zhong
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jianwei Dong
- Guangzhou National Laboratory, Guangzhou, 510005, China
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Sijie Chen
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Fan Wang
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yuan Zhou
- Guangzhou National Laboratory, Guangzhou, 510005, China
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jie Zheng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou lnstitute for Advanced Study, UCAS, Hangzhou, 310024, China
| | - Wei Peng
- Guangzhou National Laboratory, Guangzhou, 510005, China.
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Jinsai Shang
- Guangzhou National Laboratory, Guangzhou, 510005, China.
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Xinwen Chen
- Guangzhou National Laboratory, Guangzhou, 510005, China.
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
6
|
Nomura K, Fiala T, Wennemers H. Carbohydrate Co-Solutes Stabilize Collagen Triple Helices. Chembiochem 2024; 25:e202300860. [PMID: 38233350 DOI: 10.1002/cbic.202300860] [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: 12/21/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 01/19/2024]
Abstract
Carbohydrates are common co-solutes for the stabilization of proteins. The effect of carbohydrate solutions on the stability of collagen, the most abundant protein in mammals, is, however, underexplored. In this work, we studied the thermal stability of collagen triple helices derived from a molecularly defined collagen model peptide (CMP), Ac-(Pro-Hyp-Gly)7 -NH2 , in solutions of six common mono- and disaccharides. We show that the carbohydrates stabilize the collagen triple helix in a concentration-dependent manner, with an increase of the melting temperature of up to 17 °C. In addition, we show that the stabilizing effect is similar for all studied sugars, including trehalose, which is otherwise considered a privileged bioprotectant. The results provided insight into the effects of sugar co-solutes on collagen triple helices and can aid the selection of storage environments for collagen-based materials and probes.
Collapse
Affiliation(s)
- Kota Nomura
- Laboratory of Organic Chemistry, D-CHAB, ETH Zürich, Vladimir-Prelog-Weg 3, 8093, Zürich, Switzerland
| | - Tomas Fiala
- Laboratory of Organic Chemistry, D-CHAB, ETH Zürich, Vladimir-Prelog-Weg 3, 8093, Zürich, Switzerland
| | - Helma Wennemers
- Laboratory of Organic Chemistry, D-CHAB, ETH Zürich, Vladimir-Prelog-Weg 3, 8093, Zürich, Switzerland
| |
Collapse
|
7
|
Antoniazzi G, Schäfer RJB, Biedermann M, Rüttimann E, Wennemers H. Isonitrile-Proline - A Versatile Handle for the Chemoselective Derivatization of Collagen Peptides. Chemistry 2023; 29:e202302389. [PMID: 37498143 DOI: 10.1002/chem.202302389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 07/28/2023]
Abstract
Functional groups that allow for chemoselective and bioorthogonal derivatization are valuable tools for the labelling of peptides and proteins. The isonitrile is such a group but synthetic methods for its incorporation into peptides by solid-phase peptide synthesis are not known. Here, we introduce (4S)- and (4R)-isonitrileproline (Inp) as building blocks for solid-phase peptide synthesis. Conformational studies of (4S)- and (4R)-Inp and thermal stability analysis of Inp-containing collagen triple helices revealed that the isonitrile group exerts a stereoelectronic gauche effect. We showcase the value of Inp for bioorthogonal labelling by derivatization of Inp-containing collagen model peptides (CMPs). Dual labelling with a pair of bioorthogonal reactions of a CMP containing Inp and azidoproline residues further highlights the versatility of the new isonitrile-containing amino acids.
Collapse
Affiliation(s)
- Giuseppe Antoniazzi
- Laboratory of Organic Chemistry, ETH Zürich, Vladimir-Prelog-Weg 3, 8093, Zürich, Switzerland
| | - Rebecca J B Schäfer
- Laboratory of Organic Chemistry, ETH Zürich, Vladimir-Prelog-Weg 3, 8093, Zürich, Switzerland
| | - Maurice Biedermann
- Laboratory of Organic Chemistry, ETH Zürich, Vladimir-Prelog-Weg 3, 8093, Zürich, Switzerland
| | - Eric Rüttimann
- Laboratory of Organic Chemistry, ETH Zürich, Vladimir-Prelog-Weg 3, 8093, Zürich, Switzerland
| | - Helma Wennemers
- Laboratory of Organic Chemistry, ETH Zürich, Vladimir-Prelog-Weg 3, 8093, Zürich, Switzerland
| |
Collapse
|
8
|
Fiala T, Heeb R, Vigliotti L, Wennemers H. The Yin and Yang of How N-Terminal Acyl Caps Affect Collagen Triple Helices. Biomacromolecules 2023; 24:3954-3960. [PMID: 37227696 DOI: 10.1021/acs.biomac.3c00241] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
N-terminal acylation is a common tool for the installation of functional moieties (e.g., sensors or bioactive molecules) on collagen model peptides (CMPs). The N-acyl group and its length are generally assumed to have little or no influence on the properties of the collagen triple helix formed by the CMP. Here, we show that the length of short (C1-C4) acyl capping groups has different effects on the thermal stability of collagen triple helices in POG, OGP, and GPO frames. While the effect of different capping groups on the stability of triple helices in the GPO frame is negligible, longer acyl chains stabilize OGP triple helices but destabilize POG analogues. The observed trends arise from a combination of steric repulsion, the hydrophobic effect, and n → π* interactions. Our study provides a basis for the design of N-terminally functionalized CMPs with predictable effects on triple helix stability.
Collapse
Affiliation(s)
- Tomas Fiala
- Laboratory of Organic Chemistry, ETH Zurich, D-CHAB, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| | - Rahel Heeb
- Laboratory of Organic Chemistry, ETH Zurich, D-CHAB, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| | - Luca Vigliotti
- Laboratory of Organic Chemistry, ETH Zurich, D-CHAB, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| | - Helma Wennemers
- Laboratory of Organic Chemistry, ETH Zurich, D-CHAB, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| |
Collapse
|
9
|
Keever JM, Banzon PD, Hales MK, Sargent AL, Allen WE. Association between N-Terminal Pyrenes Stabilizes the Collagen Triple Helix. J Org Chem 2023; 88:11885-11894. [PMID: 37531574 DOI: 10.1021/acs.joc.3c01175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
Collagen model peptides featuring the fluorophore pyrene at their N-termini have been synthesized, and their thermal denaturation has been examined using circular dichroism (CD) and fluorescence spectroscopies. Flanking the (Pro-Hyp-Gly)7 core of the peptide monomers at positions 1 and/or 23 in the primary sequence, Lys residues were introduced to ensure water solubility. Triple helices derived from such peptides show a broad excimer emission at ∼480 nm, indicative of interaction between the pyrene units. CD experiments show that the fluorophores enhance helix stability primarily through entropic effects. Unfolding temperatures (Tm) increase by up to 7 °C for systems with N-terminal lysine residues and by up to 21 °C for systems in which the first-position Lys is replaced by Ala. Tm values derived from fluorescence measurements (at 50 μM) typically lie within ∼1 °C of those obtained using CD (at 200 μM). Computational modeling in a water continuum using B3LYP-GD3 and M06-2X functionals predicts that face-to-face association of fluorophores can occur while H-bonding within the [(POG)n]3 assembly is retained. Such parallel stacking is consistent with hydrophobically driven stabilization. Labeling collagen peptides with pyrene is a synthetically simple way to promote triple helicity while providing a means to obtain Tm data on relatively dilute samples.
Collapse
Affiliation(s)
- Jared M Keever
- Department of Chemistry, Science and Technology Building, East Carolina University, Greenville, North Carolina 27858-4353, United States
| | - Patrick D Banzon
- Department of Chemistry, Science and Technology Building, East Carolina University, Greenville, North Carolina 27858-4353, United States
| | - Megan K Hales
- Department of Chemistry, Science and Technology Building, East Carolina University, Greenville, North Carolina 27858-4353, United States
| | - Andrew L Sargent
- Department of Chemistry, Science and Technology Building, East Carolina University, Greenville, North Carolina 27858-4353, United States
| | - William E Allen
- Department of Chemistry, Science and Technology Building, East Carolina University, Greenville, North Carolina 27858-4353, United States
| |
Collapse
|
10
|
Zhao Z, Deng J, Fan D. Green biomanufacturing in recombinant collagen biosynthesis: trends and selection in various expression systems. Biomater Sci 2023; 11:5439-5461. [PMID: 37401335 DOI: 10.1039/d3bm00724c] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2023]
Abstract
Collagen, classically derived from animal tissue, is an all-important protein material widely used in biomedical materials, cosmetics, fodder, food, etc. The production of recombinant collagen through different biological expression systems using bioengineering techniques has attracted significant interest in consideration of increasing market demand and the process complexity of extraction. Green biomanufacturing of recombinant collagen has become one of the focus topics. While the bioproduction of recombinant collagens (type I, II, III, etc.) has been commercialized in recent years, the biosynthesis of recombinant collagen is extremely challenging due to protein immunogenicity, yield, degradation, and other issues. The rapid development of synthetic biology allows us to perform a heterologous expression of proteins in diverse expression systems, thus optimizing the production and bioactivities of recombinant collagen. This review describes the research progress in the bioproduction of recombinant collagen over the past two decades, focusing on different expression systems (prokaryotic organisms, yeasts, plants, insects, mammalian and human cells, etc.). We also discuss the challenges and future trends in developing market-competitive recombinant collagens.
Collapse
Affiliation(s)
- Zilong Zhao
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Xi'an 710069, Shaanxi, China.
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, Shaanxi, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, Shaanxi, China
| | - Jianjun Deng
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Xi'an 710069, Shaanxi, China.
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, Shaanxi, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, Shaanxi, China
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Xi'an 710069, Shaanxi, China.
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, Shaanxi, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, Shaanxi, China
| |
Collapse
|
11
|
Boudko SP, Konopka EH, Kim W, Taga Y, Mizuno K, Springer TA, Hudson BG, Moy TI, Lin FY. A recombinant technique for mapping functional sites of heterotrimeric collagen helices: Collagen IV CB3 fragment as a prototype for integrin binding. J Biol Chem 2023; 299:104901. [PMID: 37302550 PMCID: PMC10404678 DOI: 10.1016/j.jbc.2023.104901] [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: 05/08/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/13/2023] Open
Abstract
Collagen superfamily of proteins is a major component of the extracellular matrix. Defects in collagens underlie the cause of nearly 40 human genetic diseases in millions of people worldwide. Pathogenesis typically involves genetic alterations of the triple helix, a hallmark structural feature that bestows exceptional mechanical resistance to tensile forces and a capacity to bind a plethora of macromolecules. Yet, there is a paramount knowledge gap in understanding the functionality of distinct sites along the triple helix. Here, we present a recombinant technique to produce triple helical fragments for functional studies. The experimental strategy utilizes the unique capacity of the NC2 heterotrimerization domain of collagen IX to drive three α-chain selection and registering the triple helix stagger. For proof of principle, we produced and characterized long triple helical fragments of collagen IV that were expressed in a mammalian system. The heterotrimeric fragments encompassed the CB3 trimeric peptide of collagen IV, which harbors the binding motifs for α1β1 and α2β1 integrins. Fragments were characterized and shown to have a stable triple helix, post-translational modifications, and high affinity and specific binding of integrins. The NC2 technique is a universal tool for the high-yield production of heterotrimeric fragments of collagens. Fragments are suitable for mapping functional sites, determining coding sequences of binding sites, elucidating pathogenicity and pathogenic mechanisms of genetic mutations, and production of fragments for protein replacement therapy.
Collapse
Affiliation(s)
- Sergei P Boudko
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA.
| | | | - Woojin Kim
- Morphic Therapeutic, Inc, Waltham, Massachusetts, USA
| | - Yuki Taga
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, Japan
| | - Kazunori Mizuno
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, Japan
| | - Timothy A Springer
- Department of Biological Chemistry and Molecular Pharmacology, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Billy G Hudson
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Terence I Moy
- Morphic Therapeutic, Inc, Waltham, Massachusetts, USA
| | - Fu-Yang Lin
- Morphic Therapeutic, Inc, Waltham, Massachusetts, USA.
| |
Collapse
|
12
|
Li Y, Yang M, Nan Y, Wang J, Wang S, Cui D, Guo J, He P, Dai W, Zhou S, Zhang Y, Ma W. SARS-CoV-2 spike host cell surface exposure promoted by a COPI sorting inhibitor. Acta Pharm Sin B 2023:S2211-3835(23)00123-5. [PMID: 37360012 PMCID: PMC10110937 DOI: 10.1016/j.apsb.2023.04.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/26/2023] [Accepted: 04/10/2023] [Indexed: 06/28/2023] Open
Abstract
Via an insufficient coat protein complex I (COPI) retrieval signal, the majority of SARS-CoV-2 spike (S) is resident in host early secretory organelles and a tiny amount is leaked out in cell surface. Only surface-exposed S can be recognized by B cell receptor (BCR) or anti-S therapeutic monoclonal antibodies (mAbs) that is the trigger step for B cell activation after S mRNA vaccination or infected cell clearance by S mAbs. Now, a drug strategy to promote S host surface exposure is absent. Here, we first combined structural and biochemical analysis to characterize S COPI sorting signals. A potent S COPI sorting inhibitor was then invented, evidently capable of promoting S surface exposure and facilitating infected cell clearance by S antibody-dependent cellular cytotoxicity (ADCC). Importantly, with the inhibitor as a probe, we revealed Omicron BA.1 S is less cell surface exposed than prototypes because of a constellation of S folding mutations, possibly corresponding to its ER chaperone association. Our findings not only suggest COPI is a druggable target against COVID-19, but also highlight SARS-CoV-2 evolution mechanism driven by S folding and trafficking mutations.
Collapse
Affiliation(s)
- Yiqun Li
- School of Life Science, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Mingrui Yang
- School of Life Science, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yanan Nan
- School of Life Science, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jiaming Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Sanjiao Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Dongxiao Cui
- School of Life Science, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jiajian Guo
- School of Life Science, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Pengfei He
- School of Life Science, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Wenxin Dai
- School of Life Science, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shuqi Zhou
- School of Life Science, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yue Zhang
- School of Life Science, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Wenfu Ma
- School of Life Science, Beijing University of Chinese Medicine, Beijing 102488, China
| |
Collapse
|
13
|
Beal J, Clore A, Manthey J. Studying pathogens degrades BLAST-based pathogen identification. Sci Rep 2023; 13:5390. [PMID: 37012314 PMCID: PMC10068195 DOI: 10.1038/s41598-023-32481-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 03/28/2023] [Indexed: 04/05/2023] Open
Abstract
As synthetic biology becomes increasingly capable and accessible, it is likewise increasingly critical to be able to make accurate biosecurity determinations regarding the pathogenicity or toxicity of particular nucleic acid or amino acid sequences. At present, this is typically done using the BLAST algorithm to determine the best match with sequences in the NCBI nucleic acid and protein databases. Neither BLAST nor any of the NCBI databases, however, are actually designed for biosafety determination. Critically, taxonomic errors or ambiguities in the NCBI nucleic acid and protein databases can also cause errors in BLAST-based taxonomic categorization. With heavily studied taxa and frequently used biotechnology tools, even low frequency taxonomic categorization issues can lead to high rates of errors in biosecurity decision-making. Here we focus on the implications for false positives, finding that BLAST against NCBI's protein database will now incorrectly categorize a number of commonly used biotechnology tool sequences as the pathogens or toxins with which they have been used. Paradoxically, this implies that problems are expected to be most acute for the pathogens and toxins of highest interest and for the most widely used biotechnology tools. We thus conclude that biosecurity tools should shift away from BLAST against general purpose databases and towards new methods that are specifically tailored for biosafety purposes.
Collapse
Affiliation(s)
- Jacob Beal
- Raytheon BBN, 10 Moulton Street, Cambridge, MA, 02138, USA.
| | - Adam Clore
- Integrated DNA Technologies, 1710 Commercial Park, Coralville, IA, 52241, USA
| | - Jeff Manthey
- Integrated DNA Technologies, 1710 Commercial Park, Coralville, IA, 52241, USA
| |
Collapse
|
14
|
McGinnes Cullen L, Luo B, Wen Z, Zhang L, Durr E, Morrison TG. The Respiratory Syncytial Virus (RSV) G Protein Enhances the Immune Responses to the RSV F Protein in an Enveloped Virus-Like Particle Vaccine Candidate. J Virol 2023; 97:e0190022. [PMID: 36602367 PMCID: PMC9888267 DOI: 10.1128/jvi.01900-22] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 01/06/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a serious human respiratory pathogen, but no RSV vaccine has been licensed. Many vaccine candidates are focused on the viral F protein since the F protein is more conserved than the viral G protein across RSV strains and serotypes; thus, the F protein is thought more likely to induce a broader range of protection from infection. However, it is the G protein that binds the likely receptor, CX3CR1, in lung ciliated epithelial cells, raising the question of the importance of the G protein in vaccine candidates. Using virus-like particle (VLP) vaccine candidates, we have directly compared VLPs containing only the prefusion F protein (pre-F), only the G protein, or both glycoproteins. We report that VLPs containing both glycoproteins bind to anti-F-protein-specific monoclonal antibodies differently than do VLPs containing only the prefusion F protein. In RSV-naive cotton rats, VLPs assembled with only the pre-F protein stimulated extremely weak neutralizing antibody (NAb) titers, as did VLPs assembled with G protein. However, VLPs assembled with both glycoproteins stimulated quite robust neutralizing antibody titers, induced improved protection of the animals from RSV challenge compared to pre-F VLPs, and induced significantly higher levels of antibodies specific for F protein antigenic site 0, site III, and the AM14 binding site than did VLPs containing only the pre-F protein. These results indicate that assembly of pre-F protein with G protein in VLPs further stabilized the prefusion conformation or otherwise altered the conformation of the F protein, increasing the induction of protective antibodies. IMPORTANCE Respiratory syncytial virus (RSV) results in significant disease in infants, young children, and the elderly. Thus, development of an effective vaccine for these populations is a priority. Most ongoing efforts in RSV vaccine development have focused on the viral fusion (F) protein; however, the importance of the inclusion of G in vaccine candidates is unclear. Here, using virus-like particles (VLPs) assembled with only the F protein, only the G protein, or both glycoproteins, we show that VLPs assembled with both glycoproteins are a far superior vaccine in a cotton rat model compared with VLPs containing only F protein or only G protein. The results show that the presence of G protein in the VLPs influences the conformation of the F protein and the immune responses to F protein, resulting in significantly higher neutralizing antibody titers and better protection from RSV challenge. These results suggest that inclusion of G protein in a vaccine candidate may improve its effectiveness.
Collapse
Affiliation(s)
- Lori McGinnes Cullen
- Department of Microbiology and Physiological Systems, Program in Immunology and Microbiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Bin Luo
- Pharmacology, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Zhiyun Wen
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Lan Zhang
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Eberhard Durr
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Trudy G. Morrison
- Department of Microbiology and Physiological Systems, Program in Immunology and Microbiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
15
|
Chen Y, Wang F, Yin L, Jiang H, Lu X, Bi Y, Zhang W, Shi Y, Burioni R, Tong Z, Song H, Qi J, Gao GF. Structural basis for a human broadly neutralizing influenza A hemagglutinin stem-specific antibody including H17/18 subtypes. Nat Commun 2022; 13:7603. [PMID: 36494358 PMCID: PMC9734383 DOI: 10.1038/s41467-022-35236-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
Influenza infection continues are a persistent threat to public health. The identification and characterization of human broadly neutralizing antibodies can facilitate the development of antibody drugs and the design of universal influenza vaccines. Here, we present structural information for the human antibody PN-SIA28's heterosubtypic binding of hemagglutinin (HA) from circulating and emerging potential influenza A viruses (IAVs). Aside from group 1 and 2 conventional IAV HAs, PN-SIA28 also inhibits membrane fusion mediated by bat-origin H17 and H18 HAs. Crystallographic analyses of Fab alone or in complex with H1, H14, and H18 HA proteins reveal that PN-SIA28 binds to a highly conserved epitope in the fusion domain of different HAs, with the same CDRHs but different CDRLs for different HAs tested, distinguishing it from other structurally characterized anti-stem antibodies. The binding characteristics of PN-SIA28 provides information to support the design of increasingly potent engineered antibodies, antiviral drugs, and/or universal influenza vaccines.
Collapse
Affiliation(s)
- Yulu Chen
- grid.9227.e0000000119573309CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Fei Wang
- grid.9227.e0000000119573309CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101 China
| | - Liwei Yin
- grid.9227.e0000000119573309CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101 China
| | - Haihai Jiang
- grid.9227.e0000000119573309CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101 China
| | - Xishan Lu
- grid.9227.e0000000119573309CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101 China
| | - Yuhai Bi
- grid.9227.e0000000119573309CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China ,grid.9227.e0000000119573309Center for Influenza Research and Early-warning (CASCIRE), Chinese Academy of Sciences, Beijing, 100101 China
| | - Wei Zhang
- grid.9227.e0000000119573309CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101 China
| | - Yi Shi
- grid.9227.e0000000119573309CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Roberto Burioni
- grid.15496.3f0000 0001 0439 0892Università Vita-Salute San Raffaele, Milano, 20132 Italy
| | - Zhou Tong
- grid.9227.e0000000119573309CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101 China
| | - Hao Song
- grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China ,grid.9227.e0000000119573309Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, 100101 China
| | - Jianxun Qi
- grid.9227.e0000000119573309CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - George F. Gao
- grid.9227.e0000000119573309CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China ,grid.9227.e0000000119573309Center for Influenza Research and Early-warning (CASCIRE), Chinese Academy of Sciences, Beijing, 100101 China ,grid.9227.e0000000119573309Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, 100101 China
| |
Collapse
|
16
|
Potently neutralizing and protective anti-human metapneumovirus antibodies target diverse sites on the fusion glycoprotein. Immunity 2022; 55:1710-1724.e8. [DOI: 10.1016/j.immuni.2022.07.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/16/2022] [Accepted: 07/06/2022] [Indexed: 11/21/2022]
|
17
|
Immunogenicity and protective efficacy of RSV G central conserved domain vaccine with a prefusion nanoparticle. NPJ Vaccines 2022; 7:74. [PMID: 35773301 PMCID: PMC9244890 DOI: 10.1038/s41541-022-00487-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 05/19/2022] [Indexed: 11/10/2022] Open
Abstract
Respiratory syncytial virus (RSV) G glycoprotein has recently reemerged as a vaccine antigen due to its ability to elicit potent neutralizing antibodies and ameliorate disease in animal models. Here we designed three constructs to display the G central conserved domain (Gcc) focused on inducing broad and potent neutralizing antibodies. One construct displaying Gcc from both RSV subgroups trimerized via a C-terminal foldon (Gcc-Foldon) was highly immunogenic in mice and in MIMIC, a pre-immune human in vitro model. To explore an optimal RSV vaccine, we combined the Gcc-Foldon antigen with a stabilized pre-fusion-F nanoparticle (pre-F-NP) as a bivalent vaccine and detected no antigenic interference between the two antigens in the MIMIC model. In RSV-primed macaques, the bivalent vaccine elicited potent humoral responses. Furthermore, both Gcc-Foldon and the bivalent vaccine conferred effective protection against RSV challenge in mice. This two-component vaccine could potentially provide effective protection against RSV infection in humans and warrants further clinical evaluation.
Collapse
|
18
|
Structural basis of protein substrate processing by human mitochondrial high-temperature requirement A2 protease. Proc Natl Acad Sci U S A 2022; 119:e2203172119. [PMID: 35452308 PMCID: PMC9170070 DOI: 10.1073/pnas.2203172119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Protein aggregates are often toxic, leading to impaired cellular activities and disease. The human HtrA2 trimeric enzyme cleaves such aggregates, and mutations in HtrA2 are causative for various neurodegenerative disorders, such as Parkinson’s disease and essential tremor. The mechanism by which cleavage occurs has been studied using small peptides, but little information is available as to how HtrA2 protects cells from the pathologic effects of aggregation involving protein molecules that can form well-folded structures. Using solution NMR spectroscopy, we investigated the structural dynamics of the interaction between HtrA2 and a model protein substrate, demonstrating that HtrA2 preferentially binds to an unfolded substrate ensemble and providing insights into how HtrA2 function is regulated. The human high-temperature requirement A2 (HtrA2) protein is a trimeric protease that cleaves misfolded proteins to protect cells from stresses caused by toxic, proteinaceous aggregates, and the aberrant function of HtrA2 is closely related to the onset of neurodegenerative disorders. Our methyl-transverse relaxation optimized spectroscopy (TROSY)–based NMR studies using small-peptide ligands have previously revealed a stepwise activation mechanism involving multiple distinct conformational states. However, very little is known about how HtrA2 binds to protein substrates and if the distinct conformational states observed in previous peptide studies might be involved in the processing of protein clients. Herein, we use solution-based NMR spectroscopy to investigate the interaction between the N-terminal Src homology 3 domain from downstream of receptor kinase (drk) with an added C-terminal HtrA2-binding motif (drkN SH3-PDZbm) that exhibits marginal folding stability and serves as a mimic of a physiological protein substrate. We show that drkN SH3-PDZbm binds to HtrA2 via a two-pronged interaction, involving both its C-terminal PDZ-domain binding motif and a central hydrophobic region, with binding occurring preferentially via an unfolded ensemble of substrate molecules. Multivalent interactions between several clients and a single HtrA2 trimer significantly stimulate the catalytic activity of HtrA2, suggesting that binding avidity plays an important role in regulating substrate processing. Our results provide a thermodynamic, kinetic, and structural description of the interaction of HtrA2 with protein substrates and highlight the importance of a trimeric architecture for function as a stress-protective protease that mitigates aggregation.
Collapse
|
19
|
Abraham ET, Oecal S, Mörgelin M, Schmid PWN, Buchner J, Baumann U, Gebauer JM. Collagen's primary structure determines collagen:HSP47 complex stoichiometry. J Biol Chem 2021; 297:101169. [PMID: 34487762 PMCID: PMC8626583 DOI: 10.1016/j.jbc.2021.101169] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/27/2021] [Accepted: 09/02/2021] [Indexed: 11/21/2022] Open
Abstract
Collagens play important roles in development and homeostasis in most higher organisms. In order to function, collagens require the specific chaperone HSP47 for proper folding and secretion. HSP47 is known to bind to the collagen triple helix, but the exact positions and numbers of binding sites are not clear. Here, we employed a collagen II peptide library to characterize high-affinity binding sites for HSP47. We show that many previously predicted binding sites have very low affinities due to the presence of a negatively charged amino acid in the binding motif. In contrast, large hydrophobic amino acids such as phenylalanine at certain positions in the collagen sequence increase binding strength. For further characterization, we determined two crystal structures of HSP47 bound to peptides containing phenylalanine or leucine. These structures deviate significantly from previously published ones in which different collagen sequences were used. They reveal local conformational rearrangements of HSP47 at the binding site to accommodate the large hydrophobic side chain from the middle strand of the collagen triple helix and, most surprisingly, possess an altered binding stoichiometry in the form of a 1:1 complex. This altered stoichiometry is explained by steric collisions with the second HSP47 molecule present in all structures determined thus far caused by the newly introduced large hydrophobic residue placed on the trailing strand. This exemplifies the importance of considering all three sites of homotrimeric collagen as independent interaction surfaces and may provide insight into the formation of higher oligomeric complexes at promiscuous collagen-binding sites.
Collapse
Affiliation(s)
- Elena T Abraham
- Faculty of Mathematics and Natural Sciences, Institute of Biochemistry, University of Cologne, Cologne, Germany
| | - Sinan Oecal
- Faculty of Mathematics and Natural Sciences, Institute of Biochemistry, University of Cologne, Cologne, Germany
| | - Matthias Mörgelin
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden; Colzyx AB, Lund, Sweden
| | - Philipp W N Schmid
- Department of Chemistry, Center for Integrated Protein Science, Technische Universität München, Garching, Germany
| | - Johannes Buchner
- Department of Chemistry, Center for Integrated Protein Science, Technische Universität München, Garching, Germany
| | - Ulrich Baumann
- Faculty of Mathematics and Natural Sciences, Institute of Biochemistry, University of Cologne, Cologne, Germany.
| | - Jan M Gebauer
- Faculty of Mathematics and Natural Sciences, Institute of Biochemistry, University of Cologne, Cologne, Germany.
| |
Collapse
|
20
|
Lam JH, Khan AK, Cornell TA, Chia TW, Dress RJ, Yeow WWW, Mohd-Ismail NK, Venkataraman S, Ng KT, Tan YJ, Anderson DE, Ginhoux F, Nallani M. Polymersomes as Stable Nanocarriers for a Highly Immunogenic and Durable SARS-CoV-2 Spike Protein Subunit Vaccine. ACS NANO 2021; 15:15754-15770. [PMID: 34618423 PMCID: PMC8525042 DOI: 10.1021/acsnano.1c01243] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 09/30/2021] [Indexed: 05/05/2023]
Abstract
Multiple successful vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are urgently needed to address the ongoing coronavirus disease 2019 (Covid-19) pandemic. In the present work, we describe a subunit vaccine based on the SARS-CoV-2 spike protein coadministered with CpG adjuvant. To enhance the immunogenicity of our formulation, both antigen and adjuvant were encapsulated with our proprietary artificial cell membrane (ACM) polymersome technology. Structurally, ACM polymersomes are self-assembling nanoscale vesicles made up of an amphiphilic block copolymer comprising poly(butadiene)-b-poly(ethylene glycol) and a cationic lipid, 1,2-dioleoyl-3-trimethylammonium-propane. Functionally, ACM polymersomes serve as delivery vehicles that are efficiently taken up by dendritic cells (DC1 and DC2), which are key initiators of the adaptive immune response. Two doses of our formulation elicit robust neutralizing antibody titers in C57BL/6 mice that persist at least 40 days. Furthermore, we confirm the presence of functional memory CD4+ and CD8+ T cells that produce T helper type 1 cytokines. This study is an important step toward the development of an efficacious vaccine in humans.
Collapse
Affiliation(s)
| | - Amit K. Khan
- ACM
Biolabs Pte Ltd, Singapore 638075, Singapore
| | | | | | - Regine J. Dress
- Singapore Immunology
Network, Agency for Science, Technology and Research, Singapore 138648, Singapore
| | | | - Nur Khairiah Mohd-Ismail
- Infectious
Diseases Translational Research Program, Department of Microbiology
and Immunology, Yong Loo Lin School of Medicine, National University
Health System, National University of Singapore, Singapore 117545, Singapore
| | | | - Kim Tien Ng
- Infectious
Diseases Translational Research Program, Department of Microbiology
and Immunology, Yong Loo Lin School of Medicine, National University
Health System, National University of Singapore, Singapore 117545, Singapore
| | - Yee-Joo Tan
- Infectious
Diseases Translational Research Program, Department of Microbiology
and Immunology, Yong Loo Lin School of Medicine, National University
Health System, National University of Singapore, Singapore 117545, Singapore
- Institute
of Molecular and Cell Biology, Agency for Science, Technology
and Research, Singapore 138673, Singapore
| | - Danielle E. Anderson
- Program
in Emerging Infectious Diseases, Duke-NUS
Medical School, Singapore 169857, Singapore
| | - Florent Ginhoux
- Singapore Immunology
Network, Agency for Science, Technology and Research, Singapore 138648, Singapore
- SingHealth
Translational Immunology and Inflammation Centre, Singapore 169856, Singapore
| | | |
Collapse
|
21
|
Fassini D, Wilkie IC, Pozzolini M, Ferrario C, Sugni M, Rocha MS, Giovine M, Bonasoro F, Silva TH, Reis RL. Diverse and Productive Source of Biopolymer Inspiration: Marine Collagens. Biomacromolecules 2021; 22:1815-1834. [PMID: 33835787 DOI: 10.1021/acs.biomac.1c00013] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Marine biodiversity is expressed through the huge variety of vertebrate and invertebrate species inhabiting intertidal to deep-sea environments. The extraordinary variety of "forms and functions" exhibited by marine animals suggests they are a promising source of bioactive molecules and provides potential inspiration for different biomimetic approaches. This diversity is familiar to biologists and has led to intensive investigation of metabolites, polysaccharides, and other compounds. However, marine collagens are less well-known. This review will provide detailed insight into the diversity of collagens present in marine species in terms of their genetics, structure, properties, and physiology. In the last part of the review the focus will be on the most common marine collagen sources and on the latest advances in the development of innovative materials exploiting, or inspired by, marine collagens.
Collapse
Affiliation(s)
- Dario Fassini
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Iain C Wilkie
- Institute of Biodiversity Animal Health & Comparative Medicine, University of Glasgow, Glasgow G12 8QQ, Scotland
| | - Marina Pozzolini
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, Via Pastore 3, 16132 Genova, Italy
| | - Cinzia Ferrario
- Dipartimento di Scienze e Politiche Ambientali, Università degli Studi di Milano, Milano, Italy, Center for Complexity & Biosystems, Dipartimento di Fisica, Università degli Studi di Milano, 20122 Milano, Italy
| | - Michela Sugni
- Dipartimento di Scienze e Politiche Ambientali, Università degli Studi di Milano, Milano, Italy, Center for Complexity & Biosystems, Dipartimento di Fisica, Università degli Studi di Milano, 20122 Milano, Italy
| | - Miguel S Rocha
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Marco Giovine
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, Via Pastore 3, 16132 Genova, Italy
| | - Francesco Bonasoro
- Dipartimento di Scienze e Politiche Ambientali, Università degli Studi di Milano, Milano, Italy, Center for Complexity & Biosystems, Dipartimento di Fisica, Università degli Studi di Milano, 20122 Milano, Italy
| | - Tiago H Silva
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| |
Collapse
|
22
|
Koivunen J, Tu H, Kemppainen A, Anbazhagan P, Finnilä MA, Saarakkala S, Käpylä J, Lu N, Heikkinen A, Juffer AH, Heino J, Gullberg D, Pihlajaniemi T. Integrin α11β1 is a receptor for collagen XIII. Cell Tissue Res 2021; 383:1135-1153. [PMID: 33306155 PMCID: PMC7960628 DOI: 10.1007/s00441-020-03300-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/14/2020] [Indexed: 12/28/2022]
Abstract
Collagen XIII is a conserved transmembrane collagen mainly expressed in mesenchymal tissues. Previously, we have shown that collagen XIII modulates tissue development and homeostasis. Integrins are a family of receptors that mediate signals from the environment into the cells and vice versa. Integrin α11β1 is a collagen receptor known to recognize the GFOGER (O=hydroxyproline) sequence in collagens. Interestingly, collagen XIII and integrin α11β1 both have a role in the regulation of bone homeostasis. To study whether α11β1 is a receptor for collagen XIII, we utilized C2C12 cells transfected to express α11β1 as their only collagen receptor. The interaction between collagen XIII and integrin α11β1 was also confirmed by surface plasmon resonance and pull-down assays. We discovered that integrin α11β1 mediates cell adhesion to two collagenous motifs, namely GPKGER and GF(S)QGEK, that were shown to act as the recognition sites for the integrin α11-I domain. Furthermore, we studied the in vivo significance of the α11β1-collagen XIII interaction by crossbreeding α11 null mice (Itga11-/-) with mice overexpressing Col13a1 (Col13a1oe). When we evaluated the bone morphology by microcomputed tomography, Col13a1oe mice had a drastic bone overgrowth followed by severe osteoporosis, whereas the double mutant mouse line showed a much milder bone phenotype. To conclude, our data identifies integrin α11β1 as a new collagen XIII receptor and demonstrates that this ligand-receptor pair has a role in the maintenance of bone homeostasis.
Collapse
Affiliation(s)
- Jarkko Koivunen
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5400, FIN-90014, Oulu, Finland
| | - Hongmin Tu
- Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5000, FIN-90014, Oulu, Finland
| | - Antti Kemppainen
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5400, FIN-90014, Oulu, Finland
| | - Padmanabhan Anbazhagan
- Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5000, FIN-90014, Oulu, Finland
| | - Mikko A Finnilä
- Research Unit of Medical Imaging, Physics and Technology, University of Oulu, P.O. Box 5000, FIN-90014, Oulu, Finland
| | - Simo Saarakkala
- Research Unit of Medical Imaging, Physics and Technology, University of Oulu, P.O. Box 5000, FIN-90014, Oulu, Finland
| | - Jarmo Käpylä
- Department of Biochemistry and MediCity Research Laboratory, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | - Ning Lu
- Department of Biomedicine and Center of Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, N-5009, Bergen, Norway
| | - Anne Heikkinen
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5400, FIN-90014, Oulu, Finland
| | - André H Juffer
- Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5000, FIN-90014, Oulu, Finland
| | - Jyrki Heino
- Department of Biochemistry and MediCity Research Laboratory, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | - Donald Gullberg
- Department of Biomedicine and Center of Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, N-5009, Bergen, Norway
| | - Taina Pihlajaniemi
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5400, FIN-90014, Oulu, Finland.
| |
Collapse
|
23
|
The Versatile Manipulations of Self-Assembled Proteins in Vaccine Design. Int J Mol Sci 2021; 22:ijms22041934. [PMID: 33669238 PMCID: PMC7919822 DOI: 10.3390/ijms22041934] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/06/2021] [Accepted: 02/11/2021] [Indexed: 12/16/2022] Open
Abstract
Protein assemblies provide unique structural features which make them useful as carrier molecules in biomedical and chemical science. Protein assemblies can accommodate a variety of organic, inorganic and biological molecules such as small proteins and peptides and have been used in development of subunit vaccines via display parts of viral pathogens or antigens. Such subunit vaccines are much safer than traditional vaccines based on inactivated pathogens which are more likely to produce side-effects. Therefore, to tackle a pandemic and rapidly produce safer and more effective subunit vaccines based on protein assemblies, it is necessary to understand the basic structural features which drive protein self-assembly and functionalization of portions of pathogens. This review highlights recent developments and future perspectives in production of non-viral protein assemblies with essential structural features of subunit vaccines.
Collapse
|
24
|
Ziegler SJ, Mallinson SJ, St. John PC, Bomble YJ. Advances in integrative structural biology: Towards understanding protein complexes in their cellular context. Comput Struct Biotechnol J 2020; 19:214-225. [PMID: 33425253 PMCID: PMC7772369 DOI: 10.1016/j.csbj.2020.11.052] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 11/25/2020] [Accepted: 11/28/2020] [Indexed: 01/26/2023] Open
Abstract
Microorganisms rely on protein interactions to transmit signals, react to stimuli, and grow. One of the best ways to understand these protein interactions is through structural characterization. However, in the past, structural knowledge was limited to stable, high-affinity complexes that could be crystallized. Recent developments in structural biology have revolutionized how protein interactions are characterized. The combination of multiple techniques, known as integrative structural biology, has provided insight into how large protein complexes interact in their native environment. In this mini-review, we describe the past, present, and potential future of integrative structural biology as a tool for characterizing protein interactions in their cellular context.
Collapse
Key Words
- CLEM, correlated light and electron microscopy
- Crosslinking mass spectrometry
- Cryo-electron microscopy
- Cryo-electron tomography
- EPR, electron paramagnetic resonance
- FRET, Forster resonance energy transfer
- ISB, Integrative structural biology
- Integrative structural biology
- ML, machine learning
- MR, molecular replacement
- MSAs, multiple sequence alignments
- MX, macromolecular crystallography
- NMR, nuclear magnetic resonance
- PDB, Protein Data Bank
- Protein docking
- Protein structure prediction
- Quinary interactions
- SAD, single-wavelength anomalous dispersion
- SANS, small angle neutron scattering
- SAXS, small angle X-ray scattering
- X-ray crystallography
- XL-MS, cross-linking mass spectrometry
- cryo-EM SPA, cryo-EM single particle analysis
- cryo-EM, cryo-electron microscopy
- cryo-ET, cryo-electron tomography
Collapse
Affiliation(s)
- Samantha J. Ziegler
- Biosciences Center, National Renewable Energy Laboratory, 15013 Denver West Parkway, Golden, CO 80401, USA
| | - Sam J.B. Mallinson
- Biosciences Center, National Renewable Energy Laboratory, 15013 Denver West Parkway, Golden, CO 80401, USA
| | - Peter C. St. John
- Biosciences Center, National Renewable Energy Laboratory, 15013 Denver West Parkway, Golden, CO 80401, USA
| | - Yannick J. Bomble
- Biosciences Center, National Renewable Energy Laboratory, 15013 Denver West Parkway, Golden, CO 80401, USA
| |
Collapse
|
25
|
Ting-Hui-Lin, Chia MY, Lin CY, Yeh YQ, Jeng US, Wu WG, Lee MS. Improving immunogenicity of influenza virus H7N9 recombinant hemagglutinin for vaccine development. Vaccine 2020; 37:1897-1903. [PMID: 30857635 DOI: 10.1016/j.vaccine.2018.09.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/08/2018] [Accepted: 09/13/2018] [Indexed: 12/11/2022]
Abstract
Human infections of novel avian influenza A virus (H7N9) emerged in early 2013 and caused about 40% case-fatality through 2017. Therefore, development of influenza H7N9 vaccines is critical for pandemic preparedness. Currently, there are three means of production of commercial influenza vaccines: egg-based, mammalian cell-based, and insect cell-based platforms. The insect cell-based platform has the advantage of high speed in producing recombinant protein. In this study, we evaluate the stability and immunogenicity of two different influenza H7 HA expression constructs generated using the baculovirus system, including membrane-based full-length HA (mH7) and secreted ectodomain-based H7 (sH7). The mH7 construct could form an oligomer-rosette structure and had a high hemagglutinin (HA) titer 8192. In contrast to mH7, the sH7 construct could not form an oligomer-rosette structure and did not have HA titer before cross-linking with anti-His antibody. Thermal stability tests showed that the sH7 and mH7 constructs were unstable at 43 °C and 52 °C, respectively. In a mice immunization study, the mH7 construct but not the sH7 construct could induce robust HI and neutralizing antibody titers. In conclusion, further development of the mH7 vaccine candidate is desirable.
Collapse
Affiliation(s)
- Ting-Hui-Lin
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli County, Taiwan; College of Life Science, National Tsing-Hua University, Hsinchu, Taiwan
| | - Min-Yuan Chia
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli County, Taiwan; Department of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Chun-Yang Lin
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Yi-Qi Yeh
- National Synchrotron Radiation Research Center, Hsinchu, Taiwan
| | - U-Ser Jeng
- National Synchrotron Radiation Research Center, Hsinchu, Taiwan; Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Wen-Guey Wu
- College of Life Science, National Tsing-Hua University, Hsinchu, Taiwan
| | - Min-Shi Lee
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli County, Taiwan.
| |
Collapse
|
26
|
Willis E, Pardi N, Parkhouse K, Mui BL, Tam YK, Weissman D, Hensley SE. Nucleoside-modified mRNA vaccination partially overcomes maternal antibody inhibition of de novo immune responses in mice. Sci Transl Med 2020; 12:eaav5701. [PMID: 31915303 PMCID: PMC7339908 DOI: 10.1126/scitranslmed.aav5701] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 06/21/2019] [Accepted: 09/23/2019] [Indexed: 12/12/2022]
Abstract
Maternal antibodies provide short-term protection to infants against many infections. However, they can inhibit de novo antibody responses in infants elicited by infections or vaccination, leading to increased long-term susceptibility to infectious diseases. Thus, there is a need to develop vaccines that are able to elicit protective immune responses in the presence of antigen-specific maternal antibodies. Here, we used a mouse model to demonstrate that influenza virus-specific maternal antibodies inhibited de novo antibody responses in mouse pups elicited by influenza virus infection or administration of conventional influenza vaccines. We found that a recently developed influenza vaccine, nucleoside-modified mRNA encapsulated in lipid nanoparticles (mRNA-LNP), partially overcame this inhibition by maternal antibodies. The mRNA-LNP influenza vaccine established long-lived germinal centers in the mouse pups and elicited stronger antibody responses than did a conventional influenza vaccine approved for use in humans. Vaccination with mRNA-LNP vaccines may offer a promising strategy for generating robust immune responses in infants in the presence of maternal antibodies.
Collapse
Affiliation(s)
- Elinor Willis
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Norbert Pardi
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kaela Parkhouse
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Ying K Tam
- Acuitas Therapeutics, Vancouver, BC V6T 1Z3, Canada
| | - Drew Weissman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Scott E Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
27
|
Joyce MG, Bao A, Chen M, Georgiev IS, Ou L, Bylund T, Druz A, Kong WP, Peng D, Rundlet EJ, Van Galen JG, Wang S, Yang Y, Zhang B, Chuang GY, McLellan JS, Graham BS, Mascola JR, Kwong PD. Crystal Structure and Immunogenicity of the DS-Cav1-Stabilized Fusion Glycoprotein From Respiratory Syncytial Virus Subtype B. Pathog Immun 2019; 4:294-323. [PMID: 31893251 PMCID: PMC6922093 DOI: 10.20411/pai.v4i2.338] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 11/21/2019] [Indexed: 11/23/2022] Open
Abstract
Background: Respiratory syncytial virus (RSV) subtypes, A and B, co-circulate in annual epidemics and alternate in dominance. We have shown that a subtype A RSV fusion (F) glycoprotein, stabilized in its prefusion conformation by DS-Cav1 mutations, is a promising RSV-vaccine immunogen, capable of boosting RSV-neutralizing titers in healthy adults. In both humans and vaccine-tested animals, neutralizing titers elicited by this subtype A DS-Cav1 immunogen were ~ 2- to 3-fold higher against the homologous subtype A virus than against the heterologous subtype B virus. Methods: To understand the molecular basis for this subtype difference, we introduced DS-Cav1 mutations into RSV strain B18537 F, determined the trimeric crystal structure, and carried out immunogenicity studies. Results: The B18537 DS-Cav1 F structure at 2-Å resolution afforded a precise delineation of prefusion F characteristics, including those of antigenic site Ø, a key trimer-apex site. Structural comparison with the subtype A prefusion F indicated 11% of surface residues to be different, with an alpha-carbon root-mean-square deviation (RMSD) of 1.2 Å; antigenic site Ø, however, differed in 23% of its surface residues and had an alpha-carbon RMSD of 2.2 Å. Immunization of vaccine-tested animals with DS-Cav1-stabilized B18537 F induced neutralizing responses ~100-fold higher than with postfusion B18537 F. Notably, elicited responses neutralized RSV subtypes A and B at similar levels and were directed towards both conserved equatorial and diverse apical regions. Conclusion: We propose that structural differences in apical and equatorial sites–coupled to differently focused immune responses–provide a molecular explanation for observed differences in elicited subtype A and B neutralizing responses.
Collapse
Affiliation(s)
- M Gordon Joyce
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland
| | - Amy Bao
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Man Chen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Ivelin S Georgiev
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland.,Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, Tennessee
| | - Li Ou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Tatsiana Bylund
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Aliaksandr Druz
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Wing-Pui Kong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Dongjun Peng
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Emily J Rundlet
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Joseph G Van Galen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Shuishu Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Yongping Yang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Baoshan Zhang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Gwo-Yu Chuang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jason S McLellan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland.,Department of Molecular Biosciences, College of Natural Sciences, The University of Texas at Austin, Austin, Texas
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
28
|
Köhler M, Marchand A, Hentzen NB, Egli J, Begley AI, Wennemers H, Zenobi R. Temperature-controlled electrospray ionization mass spectrometry as a tool to study collagen homo- and heterotrimers. Chem Sci 2019; 10:9829-9835. [PMID: 32015805 PMCID: PMC6977553 DOI: 10.1039/c9sc03248g] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 08/29/2019] [Indexed: 12/13/2022] Open
Abstract
Collagen model peptides are useful for understanding the assembly and structure of collagen triple helices. The design of self-assembling heterotrimeric helices is particularly challenging and often affords mixtures of non-covalent assemblies that are difficult to characterize by conventional NMR and CD spectroscopic techniques. This can render a detailed understanding of the factors that control heterotrimer formation difficult and restrict rational design. Here, we present a novel method based on electrospray ionization mass spectrometry to investigate homo- and heterotrimeric collagen model peptides. Under native conditions, the high resolving power of mass spectrometry was used to access the stoichiometric composition of different triple helices in complex mixtures. A temperature-controlled electrospray ionization source was built to perform thermal denaturation experiments and provided melting temperatures of triple helices. These were found to be in good agreement with values obtained from CD spectroscopic measurements. Importantly, for mixtures of coexisting homo- and heterotrimers, which are difficult to analyze by conventional methods, our technique allowed for the identification and monitoring of the unfolding of each individual species. Their respective melting temperatures could easily be accessed in a single experiment, using small amounts of sample.
Collapse
Affiliation(s)
- Martin Köhler
- Department of Chemistry and Applied Biosciences , ETH Zurich , 8093 , Zurich , Switzerland . ;
| | - Adrien Marchand
- Department of Chemistry and Applied Biosciences , ETH Zurich , 8093 , Zurich , Switzerland . ;
| | - Nina B Hentzen
- Department of Chemistry and Applied Biosciences , ETH Zurich , 8093 , Zurich , Switzerland . ;
| | - Jasmine Egli
- Department of Chemistry and Applied Biosciences , ETH Zurich , 8093 , Zurich , Switzerland . ;
| | - Alina I Begley
- Department of Chemistry and Applied Biosciences , ETH Zurich , 8093 , Zurich , Switzerland . ;
| | - Helma Wennemers
- Department of Chemistry and Applied Biosciences , ETH Zurich , 8093 , Zurich , Switzerland . ;
| | - Renato Zenobi
- Department of Chemistry and Applied Biosciences , ETH Zurich , 8093 , Zurich , Switzerland . ;
| |
Collapse
|
29
|
Abstract
The combination of supramolecular aggregation of collagen model peptides with reversible covalent end‐capping of the formed triple helix in a single experimental set‐up yielded minicollagens, which were characterized by a single melting temperature. In spite of the numerous possible reaction intermediates, a specific synthetic collagen with a leading, middle and trailing strand is formed in a highly cooperative self‐assembly process.
Collapse
Affiliation(s)
- Christoph Priem
- Department of Chemistry, Philipps-Universität Marburg, Hans-Meerwein-Straße 4, 35032, Marburg, Germany
| | - Armin Geyer
- Department of Chemistry, Philipps-Universität Marburg, Hans-Meerwein-Straße 4, 35032, Marburg, Germany
| |
Collapse
|
30
|
Ito S, Saito M, Yoshida M, Takeuchi K, Doi T, Nagata K. A BRET-based assay reveals collagen-Hsp47 interaction dynamics in the endoplasmic reticulum and small-molecule inhibition of this interaction. J Biol Chem 2019; 294:15962-15972. [PMID: 31492754 PMCID: PMC6827286 DOI: 10.1074/jbc.ra119.010567] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 08/23/2019] [Indexed: 01/01/2023] Open
Abstract
Molecular chaperones perform pivotal roles in proteostasis by engaging in protein–protein interactions (PPIs). The collagen-specific molecular chaperone Hsp47 (heat shock protein 47) interacts with procollagen in the endoplasmic reticulum (ER) and plays crucial roles in collagen synthesis. PPIs between Hsp47 and collagen could offer a therapeutic target for fibrosis, which is characterized by abnormal collagen accumulation in the extracellular matrix of fibrotic organs. Herein, we established a bioluminescence resonance energy transfer (BRET) system for assessing Hsp47–collagen interaction dynamics within the ER. After optimization and validation of the method, we could demonstrate inhibition of the interaction between Hsp47 and collagen by a small molecule (Col003) in the ER. Using the BRET system, we also found that Hsp47 interacts not only with the Gly-Pro-Arg motif but also weakly with Gly-Pro-Hyp motifs of triple-helical collagen in cells. Moreover, we found that the serpin loop of Hsp47 (SerpinH1) contributes to its binding to collagen. We propose that the method developed here can provide valuable information on PPIs between Hsp47 and collagen and on the effects of PPI inhibitors important for the management of fibrotic disorders.
Collapse
Affiliation(s)
- Shinya Ito
- Institute for Protein Dynamics, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Masazumi Saito
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Masahito Yoshida
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Koh Takeuchi
- National Institute of Advanced Industrial Science and Technology, Tokyo 135-0064, Japan
| | - Takayuki Doi
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Kazuhiro Nagata
- Institute for Protein Dynamics, Kyoto Sangyo University, Kyoto 603-8555, Japan .,Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan.,CREST, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan
| |
Collapse
|
31
|
Gellermann P, Schneider-Barthold C, Bolten SN, Overfelt E, Scheper T, Pepelanova I. Production of a Recombinant Non-Hydroxylated Gelatin Mimetic in Pichia pastoris for Biomedical Applications. J Funct Biomater 2019; 10:E39. [PMID: 31480684 PMCID: PMC6787575 DOI: 10.3390/jfb10030039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/23/2019] [Accepted: 08/29/2019] [Indexed: 01/10/2023] Open
Abstract
Proteins derived from the natural extracellular matrix like collagen or gelatin are common in clinical research, where they are prized for their biocompatibility and bioactivity. Cells are able to adhere, grow and remodel scaffolds based on these materials. Usually, collagen and gelatin are sourced from animal material, risking pathogenic transmission and inconsistent batch-to-batch product quality. A recombinant production in yeast circumvents these disadvantages by ensuring production with a reproducible quality in animal-component-free media. A gelatin mimetic protein, based on the alpha chain of human collagen I, was cloned in Pichia pastoris under the control of the methanol-inducible alcohol oxidase (AOX1) promoter. A producing clone was selected and cultivated at the 30 L scale. The protein was secreted into the cultivation medium and the final yield was 3.4 g·L-1. Purification of the target was performed directly from the cell-free medium by size exclusion chromatography. The gelatin mimetic protein was tested in cell culture for biocompatibility and for promoting cell adhesion. It supported cell growth and its performance was indistinguishable from animal-derived gelatin. The gelatin-mimetic protein represents a swift strategy to produce recombinant and human-based extracellular matrix proteins for various biomedical applications.
Collapse
Affiliation(s)
- Pia Gellermann
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 5, 30167 Hannover, Germany
| | | | - Svenja Nicolin Bolten
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 5, 30167 Hannover, Germany
| | - Ethan Overfelt
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 5, 30167 Hannover, Germany
| | - Thomas Scheper
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 5, 30167 Hannover, Germany
| | - Iliyana Pepelanova
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 5, 30167 Hannover, Germany.
| |
Collapse
|
32
|
Werten MWT, Eggink G, Cohen Stuart MA, de Wolf FA. Production of protein-based polymers in Pichia pastoris. Biotechnol Adv 2019; 37:642-666. [PMID: 30902728 PMCID: PMC6624476 DOI: 10.1016/j.biotechadv.2019.03.012] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 02/03/2019] [Accepted: 03/17/2019] [Indexed: 01/09/2023]
Abstract
Materials science and genetic engineering have joined forces over the last three decades in the development of so-called protein-based polymers. These are proteins, typically with repetitive amino acid sequences, that have such physical properties that they can be used as functional materials. Well-known natural examples are collagen, silk, and elastin, but also artificial sequences have been devised. These proteins can be produced in a suitable host via recombinant DNA technology, and it is this inherent control over monomer sequence and molecular size that renders this class of polymers of particular interest to the fields of nanomaterials and biomedical research. Traditionally, Escherichia coli has been the main workhorse for the production of these polymers, but the methylotrophic yeast Pichia pastoris is finding increased use in view of the often high yields and potential bioprocessing benefits. We here provide an overview of protein-based polymers produced in P. pastoris. We summarize their physicochemical properties, briefly note possible applications, and detail their biosynthesis. Some challenges that may be faced when using P. pastoris for polymer production are identified: (i) low yields and poor process control in shake flask cultures; i.e., the need for bioreactors, (ii) proteolytic degradation, and (iii) self-assembly in vivo. Strategies to overcome these challenges are discussed, which we anticipate will be of interest also to readers involved in protein expression in P. pastoris in general.
Collapse
Affiliation(s)
- Marc W T Werten
- Wageningen Food & Biobased Research, NL-6708 WG Wageningen, The Netherlands.
| | - Gerrit Eggink
- Wageningen Food & Biobased Research, NL-6708 WG Wageningen, The Netherlands; Bioprocess Engineering, Wageningen University & Research, NL-6708 PB Wageningen, The Netherlands
| | - Martien A Cohen Stuart
- Physical Chemistry and Soft Matter, Wageningen University & Research, NL-6708 WE Wageningen, The Netherlands
| | - Frits A de Wolf
- Wageningen Food & Biobased Research, NL-6708 WG Wageningen, The Netherlands
| |
Collapse
|
33
|
Kubyshkin V. Stabilization of the triple helix in collagen mimicking peptides. Org Biomol Chem 2019; 17:8031-8047. [PMID: 31464337 DOI: 10.1039/c9ob01646e] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Collagen mimics are peptides designed to reproduce structural features of natural collagen. A triple helix is the first element in the hierarchy of collagen folding. It is an assembly of three parallel peptide chains stabilized by packing and interchain hydrogen bonds. In this review we summarize the existing chemical approaches towards stabilization of this structure including the most recent developments. Currently proposed methods include manipulation of the amino acid composition, application of unnatural amino acid analogues, stimuli-responsive modifications, chain tethering approaches, peptide amphiphiles, modifications that target interchain interactions and more. This ability to manipulate the triple helix as a supramolecular self-assembly contributes to our understanding of the collagen folding. It also provides essential information needed to design collagen-based biomaterials of the future.
Collapse
Affiliation(s)
- Vladimir Kubyshkin
- Institute of Chemistry, University of Manitoba, Dysart Rd. 144, R3T 2N2, Winnipeg, Manitoba, Canada.
| |
Collapse
|
34
|
Cullen LM, Schmidt MR, Torres GM, Capoferri AA, Morrison TG. Comparison of Immune Responses to Different Versions of VLP Associated Stabilized RSV Pre-Fusion F Protein. Vaccines (Basel) 2019; 7:vaccines7010021. [PMID: 30769923 PMCID: PMC6466353 DOI: 10.3390/vaccines7010021] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/07/2019] [Accepted: 02/11/2019] [Indexed: 11/16/2022] Open
Abstract
Efforts to develop a vaccine for respiratory syncytial virus (RSV) have primarily focused on the RSV fusion protein. The pre-fusion conformation of this protein induces the most potent neutralizing antibodies and is the focus of recent efforts in vaccine development. Following the first identification of mutations in the RSV F protein (DS-Cav1 mutant protein) that stabilized the pre-fusion conformation, other mutant stabilized pre-fusion F proteins have been described. To determine if there are differences in alternate versions of stabilized pre-fusion F proteins, we explored the use, as vaccine candidates, of virus-like particles (VLPs) containing five different pre-fusion F proteins, including the DS-Cav1 protein. The expression of these five pre-F proteins, their assembly into VLPs, their pre-fusion conformation stability in VLPs, their reactivity with anti-F monoclonal antibodies, and their induction of immune responses after the immunization of mice, were characterized, comparing VLPs containing the DS-Cav1 pre-F protein with VLPs containing four alternative pre-fusion F proteins. The concentrations of anti-F IgG induced by each VLP that blocked the binding of prototype monoclonal antibodies using two different soluble pre-fusion F proteins as targets were measured. Our results indicate that both the conformation and immunogenicity of alternative VLP associated stabilized pre-fusion RSV F proteins are different from those of DS-Cav1 VLPs.
Collapse
Affiliation(s)
- Lori M Cullen
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | - Madelyn R Schmidt
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655, USA.
- Program of Immunology and Microbiology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | - Gretel M Torres
- Program of Immunology and Microbiology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | - Adam A Capoferri
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | - Trudy G Morrison
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655, USA.
- Program of Immunology and Microbiology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| |
Collapse
|
35
|
Strawn R, Chen F, Jeet Haven P, Wong S, Park-Arias A, De Leeuw M, Xu Y. To achieve self-assembled collagen mimetic fibrils using designed peptides. Biopolymers 2018; 109:e23226. [PMID: 30133697 DOI: 10.1002/bip.23226] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 04/14/2018] [Accepted: 04/16/2018] [Indexed: 11/07/2022]
Abstract
It has proven challenging to obtain collagen-mimetic fibrils by protein design. We recently reported the self-assembly of a mini-fibril showing a 35 nm, D-period like, axially repeating structure using the designed triple helix Col108. Peptide Col108 was made by bacterial expression using a synthetic gene; its triple helix domain consists of three pseudo-identical units of amino acid sequence arranged in tandem. It was postulated that the 35 nm d-period of Col108 mini-fibrils originates from the periodicity of the Col108 primary structure. A mutual staggering of one sequence unit of the associating Col108 triple helices can maximize the inter-helical interactions and produce the observed 35 nm d-period. Based on this unit-staggered model, a triple helix consisting of only two sequence units is expected to have the potential to form the same d-periodic mini-fibrils. Indeed, when such a peptide, peptide 2U108, was made it was found to self-assemble into mini-fibrils having the same d-period of 35 nm. In contrast, no d-periodic mini-fibrils were observed for peptide 1U108, which does not have long-range repeating sequences in its primary structure. The findings of the periodic mini-fibrils of Col108 and 2U108 suggest a way forward to create collagen-mimetic fibrils for biomedical and industrial applications.
Collapse
Affiliation(s)
- Rebecca Strawn
- SGS, 606 Brandywine Pkwy, West Chester, Pennsylvania 19380, U.S.A
| | - FangFang Chen
- Department of Chemistry, Hunter College, Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, New York 10065, U.S.A
| | - Parminder Jeet Haven
- Department of Chemistry, Hunter College, Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, New York 10065, U.S.A
| | - Sam Wong
- Department of Chemistry, Hunter College, Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, New York 10065, U.S.A
| | - Anne Park-Arias
- Radium2 Capital Inc., 300 RXR Plaza, Uniondale, New York 11556, U.S.A
| | - Monique De Leeuw
- Delft University of Technology, Mekelweg 2, 2628 CD Delft, 347-205-0465, Netherlands
| | - Yujia Xu
- Department of Chemistry, Hunter College, Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, New York 10065, U.S.A
| |
Collapse
|
36
|
Ornelas A, Williams KN, Hatch KA, Paez A, Aguilar AC, Ellis CC, Tasnim N, Ray S, Dirk CW, Boland T, Joddar B, Li C, Michael K. Synthesis and characterization of a photocleavable collagen-like peptide. Org Biomol Chem 2018; 16:1000-1013. [PMID: 29345707 PMCID: PMC5951682 DOI: 10.1039/c7ob02198d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A 34-amino acid long collagen-like peptide rich in proline, hydroxyproline, and glycine, and with four photoreactive N-acyl-7-nitroindoline units incorporated into the peptide backbone was synthesized by on-resin fragment condensation. Its circular dichroism supports a stable triple helix structure. The built-in photochemical function enables the decomposition of the peptide into small peptide fragments by illumination with UV light of 350 nm in aqueous solution. Illumination of a thin film of the peptide, or a thin film of a photoreactive amino acid model compound containing a 5-bromo-7-nitroindoline moiety, with femtosecond laser light at 710 nm allows for the creation of well-resolved micropatterns. The cytocompatibility of the peptide was demonstrated using human mesenchymal stem cells and mouse embryonic fibroblasts. Our data show that the full-length peptide is cytocompatible as it can support cell growth and maintain cell viability. In contrast, the small peptide fragments created by photolysis are somewhat cytotoxic and therefore less cytocompatible. These data suggest that biomimetic collagen-like photoreactive peptides could potentially be used for growing cells in 2D micropatterns based on patterns generated by photolysis prior to cell growth.
Collapse
Affiliation(s)
- Alfredo Ornelas
- Department of Chemistry and Biochemistry, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Liu H, Su D, Zhang J, Ge S, Li Y, Wang F, Gravel M, Roulston A, Song Q, Xu W, Liang JG, Shore G, Wang X, Liang P. Improvement of Pharmacokinetic Profile of TRAIL via Trimer-Tag Enhances its Antitumor Activity in vivo. Sci Rep 2017; 7:8953. [PMID: 28827692 PMCID: PMC5566391 DOI: 10.1038/s41598-017-09518-1] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 07/24/2017] [Indexed: 12/11/2022] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL/Apo2L) has long been considered a tantalizing target for cancer therapy because it mediates activation of the extrinsic apoptosis pathway in a tumor-specific manner by binding to and trimerizing its functional receptors DR4 or DR5. Despite initial promise, both recombinant human TRAIL (native TRAIL) and dimeric DR4/DR5 agonist monoclonal antibodies (mAbs) failed in multiple human clinical trials. Here we show that in-frame fusion of human C-propeptide of α1(I) collagen (Trimer-Tag) to the C-terminus of mature human TRAIL leads to a disulfide bond-linked homotrimer which can be expressed at high levels as a secreted protein from CHO cells. The resulting TRAIL-Trimer not only retains similar bioactivity and receptor binding kinetics as native TRAIL in vitro which are 4-5 orders of magnitude superior to that of dimeric TRAIL-Fc, but also manifests more favorable pharmacokinetic and antitumor pharmacodynamic profiles in vivo than that of native TRAIL. Taken together, this work provides direct evidence for the in vivo antitumor efficacy of TRAIL being proportional to systemic drug exposure and suggests that the previous clinical failures may have been due to rapid systemic clearance of native TRAIL and poor apoptosis-inducing potency of dimeric agonist mAbs despite their long serum half-lives.
Collapse
Affiliation(s)
- Haipeng Liu
- Department of Biochemistry & Molecular Biology, College of Life Sciences, Sichuan University, Chengdu, China
| | - Danmei Su
- Department of Biochemistry & Molecular Biology, College of Life Sciences, Sichuan University, Chengdu, China
| | - Jinlong Zhang
- Department of Biochemistry & Molecular Biology, College of Life Sciences, Sichuan University, Chengdu, China
| | - Shuaishuai Ge
- Department of Biochemistry & Molecular Biology, College of Life Sciences, Sichuan University, Chengdu, China
| | - Youwei Li
- Department of Biochemistry & Molecular Biology, College of Life Sciences, Sichuan University, Chengdu, China
| | - Fei Wang
- Department of Biochemistry & Molecular Biology, College of Life Sciences, Sichuan University, Chengdu, China
| | - Michel Gravel
- Laboratory for Therapeutic Development, Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal (QC), Canada
| | - Anne Roulston
- Laboratory for Therapeutic Development, Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal (QC), Canada
| | - Qin Song
- Clover Biopharmaceuticals, Chengdu, China
| | - Wei Xu
- Clover Biopharmaceuticals, Chengdu, China
| | | | - Gordon Shore
- Laboratory for Therapeutic Development, Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal (QC), Canada
| | - Xiaodong Wang
- National Institute of Biological Sciences, Beijing, China
| | - Peng Liang
- Department of Biochemistry & Molecular Biology, College of Life Sciences, Sichuan University, Chengdu, China.
- Clover Biopharmaceuticals, Chengdu, China.
- GenHunter Corporation, 624 Grassmere Park, Nashville, TN, 37211, USA.
| |
Collapse
|
38
|
Improved Prefusion Stability, Optimized Codon Usage, and Augmented Virion Packaging Enhance the Immunogenicity of Respiratory Syncytial Virus Fusion Protein in a Vectored-Vaccine Candidate. J Virol 2017; 91:JVI.00189-17. [PMID: 28539444 DOI: 10.1128/jvi.00189-17] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 05/05/2017] [Indexed: 12/13/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the most important viral agent of severe pediatric respiratory tract disease worldwide, but it lacks a licensed vaccine or suitable antiviral drug. A live attenuated chimeric bovine/human parainfluenza virus type 3 (rB/HPIV3) was developed previously as a vector expressing RSV fusion (F) protein to confer bivalent protection against RSV and HPIV3. In a previous clinical trial in virus-naive children, rB/HPIV3 was well tolerated but the immunogenicity of wild-type RSV F was unsatisfactory. We previously modified RSV F with a designed disulfide bond (DS) to increase stability in the prefusion (pre-F) conformation and to be efficiently packaged in the vector virion. Here, we further stabilized pre-F by adding both disulfide and cavity-filling mutations (DS-Cav1), and we also modified RSV F codon usage to have a lower CpG content and a higher level of expression. This RSV F open reading frame was evaluated in rB/HPIV3 in three forms: (i) pre-F without vector-packaging signal, (ii) pre-F with vector-packaging signal, and (iii) secreted pre-F ectodomain trimer. Despite being efficiently expressed, the secreted pre-F was poorly immunogenic. DS-Cav1 stabilized pre-F, with or without packaging, induced higher titers of pre-F specific antibodies in hamsters, and improved the quality of RSV-neutralizing serum antibodies. Codon-optimized RSV F containing fewer CpG dinucleotides had higher F expression, replicated more efficiently in vivo, and was more immunogenic. The combination of DS-Cav1 pre-F stabilization, optimized codon usage, reduced CpG content, and vector packaging significantly improved vector immunogenicity and protective efficacy against RSV. This provides an improved vectored RSV vaccine candidate suitable for pediatric clinical evaluation.IMPORTANCE RSV and HPIV3 are the first and second leading viral causes of severe pediatric respiratory disease worldwide. Licensed vaccines or suitable antiviral drugs are not available. We are developing a chimeric rB/HPIV3 vector expressing RSV F as a bivalent RSV/HPIV3 vaccine and have been evaluating means to increase RSV F immunogenicity. In this study, we evaluated the effects of improved stabilization of F in the pre-F conformation and of codon optimization resulting in reduced CpG content and greater pre-F expression. Reduced CpG content dampened the interferon response to infection, promoting higher replication and increased F expression. We demonstrate that improved pre-F stabilization and strategic manipulation of codon usage, together with efficient pre-F packaging into vector virions, significantly increased F immunogenicity in the bivalent RSV/HPIV3 vaccine. The improved immunogenicity included induction of increased titers of high-quality complement-independent antibodies with greater pre-F site Ø binding and greater protection against RSV challenge.
Collapse
|
39
|
Wang X, Ge H, Zhang D, Wu S, Zhang G. Oligomerization triggered by foldon: a simple method to enhance the catalytic efficiency of lichenase and xylanase. BMC Biotechnol 2017; 17:57. [PMID: 28673305 PMCID: PMC5496177 DOI: 10.1186/s12896-017-0380-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 06/28/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Effective and simple methods that lead to higher enzymatic efficiencies are highly sough. Here we proposed a foldon-triggered trimerization of the target enzymes with significantly improved catalytic performances by fusing a foldon domain at the C-terminus of the enzymes via elastin-like polypeptides (ELPs). The foldon domain comprises 27 residues and can forms trimers with high stability. RESULTS Lichenase and xylanase can hydrolyze lichenan and xylan to produce value added products and biofuels, and they have great potentials as biotechnological tools in various industrial applications. We took them as the examples and compared the kinetic parameters of the engineered trimeric enzymes to those of the monomeric and wild type ones. When compared with the monomeric ones, the catalytic efficiency (k cat /K m ) of the trimeric lichenase and xylanase increased 4.2- and 3.9- fold. The catalytic constant (k cat ) of the trimeric lichenase and xylanase increased 1.8- fold and 5.0- fold than their corresponding wild-type counterparts. Also, the specific activities of trimeric lichenase and xylanase increased by 149% and 94% than those of the monomeric ones. Besides, the recovery of the lichenase and xylanase activities increased by 12.4% and 6.1% during the purification process using ELPs as the non-chromatographic tag. The possible reason is the foldon domain can reduce the transition temperature of the ELPs. CONCLUSION The trimeric lichenase and xylanase induced by foldon have advantages in the catalytic performances. Besides, they were easier to purify with increased purification fold and decreased the loss of activities compared to their corresponding monomeric ones. Trimerizing of the target enzymes triggered by the foldon domain could improve their activities and facilitate the purification, which represents a simple and effective enzyme-engineering tool. It should have exciting potentials both in industrial and laboratory scales.
Collapse
Affiliation(s)
- Xinzhe Wang
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, Fujian, 361021, China
| | - Huihua Ge
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, Fujian, 361021, China
| | - Dandan Zhang
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, Fujian, 361021, China
| | - Shuyu Wu
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, Fujian, 361021, China
| | - Guangya Zhang
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, Fujian, 361021, China.
| |
Collapse
|
40
|
Abstract
Collagen-like peptides (CLPs), also known as collagen-mimetic peptides (CMPs), are short synthetic peptides that mimic the triple helical conformation of native collagens. Traditionally, CLPs have been widely used in deciphering the chemical basis for collagen triple helix stabilization, mimicking collagen fibril formation and fabricating other higher-order supramolecular self-assemblies. While CLPs have been used extensively for elucidation of the assembly of native collagens, less work has been reported on the use of CLP-polymer and CLP-peptide conjugates in the production of responsive assemblies. CLP triple helices have been used as physical cross-links in CLP-polymer hydrogels with predesigned thermoresponsiveness. The more recently reported ability of CLP to target native collagens via triple helix hybridization has further inspired the production of CLP-polymer and CLP-peptide bioconjugates and the employment of these conjugates in generating well-defined nanostructures for targeting collagen substrates. This review summarizes the current progress and potential of using CLPs in biomedical arenas and is intended to serve as a general guide for designing CLP-containing biomaterials.
Collapse
Affiliation(s)
| | - Kristi L Kiick
- Delaware Biotechnology Institute , Newark, Delaware 19711, United States
| |
Collapse
|
41
|
Walters CR, Szantai-Kis DM, Zhang Y, Reinert ZE, Horne WS, Chenoweth DM, Petersson EJ. The effects of thioamide backbone substitution on protein stability: a study in α-helical, β-sheet, and polyproline II helical contexts. Chem Sci 2017; 8:2868-2877. [PMID: 28553525 PMCID: PMC5428018 DOI: 10.1039/c6sc05580j] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 01/24/2017] [Indexed: 12/17/2022] Open
Abstract
Thioamides are single atom substitutions of the peptide bond that serve as versatile probes of protein structure. Effective use of thioamides requires a robust understanding of the impact that the substitution has on a protein of interest. However, the thermodynamic effects of thioamide incorporation have only been studied in small structural motifs, and their influence on secondary structure in the context of full-length proteins is not known. Here we describe a comprehensive survey of thioamide substitutions in three benchmark protein systems (calmodulin, the B1 domain of protein G, and collagen) featuring the most prevalent secondary structure motifs: α-helix, β-sheet, and polyproline type II helix. We find that in most cases, effects on thermostability can be understood in terms of the positioning and local environment of the thioamide relative to proximal structural elements and hydrogen bonding networks. These observations set the stage for the rational design of thioamide substituted proteins with predictable stabilities.
Collapse
Affiliation(s)
- Christopher R Walters
- Department of Chemistry , University of Pennsylvania , 231 S. 34th Street , Philadelphia , PA 19104 , USA
| | - D Miklos Szantai-Kis
- Biochemistry and Molecular Biophysics Graduate Group , University of Pennsylvania , 3700 Hamilton Walk , Philadelphia , PA 19104 , USA
| | - Yitao Zhang
- Department of Chemistry , University of Pennsylvania , 231 S. 34th Street , Philadelphia , PA 19104 , USA
| | - Zachary E Reinert
- Department of Chemistry , University of Pittsburgh , 219 Parkman Avenue , Pittsburgh , PA 15260 , USA
| | - W Seth Horne
- Department of Chemistry , University of Pittsburgh , 219 Parkman Avenue , Pittsburgh , PA 15260 , USA
| | - David M Chenoweth
- Department of Chemistry , University of Pennsylvania , 231 S. 34th Street , Philadelphia , PA 19104 , USA
| | - E James Petersson
- Department of Chemistry , University of Pennsylvania , 231 S. 34th Street , Philadelphia , PA 19104 , USA
| |
Collapse
|
42
|
Human antibody 3E1 targets the HA stem region of H1N1 and H5N6 influenza A viruses. Nat Commun 2016; 7:13577. [PMID: 27910950 PMCID: PMC5477490 DOI: 10.1038/ncomms13577] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 10/17/2016] [Indexed: 01/05/2023] Open
Abstract
As influenza A viruses remain a major threat to human health worldwide, the discovery of broadly neutralizing monoclonal antibodies that recognize conserved epitopes would facilitate the development of antibody-based therapeutic strategies. Here we report that a VH4-4-encoded human mAb named 3E1 could neutralize H1 and H5 subtype viruses in vitro and protect mice against the H1N1 and H5N6 viruses by inhibiting the low pH-induced conformational rearrangement of haemagglutinin (HA), hence blocking membrane fusion. The crystal structures of 3E1 Fab in complex with HA of two H1N1 strains reveal that 3E1, with both heavy and light chains, binds to a conserved epitope of the HA stem region, comprising parts of the fusion peptide, the F subdomain and the outermost β-strand preceding helix A. Altogether, these data suggest the potential of 3E1 as a therapeutic drug against H1 and H5 subtype viruses.
Collapse
|
43
|
Zhang T, Liu H, Chen X, Wang Z, Wang S, Qu C, Zhang J, Xu X. Lipidated L2 epitope repeats fused with a single-chain antibody fragment targeting human FcγRI elicited cross-neutralizing antibodies against a broad spectrum of human papillomavirus types. Vaccine 2016; 34:5531-5539. [PMID: 27729176 DOI: 10.1016/j.vaccine.2016.10.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Revised: 09/29/2016] [Accepted: 10/03/2016] [Indexed: 12/24/2022]
Abstract
Numerous types of human papillomaviruses (HPVs) have been identified, and the global burden of diseases associated with HPV infection is remarkable, especially in developing regions. Thus a low-cost broad-spectrum prophylactic vaccine is urgently needed. The N-terminal amino acid 17-36 of HPV 16 L2 protein is confirmed to be a major cross-neutralizing epitope (RG-1 epitope). Monomeric proteins containing RG-1 epitopes and scaffold proteins, such as bacterial thioredoxin or modified IgG1 Fc fragment and L2 epitope fusion protein, induced cross-neutralizing antibodies, arousing the possibility of the development of low-cost monomeric vaccine in bacterial expression system. Here we show that a novel immunogen-scaffold protein containing a lipidated triple-repeat HPV 16RG-1 epitope and a hFcγRI specific single-chain antibody fragment (H22scFv), named LpE3H22, elicited high titers of cross-neutralizing antibodies against a broad range of mucosal and cutaneous HPV types when adjuvanted with MF59 and poly I:C. LpE3H22 was produced in E. coli expression system. In contrast to three repeats of RG-1 epitope (E3) and unlipidated fusion protein E3H22, vaccination of LpE3H22 induced robust cross-neutralizing antibody responses in hFcγRI transgenic mice. Furthermore, the neutralizing antibody response induced by LpE3H22 was significantly weaker in WT mice than in the Tg mice. The cross-neutralizing antibodies induced by LpE3H22 sustained for at least 10months in Tg mice. Our results demonstrate that hFcγRI targeting and lipidation both contribute to the enhancement of immunogenicity of L2 antigen. Therefore, delivering the lipidated L2 antigen with H22scFv opens a new avenue for low-cost pan-HPV vaccine development.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Hongyang Liu
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Xue Chen
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Zhirong Wang
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Shuo Wang
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Chunfeng Qu
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingzhi Zhang
- Changchun Werersai Biotec Pharmaceutical Co., LTD, Changchun, China
| | - Xuemei Xu
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China.
| |
Collapse
|
44
|
Velmurugan P, Jonnalagadda RR, Sankaranarayanan K, Dhathathreyan A. Does L to D-amino acid substitution trigger helix→sheet conformations in collagen like peptides adsorbed to surfaces? MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2015; 57:249-56. [PMID: 26354261 DOI: 10.1016/j.msec.2015.07.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 06/22/2015] [Accepted: 07/28/2015] [Indexed: 11/18/2022]
Abstract
The present work reports on the structural order, self assembling behaviour and the role in adsorption to hydrophilic or hydrophobic solid surfaces of modified sequence from the triple helical peptide model of the collagenase cleavage site in type I collagen (Uniprot accession number P02452 residues from 935 to 970) using (D)Ala and (D)Ile substitutions as given in the models below: Model-1: GSOGADGPAGAOGTOGPQGIAGQRGVV GLOGQRGER. Model-2: GSOGADGP(D)AGAOGTOGPQGIAGQRGVVGLOGQRGER. Model-3: GSOGADGPAGAOGTOGPQG(D)IAGQRGVVGLOGQRGER. Collagenase is an important enzyme that plays an important role in degrading collagen in wound healing, cancer metastasis and even in embryonic development. However, the mechanism by which this degradation occurs is not completely understood. Our results show that adsorption of the peptides to the solid surfaces, specifically hydrophobic triggers a helix to beta transition with order increasing in peptide models 2 and 3. This restricts the collagenolytic behaviour of collagenase and may find application in design of peptides and peptidomimetics for enzyme-substrate interaction, specifically with reference to collagen and other extra cellular matrix proteins.
Collapse
Affiliation(s)
- Punitha Velmurugan
- Council of Scientific and Industrial Research-Central Leather Research Institute, Chemical Laboratory, Adyar, Chennai 600 020, India; University of Madras, Centre for Advanced Study in Crystallography and Biophysics, Guindy Campus, Chennai 600 025, India
| | - Raghava Rao Jonnalagadda
- Council of Scientific and Industrial Research-Central Leather Research Institute, Chemical Laboratory, Adyar, Chennai 600 020, India.
| | - Kamatchi Sankaranarayanan
- Council of Scientific and Industrial Research-Central Leather Research Institute, Chemical Laboratory, Adyar, Chennai 600 020, India
| | - Aruna Dhathathreyan
- Council of Scientific and Industrial Research-Central Leather Research Institute, Biophysics Laboratory, Adyar, Chennai 600 020, India
| |
Collapse
|
45
|
Cullen LM, Blanco JCG, Morrison TG. Cotton rat immune responses to virus-like particles containing the pre-fusion form of respiratory syncytial virus fusion protein. J Transl Med 2015; 13:350. [PMID: 26541285 PMCID: PMC4636065 DOI: 10.1186/s12967-015-0705-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 10/20/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Virus-like particles (VLPs) based on Newcastle disease virus (NDV) core proteins, M and NP, and containing two chimera proteins, F/F and H/G, composed of the respiratory syncytial virus (RSV) fusion protein (F) and glycoprotein (G) ectodomains fused to the transmembrane and cytoplasmic domains of the NDV F and HN proteins, respectively, stimulate durable, protective anti-RSV neutralizing antibodies in mice. Furthermore, immunization of mice with a VLP containing a F/F chimera protein with modifications previously reported to stabilize the pre-fusion form of the RSV F protein resulted in significantly improved neutralizing antibody titers over VLPs containing the wild type F protein. The goal of this study was to determine if VLPs containing the pre-fusion form of the RSV F protein stimulated protective immune responses in cotton rats, a more RSV permissive animal model than mice. METHODS Cotton rats were immunized intramuscularly with VLPs containing stabilized pre-fusion F/F chimera protein as well as the H/G chimera protein. The anti-RSV F and RSV G antibody responses were determined by ELISA. Neutralizing antibody titers in sera of immunized animals were determined in plaque reduction assays. Protection of the animals from RSV challenge was assessed. The safety of the VLP vaccine was determined by monitoring lung pathology upon RSV challenge of immunized animals. RESULTS The Pre-F/F VLP induced neutralizing titers that were well above minimum levels previously proposed to be required for a successful vaccine and titers significantly higher than those stimulated by RSV infection. In addition, Pre-F/F VLP immunization stimulated higher IgG titers to the soluble pre-fusion F protein than RSV infection. Cotton rats immunized with Pre-F/F VLPs were protected from RSV challenge, and, importantly, the VLP immunization did not result in enhanced respiratory disease upon RSV challenge. CONCLUSIONS VLPs containing the pre-fusion RSV F protein have characteristics required for a safe, effective RSV vaccine.
Collapse
Affiliation(s)
- Lori McGinnes Cullen
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, 01655, USA.
| | | | - Trudy G Morrison
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, 01655, USA.
- Program in Immunology and Microbiology, University of Massachusetts Medical School, Worcester, MA, 01655, USA.
| |
Collapse
|
46
|
Jungst T, Smolan W, Schacht K, Scheibel T, Groll J. Strategies and Molecular Design Criteria for 3D Printable Hydrogels. Chem Rev 2015; 116:1496-539. [PMID: 26492834 DOI: 10.1021/acs.chemrev.5b00303] [Citation(s) in RCA: 436] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Tomasz Jungst
- Department for Functional Materials in Medicine and Dentistry, University of Würzburg , Pleicherwall 2, 97070 Würzburg, Germany
| | - Willi Smolan
- Department for Functional Materials in Medicine and Dentistry, University of Würzburg , Pleicherwall 2, 97070 Würzburg, Germany
| | - Kristin Schacht
- Chair of Biomaterials, Faculty of Engineering Science, University of Bayreuth , Universitätsstrasse 30, 95447 Bayreuth, Germany
| | - Thomas Scheibel
- Chair of Biomaterials, Faculty of Engineering Science, University of Bayreuth , Universitätsstrasse 30, 95447 Bayreuth, Germany
| | - Jürgen Groll
- Department for Functional Materials in Medicine and Dentistry, University of Würzburg , Pleicherwall 2, 97070 Würzburg, Germany
| |
Collapse
|
47
|
White EM, Miranker AD. A solenoid design for assessing determinants of parallel β-sheet registration. Protein Eng Des Sel 2015; 28:577-83. [PMID: 26487712 DOI: 10.1093/protein/gzv053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 09/17/2015] [Indexed: 11/12/2022] Open
Abstract
A novel protein construct is presented that combines a homotrimeric, triple-stranded β-helix as a guest to a homotrimeric foldon unit from bacteriophage T4 fibritin. The β-helical solenoid selected is short (46 residues) and is part of a subdomain of the T4 cell-puncturing device. The resultant design is trimeric and displays greatly enhanced stability over each sub-component alone. The intended goal is a design that will enable evaluation of sequence determinants that promote in-register versus out-of-register parallel β-sheet homotrimerization. Towards that end, the importance of a set of three buried salt-bridges was evaluated by converting them to residues otherwise consistently found throughout the natural solenoid at the same positions. The critical role of the charged residues in the salt-bridges was evident in that their elimination resulted in amyloid-like aggregation.
Collapse
Affiliation(s)
- Ellen M White
- Department of Molecular Biophysics and Biochemistry, Yale University, 260 Whitney Avenue, New Haven, CT 06520-8114, USA
| | - Andrew D Miranker
- Department of Molecular Biophysics and Biochemistry, Yale University, 260 Whitney Avenue, New Haven, CT 06520-8114, USA
| |
Collapse
|
48
|
Körling M, Geyer A. Beyond Natural Limitations: Long-Range Influence of Non-Natural Flexible and Rigid β-Turn Mimetics in a Native β-Hairpin Motif. European J Org Chem 2015. [DOI: 10.1002/ejoc.201500724] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
49
|
Parmar AS, Xu F, Pike DH, Belure SV, Hasan NF, Drzewiecki KE, Shreiber DI, Nanda V. Metal Stabilization of Collagen and de Novo Designed Mimetic Peptides. Biochemistry 2015; 54:4987-97. [PMID: 26225466 PMCID: PMC5335877 DOI: 10.1021/acs.biochem.5b00502] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We explore the design of metal binding sites to modulate triple-helix stability of collagen and collagen-mimetic peptides. Globular proteins commonly utilize metals to connect tertiary structural elements that are well separated in sequence, constraining structure and enhancing stability. It is more challenging to engineer structural metals into fibrous protein scaffolds, which lack the extensive tertiary contacts seen in globular proteins. In the collagen triple helix, the structural adjacency of the carboxy-termini of the three chains makes this region an attractive target for introducing metal binding sites. We engineered His3 sites based on structural modeling constraints into a series of designed homotrimeric and heterotrimeric peptides, assessing the capacity of metal binding to improve stability and in the case of heterotrimers, affect specificity of assembly. Notable enhancements in stability for both homo- and heteromeric systems were observed upon addition of zinc(II) and several other metal ions only when all three histidine ligands were present. Metal binding affinities were consistent with the expected Irving-Williams series for imidazole. Unlike other metals tested, copper(II) also bound to peptides lacking histidine ligands. Acetylation of the peptide N-termini prevented copper binding, indicating proline backbone amide metal-coordination at this site. Copper similarly stabilized animal extracted Type I collagen in a metal-specific fashion, highlighting the potential importance of metal homeostasis within the extracellular matrix.
Collapse
Affiliation(s)
- Avanish S. Parmar
- Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad-500046, Telangana, INDIA
- Center for Advanced Biotechnology and Medicine, Department of Biochemistry and Molecular Biology Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Fei Xu
- Center for Advanced Biotechnology and Medicine, Department of Biochemistry and Molecular Biology Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Douglas H. Pike
- Center for Advanced Biotechnology and Medicine, Department of Biochemistry and Molecular Biology Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Sandeep V. Belure
- Center for Advanced Biotechnology and Medicine, Department of Biochemistry and Molecular Biology Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Nida F. Hasan
- Center for Advanced Biotechnology and Medicine, Department of Biochemistry and Molecular Biology Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Kathryn E. Drzewiecki
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey 08854, USA
| | - David I. Shreiber
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Vikas Nanda
- Center for Advanced Biotechnology and Medicine, Department of Biochemistry and Molecular Biology Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey 08854, USA
| |
Collapse
|
50
|
Sun L, Xiong Y, Bashan A, Zimmerman E, Shulman Daube S, Peleg Y, Albeck S, Unger T, Yonath H, Krupkin M, Matzov D, Yonath A. A Recombinant Collagen-mRNA Platform for Controllable Protein Synthesis. Chembiochem 2015; 16:1415-9. [PMID: 25930950 PMCID: PMC4517095 DOI: 10.1002/cbic.201500205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 04/30/2015] [Indexed: 12/19/2022]
Abstract
We have developed a collagen-mRNA platform for controllable protein production that is intended to be less prone to the problems associated with commonly used mRNA therapy as well as with collagen skin-healing procedures. A collagen mimic was constructed according to a recombinant method and was used as scaffold for translating mRNA chains into proteins. Cysteines were genetically inserted into the collagen chain at positions allowing efficient ribosome translation activity while minimizing mRNA misfolding and degradation. Enhanced green fluorescence protein (eGFP) mRNA bound to collagen was successfully translated by cell-free Escherichia coli ribosomes. This system enabled an accurate control of specific protein synthesis by monitoring expression time and level. Luciferase-mRNA was also translated on collagen scaffold by eukaryotic cell extracts. Thus we have demonstrated the feasibility of controllable protein synthesis on collagen scaffolds by ribosomal machinery.
Collapse
Affiliation(s)
- Liping Sun
- Department of Biomaterials, College of Materials, Xiamen University, 422, Siming South Road, Xiamen 361005 (China)
| | - Yunjing Xiong
- Department of Biomaterials, College of Materials, Xiamen University, 422, Siming South Road, Xiamen 361005 (China)
| | - Anat Bashan
- Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001 (Israel)
| | - Ella Zimmerman
- Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001 (Israel)
| | | | - Yoav Peleg
- Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001 (Israel)
| | - Shira Albeck
- Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001 (Israel)
| | - Tamar Unger
- Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001 (Israel)
| | - Hagith Yonath
- Sheba Medical Center, 1 Sheba Street, Tel Hashomer 52621 (Israel)
- Sackler School of Medicine, Tel Aviv University, 10 Levanon Street, Tel Aviv 69978 (Israel)
| | - Miri Krupkin
- Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001 (Israel)
| | - Donna Matzov
- Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001 (Israel)
| | - Ada Yonath
- Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001 (Israel).
| |
Collapse
|