1
|
Rončević T, Gerdol M, Pacor S, Cvitanović A, Begić A, Weber I, Krce L, Caporale A, Mardirossian M, Tossi A, Zoranić L. Antimicrobial Peptide with a Bent Helix Motif Identified in Parasitic Flatworm Mesocestoides corti. Int J Mol Sci 2024; 25:11690. [PMID: 39519242 PMCID: PMC11546468 DOI: 10.3390/ijms252111690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 10/25/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
The urgent need for antibiotic alternatives has driven the search for antimicrobial peptides (AMPs) from many different sources, yet parasite-derived AMPs remain underexplored. In this study, three novel potential AMP precursors (mesco-1, -2 and -3) were identified in the parasitic flatworm Mesocestoides corti, via a genome-wide mining approach, and the most promising one, mesco-2, was synthesized and comprehensively characterized. It showed potent broad-spectrum antibacterial activity at submicromolar range against E. coli and K. pneumoniae and low micromolar activity against A. baumannii, P. aeruginosa and S. aureus. Mechanistic studies indicated a membrane-related mechanism of action, and circular dichroism spectroscopy confirmed that mesco-2 is unstructured in water but forms stable helical structures on contact with anionic model membranes, indicating strong interactions and helix stacking. It is, however, unaffected by neutral membranes, suggesting selective antimicrobial activity. Structure prediction combined with molecular dynamics simulations suggested that mesco-2 adopts an unusual bent helix conformation with the N-terminal sequence, when bound to anionic membranes, driven by a central GRGIGRG motif. This study highlights mesco-2 as a promising antibacterial agent and emphasizes the importance of structural motifs in modulating AMP function.
Collapse
Affiliation(s)
- Tomislav Rončević
- Department of Biology, Faculty of Science, University of Split, 21000 Split, Croatia;
| | - Marco Gerdol
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (M.G.); (S.P.); (M.M.); (A.T.)
| | - Sabrina Pacor
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (M.G.); (S.P.); (M.M.); (A.T.)
| | - Ana Cvitanović
- Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia;
| | - Anamarija Begić
- Department of Physics, Faculty of Science, University of Split, 21000 Split, Croatia; (A.B.); (I.W.); (L.K.)
| | - Ivana Weber
- Department of Physics, Faculty of Science, University of Split, 21000 Split, Croatia; (A.B.); (I.W.); (L.K.)
| | - Lucija Krce
- Department of Physics, Faculty of Science, University of Split, 21000 Split, Croatia; (A.B.); (I.W.); (L.K.)
| | - Andrea Caporale
- Institute of Crystallography, CNR, Basovizza, 34149 Trieste, Italy;
| | - Mario Mardirossian
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (M.G.); (S.P.); (M.M.); (A.T.)
| | - Alessandro Tossi
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (M.G.); (S.P.); (M.M.); (A.T.)
| | - Larisa Zoranić
- Department of Physics, Faculty of Science, University of Split, 21000 Split, Croatia; (A.B.); (I.W.); (L.K.)
| |
Collapse
|
2
|
Xu L, Fan X, He Y, Xia X, Zhang J. Design, Synthesis, and Biological Evaluation of Lysine-Stapled Peptide Inhibitors of p53-MDM2/MDMX Interactions with Potent Antitumor Activity In Vivo. J Med Chem 2024; 67:17893-17904. [PMID: 39300610 DOI: 10.1021/acs.jmedchem.4c01939] [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: 09/22/2024]
Abstract
We introduce novel lysine-stapled peptide inhibitors targeting p53-MDM2/MDMX interactions. Leveraging the model peptides pDI (LTFEHYWAQLTS) and PMI-M3 (LTFLEYWAQLMQ) as starting points, a series of lysine-stapled analogues were designed and synthesized. Through in vitro cell assay screening, two lead compounds, SPDI-48-T1 and SPMI-48-T3, were identified for their excellent antiproliferation activity. Fluorescence polarization assays revealed that both compounds exhibited strong binding affinities against MDM2 and MDMX, ascertained by Kd values within the low micromolar spectrum. Further characterization of SPDI-48-T1 and SPMI-48-T3 demonstrated that SPDI-48-T1 possessed superior cell permeability and serum stability. Notably, SPDI-48-T1 displayed a dose-dependent suppression of tumor growth in an HCT116 xenograft mouse model. Our findings indicate that SPDI-48-T1 holds promise as a lead compound for further development as an anticancer agent by modulating p53-MDM2/MDMX interactions. Additionally, this study also proved that the lysine stapling strategy may serve as a robust approach for generating peptide ligands targeting other protein-protein interactions.
Collapse
Affiliation(s)
- Lei Xu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, People's Republic of China
| | - Xin Fan
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, People's Republic of China
| | - Yi He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, People's Republic of China
| | - Xuefeng Xia
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, People's Republic of China
| | - Jinqiang Zhang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, People's Republic of China
- Chongqing University Industrial Technology Research Institute, Chongqing 401329, People's Republic of China
| |
Collapse
|
3
|
Li K, Liu L. Computational design and experimental confirmation of a disulfide-stapled YAP helix α1-trap derived from TEAD4 helical hairpin to selectively capture YAP α1-helix with potent antitumor activity. J Comput Aided Mol Des 2024; 38:31. [PMID: 39177727 DOI: 10.1007/s10822-024-00572-2] [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: 04/07/2024] [Accepted: 08/14/2024] [Indexed: 08/24/2024]
Abstract
Human Hippo signaling pathway is an evolutionarily conserved regulator network that controls organ development and has been implicated in various cancers. Transcriptional enhanced associate domain-4 (TEAD4) is the final nuclear effector of Hippo pathway, which is activated by Yes-associated protein (YAP) through binding to two separated YAP regions of α1-helix and Ω-loop. Previous efforts have all been addressed on deriving peptide inhibitors from the YAP to target TEAD4. Instead, we herein attempted to rationally design a so-called 'YAP helixα1-trap' based on the TEAD4 to target YAP by using dynamics simulation and energetics analysis as well as experimental assays at molecular and cellular levels. The trap represents a native double-stranded helical hairpin covering a specific YAP-binding site on TEAD4 surface, which is expected to form a three-helix bundle with the α1-helical region of YAP, thus competitively disrupting TEAD4-YAP interaction. The hairpin was further stapled by a disulfide bridge across its two helical arms. Circular dichroism characterized that the stapling can effectively constrain the trap into a native-like structured conformation in free state, thus largely minimizing the entropy penalty upon its binding to YAP. Affinity assays revealed that the stapling can considerably improve the trap binding potency to YAP α1-helix by up to 8.5-fold at molecular level, which also exhibited a good tumor-suppressing effect at cellular level if fused with TAT cell permeation sequence. In this respect, it is considered that the YAP helixα1-trap-mediated blockade of Hippo pathway may be a new and promising therapeutic strategy against cancers.
Collapse
Affiliation(s)
- Kaipeng Li
- School of Chemistry and Chemical Engineering, Jinggangshan University, No. 28, Xueyuan Road, Ji'an, 343009, China
| | - Lijun Liu
- School of Chemistry and Chemical Engineering, Jinggangshan University, No. 28, Xueyuan Road, Ji'an, 343009, China.
| |
Collapse
|
4
|
Bhatt MR, Zondlo NJ. Electronic Control of Polyproline II Helix Stability via the Identity of Acyl Capping Groups: the Pivaloyl Group Particularly Promotes PPII. Chemistry 2024; 30:e202401454. [PMID: 38661017 DOI: 10.1002/chem.202401454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 04/26/2024]
Abstract
The type II polyproline helix (PPII) is a fundamental secondary structure of proteins, important in globular proteins, in intrinsically disordered proteins, and at protein-protein interfaces. PPII is stabilized in part by n→π* interactions between consecutive carbonyls, via electron delocalization between an electron-donor carbonyl lone pair (n) and an electron-acceptor carbonyl (π*) on the subsequent residue. We previously demonstrated that changes to the electronic properties of the acyl donor can predictably modulate the strength of n→π* interactions, with data from model compounds, in solution in chloroform, in the solid state, and computationally. Herein, we examined whether the electronic properties of acyl capping groups could modulate the stability of PPII in peptides in water. In X-PPGY-NH2 peptides (X=10 acyl capping groups), the effect of acyl group identity on PPII was quantified by circular dichroism and NMR spectroscopy. Electron-rich acyl groups promoted PPII relative to the standard acetyl (Ac-) group, with the pivaloyl and iso-butyryl groups most significantly increasing PPII. In contrast, acyl derivatives with electron-withdrawing substituents and the formyl group relatively disfavored PPII. Similar results, though lesser in magnitude, were also observed in X-APPGY-NH2 peptides, indicating that the capping group can impact PPII conformation at both proline and non-proline residues. The pivaloyl group was particularly favorable in promoting PPII. The effects of acyl capping groups were further analyzed in X-DfpPGY-NH2 and X-ADfpPGY-NH2 peptides, Dfp=4,4-difluoroproline. Data on these peptides indicated that acyl groups induced order Piv- > Ac- > For-. These results suggest that greater consideration should be given to the identity of acyl capping groups in inducing structure in peptides.
Collapse
Affiliation(s)
- Megh R Bhatt
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, 19716, United States
| | - Neal J Zondlo
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, 19716, United States
| |
Collapse
|
5
|
Quigua-Orozco RM, Andrade IEP, Oshiro KGN, Rezende SB, Santos ADO, Pereira JAL, da Silva VG, Buccini DF, Porto WF, Macedo MLR, Cardoso MH, Franco OL. In silico optimization of analogs derived pro-adrenomedullin peptide to evaluate antimicrobial potential. Chem Biol Drug Des 2024; 104:e14588. [PMID: 39048531 DOI: 10.1111/cbdd.14588] [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: 01/30/2024] [Revised: 06/04/2024] [Accepted: 07/09/2024] [Indexed: 07/27/2024]
Abstract
Diverse computational approaches have been widely used to assist in designing antimicrobial peptides with enhanced activities. This tactic has also been used to address the need for new treatment alternatives to combat resistant bacterial infections. Herein, we have designed eight variants from a natural peptide, pro-adrenomedullin N-terminal 20 peptide (PAMP), using an in silico pattern insertion approach, the Joker algorithm. All the variants show an α-helical conformation, but with differences in the helix percentages according to circular dichroism (CD) results. We found that the C-terminal portion of PAMP may be relevant for its antimicrobial activities, as revealed by the molecular dynamics, CD, and antibacterial results. The analogs showed variable antibacterial potential, but most were not cytotoxic. Nevertheless, PAMP2 exhibited the most potent activities against human and animal-isolated bacteria, showing cytotoxicity only at a substantially higher concentration than its minimal inhibitory concentration (MIC). Our results suggest that the enhanced activity in the profile of PAMP2 may be related to their particular physicochemical properties, along with the adoption of an amphipathic α-helical arrangement with the conserved C-terminus portion. Finally, the peptides designed in this study can constitute scaffolds for the design of improved sequences.
Collapse
Affiliation(s)
- Raquel M Quigua-Orozco
- S-Inova Biotech, Programa de Pós-Graduação Em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul, Brazil
| | - Isadora E P Andrade
- Programa de Pós-Graduação Em Patologia Molecular, Faculdade de Medicina, Universidade de Brasília, Brasília, Distrito Federal, Brazil
| | - Karen G N Oshiro
- S-Inova Biotech, Programa de Pós-Graduação Em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul, Brazil
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação Em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Distrito Federal, Brazil
| | - Samilla B Rezende
- S-Inova Biotech, Programa de Pós-Graduação Em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul, Brazil
| | - Alexandre Duarte O Santos
- S-Inova Biotech, Programa de Pós-Graduação Em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul, Brazil
| | - Julia A L Pereira
- S-Inova Biotech, Programa de Pós-Graduação Em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul, Brazil
| | - Viviane G da Silva
- S-Inova Biotech, Programa de Pós-Graduação Em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul, Brazil
| | - Danieli F Buccini
- S-Inova Biotech, Programa de Pós-Graduação Em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul, Brazil
| | - William F Porto
- Programa de Pós-Graduação Em Patologia Molecular, Faculdade de Medicina, Universidade de Brasília, Brasília, Distrito Federal, Brazil
| | - Maria L R Macedo
- Laboratório de Purificação de Proteínas e Suas Funções Biológicas, Universidade Federal de Mato Grosso Do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Marlon H Cardoso
- S-Inova Biotech, Programa de Pós-Graduação Em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul, Brazil
- Programa de Pós-Graduação Em Patologia Molecular, Faculdade de Medicina, Universidade de Brasília, Brasília, Distrito Federal, Brazil
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação Em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Distrito Federal, Brazil
- Laboratório de Purificação de Proteínas e Suas Funções Biológicas, Universidade Federal de Mato Grosso Do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Octávio L Franco
- S-Inova Biotech, Programa de Pós-Graduação Em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul, Brazil
- Programa de Pós-Graduação Em Patologia Molecular, Faculdade de Medicina, Universidade de Brasília, Brasília, Distrito Federal, Brazil
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação Em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Distrito Federal, Brazil
| |
Collapse
|
6
|
Vilas Boas LCP, Buccini DF, Berlanda RLA, Santos BDPO, Maximiano MR, Lião LM, Gonçalves S, Santos NC, Franco OL. Antiviral Activities of Mastoparan-L-Derived Peptides against Human Alphaherpesvirus 1. Viruses 2024; 16:948. [PMID: 38932240 PMCID: PMC11209138 DOI: 10.3390/v16060948] [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: 03/09/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Human alphaherpesvirus 1 (HSV-1) is a significantly widespread viral pathogen causing recurrent infections that are currently incurable despite available treatment protocols. Studies have highlighted the potential of antimicrobial peptides sourced from Vespula lewisii venom, particularly those belonging to the mastoparan family, as effective against HSV-1. This study aimed to demonstrate the antiviral properties of mastoparans, including mastoparan-L [I5, R8], mastoparan-MO, and [I5, R8] mastoparan, against HSV-1. Initially, Vero cell viability was assessed in the presence of these peptides, followed by the determination of antiviral activity, mechanism of action, and dose-response curves through plaque assays. Structural analyses via circular dichroism and nuclear magnetic resonance were conducted, along with evaluating membrane fluidity changes induced by [I5, R8] mastoparan using fluorescence-labeled lipid vesicles. Cytotoxic assays revealed high cell viability (>80%) at concentrations of 200 µg/mL for mastoparan-L and mastoparan-MO and 50 µg/mL for [I5, R8] mastoparan. Mastoparan-MO and [I5, R8] mastoparan exhibited over 80% HSV-1 inhibition, with up to 99% viral replication inhibition, particularly in the early infection stages. Structural analysis indicated an α-helical structure for [I5, R8] mastoparan, suggesting effective viral particle disruption before cell attachment. Mastoparans present promising prospects for HSV-1 infection control, although further investigation into their mechanisms is warranted.
Collapse
Affiliation(s)
- Liana Costa Pereira Vilas Boas
- Pós-Graduação em Patologia Molecular, Campus Darcy Ribeiro, Universidade de Brasília, Brasília 70910-900, DF, Brazil
- Centro de Análises Bioquímicas e Proteômicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília 70790-760, DF, Brazil
| | - Danieli Fernanda Buccini
- Centro de Análises Bioquímicas e Proteômicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília 70790-760, DF, Brazil
- Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande 79117-900, MS, Brazil
| | - Rhayfa Lorrayne Araújo Berlanda
- Pós-Graduação em Patologia Molecular, Campus Darcy Ribeiro, Universidade de Brasília, Brasília 70910-900, DF, Brazil
- Centro de Análises Bioquímicas e Proteômicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília 70790-760, DF, Brazil
| | - Bruno de Paula Oliveira Santos
- Laboratório de Ressonância Magnética Nuclear, Instituto de Química, Universidade Federal de Goiás, Goiânia 74690-900, GO, Brazil
| | - Mariana Rocha Maximiano
- Centro de Análises Bioquímicas e Proteômicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília 70790-760, DF, Brazil
- Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande 79117-900, MS, Brazil
| | - Luciano Morais Lião
- Laboratório de Ressonância Magnética Nuclear, Instituto de Química, Universidade Federal de Goiás, Goiânia 74690-900, GO, Brazil
| | - Sónia Gonçalves
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal (N.C.S.)
| | - Nuno C. Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal (N.C.S.)
| | - Octávio Luiz Franco
- Pós-Graduação em Patologia Molecular, Campus Darcy Ribeiro, Universidade de Brasília, Brasília 70910-900, DF, Brazil
- Centro de Análises Bioquímicas e Proteômicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília 70790-760, DF, Brazil
- Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande 79117-900, MS, Brazil
| |
Collapse
|
7
|
Fischer N, Tóth A, Jancsó A, Thulstrup P, Diness F. Inducing α-Helicity in Peptides by Silver Coordination to Cysteine. Chemistry 2024; 30:e202304064. [PMID: 38456607 DOI: 10.1002/chem.202304064] [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: 01/18/2024] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/09/2024]
Abstract
Short peptide sequences consisting of two cysteine residues separated by three other amino acids display complete change from random coil to α-helical secondary structure in response to addition of Ag+ ions. The folded CXXXC/Ag+ complex involves formation of multinuclear Ag+ species and is stable in a wide pH range from below 3 to above 8. The complex is stable through reversed-phase HPLC separation as well as towards a physiological level of chloride ions, based on far-UV circular dichroism spectroscopy. In electrospray MS under acidic conditions a peptide dimer with four Ag+ ions bound was observed, and modelling based on potentiometric experiments supported this to be the dominating complex at neutral pH together with a peptide dimer with 3 Ag+ and one proton at lower pH. The complex was demonstrated to work as a N-terminal nucleation site for inducing α-helicity into longer peptides. This type of silver-mediated peptide assembly and folding may be of more general use for stabilizing not only peptide folding but also for controlling oligomerization even under acidic conditions.
Collapse
Affiliation(s)
- Niklas Fischer
- Department of Science and Environment, Roskilde University, Universitetsvej 1, 4000, Roskilde, Denmark
- Department of Chemistry, University of Copenhagen, Universitetsparken 5, 2100, København Ø, Denmark
| | - Annamária Tóth
- Department of Molecular and Analytical Chemistry, University of Szeged, Dómtér 7-8, H-6720, Szeged, Hungary
| | - Attila Jancsó
- Department of Molecular and Analytical Chemistry, University of Szeged, Dómtér 7-8, H-6720, Szeged, Hungary
| | - Peter Thulstrup
- Department of Chemistry, University of Copenhagen, Universitetsparken 5, 2100, København Ø, Denmark
| | - Frederik Diness
- Department of Science and Environment, Roskilde University, Universitetsvej 1, 4000, Roskilde, Denmark
- Department of Chemistry, University of Copenhagen, Universitetsparken 5, 2100, København Ø, Denmark
| |
Collapse
|
8
|
Cao HW, Chen YS, Li JZ, Chen HW, Li LY, Li ZK, Wang MQ. Development of D-π-A organic dyes for discriminating HSA from BSA and study on dye-HSA interaction. Bioorg Chem 2024; 147:107360. [PMID: 38604019 DOI: 10.1016/j.bioorg.2024.107360] [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: 02/18/2024] [Revised: 03/21/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
HSA (human serum albumin), a most abundant protein in blood serum, plays a key role in maintaining human health. Abnormal HSA level is correlated with many diseases, and thus has been used as an essential biomarker for therapeutic monitoring and biomedical diagnosis. Development of small-molecule fluorescent probes allowing the selective and sensitive recognition of HSA in in vitro and in vivo is of fundamental importance in basic biological research as well as medical diagnosis. Herein, we reported a series of new synthesized fluorescent dyes containing D-π-A constitution, which exhibited different optical properties in solution and solid state. Among them, dye M-H-SO3 with a hydrophilic sulfonate group at electron-acceptor part displayed selectivity for discrimination of HSA from BSA and other enzymes. Upon binding of dye M-H-SO3 with HSA, a significant fluorescence enhancement with a turn-on ratio about 96-fold was triggered. The detection limit was estimated to be ∼ 40 nM. Studies on the interaction mechanism revealed that dye M-H-SO3 could bind to site III of HSA with a 1:1 binding stoichiometry. Furthermore, dye M-H-SO3 has been applied to determine HSA in real urine samples with good recoveries, which provided a useful method for HSA analysis in biological fluids.
Collapse
Affiliation(s)
- Hao-Wen Cao
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Yan-Song Chen
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Jing-Zhi Li
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Hai-Wen Chen
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Lu-Yu Li
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Ze-Kai Li
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Ming-Qi Wang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China.
| |
Collapse
|
9
|
Orozco RMQ, Oshiro KGN, Pinto IB, Buccini DF, Almeida CV, Marin VN, de Souza CM, Macedo MLR, Cardoso MH, Franco OL. Employment of mastoparan-like peptides to prevent Staphylococcus aureus associated with bovine mastitis. J Bacteriol 2024; 206:e0007124. [PMID: 38629875 PMCID: PMC11112992 DOI: 10.1128/jb.00071-24] [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: 03/05/2024] [Accepted: 03/25/2024] [Indexed: 05/24/2024] Open
Abstract
Bovine mastitis is a frequent infection in lactating cattle, causing great economic losses. Staphylococcus aureus represents the main etiological agent, which causes recurrent and persistent intramammary infections because conventional antibiotics are ineffective against it. Mastoparan-like peptides are multifunctional molecules with broad antimicrobial potential, constituting an attractive alternative. Nevertheless, their toxicity to host cells has hindered their therapeutic application. Previously, our group engineered three mastoparan-L analogs, namely mastoparan-MO, mastoparan-R1, and [I5, R8] MP, to improve cell selectivity and potential. Here, we were interested in comparing the antibacterial efficacy of mastoparan-L and its analogs against bovine mastitis isolates of S. aureus strains, making a correlation with the physicochemical properties and structural arrangement changes promoted by the sequence modifications. As a result, the analog's hemolytic and/or antimicrobial activity was balanced. All the peptides displayed α-helical folding in hydrophobic and membrane-mimetic environments, as determined by circular dichroism. The peptide [I5, R8] MP stood out for its enhanced selectivity and antibacterial features related to mastoparan-L and the other derivatives. Biophysical approaches revealed that [I5, R8] MP rapidly depolarizes the bacterial membrane of S. aureus, causing cell death by subsequent membrane disruption. Our results demonstrated that the [I5, R8] MP peptide could be a starting point for the development of peptide-based drugs for the treatment of bovine mastitis, with the advantage of no residue in milk, which would help reduce the use of classical antibiotics.IMPORTANCEStaphylococcus aureus is a leading cause of mastitis, the world's most important dairy cattle disease. The multidrug resistance and zoonotic potential of S. aureus, besides the likelihood of antibiotic residues in milk, are of critical concern to public and animal health. Antimicrobial peptides offer a novel antimicrobial strategy. Here, we demonstrate that [I5, R8] MP is a potent and selective peptide, which acts on S. aureus by targeting the bacterial membrane. Therefore, understanding the physicochemical determinants and the modes of action of this class of antimicrobials opens novel prospects for peptide development with enhanced activities in the bovine mastitis context.
Collapse
Affiliation(s)
- Raquel M. Q. Orozco
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
| | - Karen G. N. Oshiro
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
- Programa de Pós-Graduação em Patologia Molecular, Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil
| | - Ingrid B. Pinto
- Laboratório de Síntese e Transformações de Moléculas Orgânicas, Instituo de Química, Universidade Federal de Mato Grosso do Sul, Campo Grande, Brazil
| | - Danieli F. Buccini
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
| | - Claudiane V. Almeida
- Laboratório de Purificação de Proteínas e suas Funções Biológicas, Universidade Federal de Mato Grosso do Sul, Campo Grande, Brazil
| | - Valentina Nieto Marin
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
| | - Camila Maurmann de Souza
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brasília, Brazil
| | - Maria L. R. Macedo
- Laboratório de Purificação de Proteínas e suas Funções Biológicas, Universidade Federal de Mato Grosso do Sul, Campo Grande, Brazil
| | - Marlon H. Cardoso
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
- Programa de Pós-Graduação em Patologia Molecular, Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil
- Laboratório de Purificação de Proteínas e suas Funções Biológicas, Universidade Federal de Mato Grosso do Sul, Campo Grande, Brazil
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brasília, Brazil
| | - Octávio L. Franco
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
- Programa de Pós-Graduação em Patologia Molecular, Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brasília, Brazil
| |
Collapse
|
10
|
Bhatt MR, Ganguly HK, Zondlo NJ. Acyl Capping Group Identity Effects on α-Helicity: On the Importance of Amide·Water Hydrogen Bonds to α-Helix Stability. Biochemistry 2024; 63:1118-1130. [PMID: 38623827 DOI: 10.1021/acs.biochem.3c00646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
Acyl capping groups stabilize α-helices relative to free N-termini by providing one additional C═Oi···Hi+4-N hydrogen bond. The electronic properties of acyl capping groups might also directly modulate α-helix stability: electron-rich N-terminal acyl groups could stabilize the α-helix by strengthening both i/i + 4 hydrogen bonds and i/i + 1 n → π* interactions. This hypothesis was tested in peptides X-AKAAAAKAAAAKAAGY-NH2, where X = different acyl groups. Surprisingly, the most electron-rich acyl groups (pivaloyl and iso-butyryl) strongly destabilized the α-helix. Moreover, the formyl group induced nearly identical α-helicity to that of the acetyl group, despite being a weaker electron donor for hydrogen bonds and for n → π* interactions. Other acyl groups exhibited intermediate α-helicity. These results indicate that the electronic properties of the acyl carbonyl do not directly determine the α-helicity in peptides in water. In order to understand these effects, DFT calculations were conducted on α-helical peptides. Using implicit solvation, α-helix stability correlated with acyl group electronics, with the pivaloyl group exhibiting closer hydrogen bonds and n → π* interactions, in contrast to the experimental results. However, DFT and MD calculations with explicit water solvation revealed that hydrogen bonding to water was impacted by the sterics of the acyl capping group. Formyl capping groups exhibited the closest water-amide hydrogen bonds, while pivaloyl groups exhibited the longest. In α-helices in the PDB, the highest frequency of close amide-water hydrogen bonds is observed when the N-cap residue is Gly. The combination of experimental and computational results indicates that solvation (hydrogen bonding of water) to the N-terminal amide groups is a central determinant of α-helix stability.
Collapse
Affiliation(s)
- Megh R Bhatt
- Department of Chemistry and Biochemistry, University of Delaware Newark, Delaware 19716, United States
| | - Himal K Ganguly
- Department of Chemistry and Biochemistry, University of Delaware Newark, Delaware 19716, United States
| | - Neal J Zondlo
- Department of Chemistry and Biochemistry, University of Delaware Newark, Delaware 19716, United States
| |
Collapse
|
11
|
Maria Medeiros Theóphilo Galvão A, Lamy Rasera M, de Figueiredo Furtado G, Grossi Bovi Karatay G, M Tavares G, Dupas Hubinger M. Lentil protein isolate (Lens culinaris) subjected to ultrasound treatment combined or not with heat-treatment: structural characterization and ability to stabilize high internal phase emulsions. Food Res Int 2024; 183:114212. [PMID: 38760140 DOI: 10.1016/j.foodres.2024.114212] [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: 11/04/2023] [Revised: 02/20/2024] [Accepted: 03/10/2024] [Indexed: 05/19/2024]
Abstract
This study evaluated the effect of ultrasound treatment combined or not with heat treatment applied to lentil protein isolate (LPI) aiming to enhance its ability to stabilize high internal phase emulsions (HIPE). LPI dispersion (2%, w/w) was ultrasound-treated at 60% (UA) and 70% (UB) amplitude for 7 min; these samples were subjected to and then heat treatments at 70 °C (UAT70 and UBT70, respectively) or 80 °C (UAT80 and UBT80, respectively) for 20 min. HIPEs were produced with 25% untreated and treated LPI dispersions and 75% soybean oil using a rotor-stator (15,500 rpm/1 min). The LPI dispersions were evaluated for particle size, solubility, differential scanning calorimetry, electrophoresis, secondary structure estimation (circular dichroism and FT-IR), intrinsic fluorescence, surface hydrophobicity, and free sulfhydryl groups content. The HIPEs were evaluated for droplet size, morphology, rheology, centrifugal stability, and the Turbiscan test. Ultrasound treatment decreased LPI dispersions' particle size (∼80%) and increased solubility (∼90%). Intrinsic fluorescence and surface hydrophobicity confirmed LPI modification due to the exposure to hydrophobic patches. The combination of ultrasound and heat treatments resulted in a reduction in the free sulfhydryl group content of LPI. HIPEs produced with ultrasound-heat-treated LPI had a lower droplet size distribution mode, greater oil retention values in the HIPE structure (> 98%), lower Turbiscan stability index (< 2), and a firmer and more homogeneous appearance compared to HIPE produced with untreated LPI, indicating higher stability for the HIPEs stabilized by treated LPI. Therefore, combining ultrasound and heat treatments could be an effective method for the functional modification of lentil proteins, allowing their application as HIPE emulsifiers.
Collapse
Affiliation(s)
- Andrêssa Maria Medeiros Theóphilo Galvão
- Departamento de Engenharia e Tecnologia de Alimentos, Faculdade de Engenharia de Alimentos, Universidade Estadual de Campinas (UNICAMP), Monteiro Lobato, 80, 13083-862 Campinas, SP, Brazil.
| | - Mariana Lamy Rasera
- Departamento de Ciência de Alimentos e Nutrição, Faculdade de Engenharia de Alimentos, Universidade Estadual de Campinas (UNICAMP), Monteiro Lobato, 80, 13083-862 Campinas, SP, Brazil
| | - Guilherme de Figueiredo Furtado
- Centro de Ciências da Natureza, Universidade Federal de São Carlos, Rod. Lauri Simões de Barros, km 12 - SP 189, Buri, SP 18290-000, Brazil
| | - Graziele Grossi Bovi Karatay
- Departamento de Engenharia e Tecnologia de Alimentos, Faculdade de Engenharia de Alimentos, Universidade Estadual de Campinas (UNICAMP), Monteiro Lobato, 80, 13083-862 Campinas, SP, Brazil
| | - Guilherme M Tavares
- Departamento de Ciência de Alimentos e Nutrição, Faculdade de Engenharia de Alimentos, Universidade Estadual de Campinas (UNICAMP), Monteiro Lobato, 80, 13083-862 Campinas, SP, Brazil
| | - Míriam Dupas Hubinger
- Departamento de Engenharia e Tecnologia de Alimentos, Faculdade de Engenharia de Alimentos, Universidade Estadual de Campinas (UNICAMP), Monteiro Lobato, 80, 13083-862 Campinas, SP, Brazil
| |
Collapse
|
12
|
Zavrtanik U, Lah J, Hadži S. Estimation of Peptide Helicity from Circular Dichroism Using the Ensemble Model. J Phys Chem B 2024; 128:2652-2663. [PMID: 38470351 PMCID: PMC10961730 DOI: 10.1021/acs.jpcb.3c07511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/13/2024]
Abstract
An established method for the quantitation of the helix content in peptides using circular dichroism (CD) relies on the linear spectroscopic model. This model assumes an average value of the helix-length correction for all peptide conformers, irrespective of the length of the helical segment. Here we assess the validity of this approximation and introduce a more physically realistic ensemble-based analysis of the CD signal in which the length correction is assigned specifically to each ensemble conformer. We demonstrate that the linear model underestimates peptide helicity, with the difference depending on the ensemble composition. We developed a computer program that implements the ensemble model to estimate the peptide helicity. Using this model and the CD data set covering a broad range of helicities, we recalibrate CD baseline parameters and redetermine helix-coil parameters for the alanine-rich peptide. We show that the ensemble model leverages small differences in signal between conformers to extract more information from the experimental data, enabling the determination of several poorly defined quantities, such as the nucleation constant and heat capacity change associated with helix folding. Overall, the presented ensemble-based treatment of the CD signal, together with the recalibrated values of the spectroscopic baseline parameters, provides a coherent framework for the analysis of the peptide helix content.
Collapse
Affiliation(s)
- Uroš Zavrtanik
- Department of Physical Chemistry,
Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Jurij Lah
- Department of Physical Chemistry,
Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - San Hadži
- Department of Physical Chemistry,
Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
13
|
Hughes RG, Zhao S, Oas TG, Schmidler SC. Efficient enumeration and visualization of helix-coil ensembles. Biophys J 2024; 123:317-333. [PMID: 38158653 PMCID: PMC10870177 DOI: 10.1016/j.bpj.2023.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/26/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024] Open
Abstract
Helix-coil models are routinely used to interpret circular dichroism data of helical peptides or predict the helicity of naturally-occurring and designed polypeptides. However, a helix-coil model contains significantly more information than mean helicity alone, as it defines the entire ensemble-the equilibrium population of every possible helix-coil configuration-for a given sequence. Many desirable quantities of this ensemble are either not obtained as ensemble averages or are not available using standard helicity-averaging calculations. Enumeration of the entire ensemble can allow calculation of a wider set of ensemble properties, but the exponential size of the configuration space typically renders this intractable. We present an algorithm that efficiently approximates the helix-coil ensemble to arbitrary accuracy by sequentially generating a list of the M highest populated configurations in descending order of population. Truncating this list of (configuration, population) pairs at a desired accuracy provides an approximating sub-ensemble. We demonstrate several uses of this approach for providing insight into helix-coil ensembles and folding mechanisms, including landscape visualization.
Collapse
Affiliation(s)
- Roy G Hughes
- Department of Biochemistry, Duke University, Durham, North Carolina
| | - Shiwen Zhao
- Program in Computational Biology and Bioinformatics, Duke University, Durham, North Carolina
| | - Terrence G Oas
- Department of Biochemistry, Duke University, Durham, North Carolina
| | - Scott C Schmidler
- Program in Computational Biology and Bioinformatics, Duke University, Durham, North Carolina; Department of Statistical Science, Duke University, Durham, North Carolina.
| |
Collapse
|
14
|
Siddiqui GA, Naeem A. Bioflavonoids ameliorate crowding induced hemoglobin aggregation: a spectroscopic and molecular docking approach. J Biomol Struct Dyn 2023; 41:10315-10325. [PMID: 36519442 DOI: 10.1080/07391102.2022.2154270] [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: 09/23/2022] [Accepted: 11/26/2022] [Indexed: 12/23/2022]
Abstract
The cellular environment is densely crowded, confining biomacromolecules including proteins to less available space. This macromolecular confinement may affect the physiological conformation of proteins in long-term processes like ageing. Changes in physiological protein structure can lead to protein conformational disorders including neurodegeneration. An intervention approach using food and plant derived bioflavonoids offered a way to find a treatment for these enervating pathological conditions as there is no remedy available. The bioflavonoids NAR (naringenin), 7HD (7 hydroxyflavanone) and CHR (chrysin) were tested for their ability to protect Hb (hemoglobin) against crowding-induced aggregation. Morphological and secondary structural transitions were studied using microscopic and circular dichroism experiments, respectively. The kinetic study was carried out using the relative thioflavin T assay. Molecular docking, AmylPred2, admetSAR and FRET were applied to understand the binding parameters of bioflavonoids with Hb and their drug likeliness. Isolated human lymphocytes were used as a cellular system to study the toxic effects of Hb aggregates. Redox perturbation and cytotoxicity were evaluated by DCFH-DA and MTT assays, respectively. This study suggests that bioflavonoids bind to Hb in the vicinity of aggregation prone amino acid sequences. Binding of the bioflavonoids stabilizes the Hb against crowding-induced structural alterations. Therefore, this study signifies the potential of bioflavonoids for future treatment of many proteopathies including neurodegeneration.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Gufran Ahmed Siddiqui
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Aabgeena Naeem
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
15
|
Johnson TS, Bourdine AA, Deber CM. Hydrophobic moment drives penetration of bacterial membranes by transmembrane peptides. J Biol Chem 2023; 299:105266. [PMID: 37734555 PMCID: PMC10585379 DOI: 10.1016/j.jbc.2023.105266] [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: 07/30/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023] Open
Abstract
With antimicrobial resistance (AMR) remaining a persistent and growing threat to human health worldwide, membrane-active peptides are gaining traction as an alternative strategy to overcome the issue. Membrane-embedded multi-drug resistant (MDR) efflux pumps are a prime target for membrane-active peptides, as they are a well-established contributor to clinically relevant AMR infections. Here, we describe a series of transmembrane peptides (TMs) to target the oligomerization motif of the AcrB component of the AcrAB-TolC MDR efflux pump from Escherichia coli. These peptides contain an N-terminal acetyl-A-(Sar)3 (sarcosine; N-methylglycine) tag and a C-terminal lysine tag-a design strategy our lab has utilized to improve the solubility and specificity of targeting for TMs previously. While these peptides have proven useful in preventing AcrB-mediated substrate efflux, the mechanisms by which these peptides associate with and penetrate the bacterial membrane remained unknown. In this study, we have shown peptide hydrophobic moment (μH)-the measure of concentrated hydrophobicity on one face of a lipopathic α-helix-drives bacterial membrane permeabilization and depolarization, likely through lateral-phase separation of negatively-charged POPG lipids and the disruption of lipid packing. Our results show peptide μH is an important consideration when designing membrane-active peptides and may be the determining factor in whether a TM will function in a permeabilizing or non-permeabilizing manner when embedded in the bacterial membrane.
Collapse
Affiliation(s)
- Tyler S Johnson
- Program in Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Aleksandra A Bourdine
- Program in Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Charles M Deber
- Program in Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
16
|
Potnis CS, Grapperhaus CA, Gupta G. Investigating BioCaRGOS, a Sol-Gel Matrix for the Stability of Heme Proteins under Enzymatic Degradation and Low pH. ACS OMEGA 2023; 8:32053-32059. [PMID: 37692240 PMCID: PMC10483679 DOI: 10.1021/acsomega.3c04012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/02/2023] [Indexed: 09/12/2023]
Abstract
There have been significant advances in the development of vaccines for the prevention of various infectious diseases in the last few decades. These vaccines are mainly composed of proteins and nucleic acids. Poor handling and storage, exposure to high temperatures that lead to enzymatic degradation, pH variation, and various other stresses can denature the proteins or nucleic acids present in any vaccine formulation. Therefore, it is necessary to maintain a proper environment to preserve the integrity of biospecimens. To overcome these challenges, we report a practical and user-friendly approach for sol-gels called "BioCaRGOS" that can stabilize heme proteins not only in the presence of degrading enzymes and acidic pH but simultaneously maintain stability at room temperature. Heme proteins, such as myoglobin and cytochrome c, have been used for this study.
Collapse
Affiliation(s)
- Chinmay S Potnis
- Department of Chemistry, University of Louisville, Louisville, Kentucky 40292, United States
| | - Craig A Grapperhaus
- Department of Chemistry, University of Louisville, Louisville, Kentucky 40292, United States
| | - Gautam Gupta
- Department of Chemical Engineering, University of Louisville, Louisville, Kentucky 40292, United States
| |
Collapse
|
17
|
Khan S, Khan M, Lohani M, Ahmad S, Sherwani S, Bhagwath S, Khan MWA, Wahid M, Aqil F, Haque S. NADP/H binding nearly doubles the stability of a Mycobacterium drug target: an unfolding study. J Biomol Struct Dyn 2023; 41:8018-8025. [PMID: 36166625 DOI: 10.1080/07391102.2022.2127910] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/17/2022] [Indexed: 10/14/2022]
Abstract
Mycobacterium Aspartate beta semialdehyde dehydrogenase (ASADH) was studied using various spectroscopic techniques and size exclusion chromatography to examine the unfolding of free (apo) and NADP/H-bound (holo) forms of ASADH. Non-cooperative guanidinium chloride (GdnHCl)-induced unfolding of the apo ASADH was discovered, and no partially folded intermediate structures were stabilized. On the other hand, it was observed that GdnHCl's unfolding of holoenzyme was a cooperative process without any stable intermediate structure. The native form of holoenzyme is found to be stable against the lower concentration of GdnHCl only (namely up to 1.25 M GdnHCl). The tryptophan environment appears to unfold cooperatively in case of the holoenzyme and is in well coordination with the overall unfolding of the holoenzyme. The presence of NADP/H shows a stabilizing effect on the tryptophan environment as well as on the native NADP/H-bound enzyme. Δ G Solvent o values reveal nearly two-fold (∼1.9) conformationally more stable folded holoenzyme compared to its native apo state. The Cm for the apo and holo forms of ASADH are 1.3 and 1.9 M, respectively. Novel drug leads targeting the NADP/H binding domain of ASADH could offer promising drugs against extremely infective Mycobacterium tuberculosis.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Saif Khan
- Department of Basic Dental and Medical Sciences, College of Dentistry, Ha'il University, Ha'il, Saudi Arabia
| | - Mahvish Khan
- Department of Biology, College of Science, University of Ha'il, Ha'il, Saudi Arabia
| | - Mohtashim Lohani
- Department of Emergency Medical Services, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Saheem Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail, Saudi Arabia
| | - Subuhi Sherwani
- Department of Biology, College of Science, University of Ha'il, Ha'il, Saudi Arabia
| | - Sundeep Bhagwath
- Department of Basic Dental and Medical Sciences, College of Dentistry, Ha'il University, Ha'il, Saudi Arabia
| | - Mohd Wajid A Khan
- Department of Chemistry, College of Sciences, University of Ha'il, Ha'il, Saudi Arabia
| | - Mohd Wahid
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Farrukh Aqil
- Department of Medicine and James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
18
|
Rizzo S, Sikorski E, Park S, Im W, Vasquez‐Montes V, Ladokhin AS, Thévenin D. Promoting the activity of a receptor tyrosine phosphatase with a novel pH-responsive transmembrane agonist inhibits cancer-associated phenotypes. Protein Sci 2023; 32:e4742. [PMID: 37515426 PMCID: PMC10461461 DOI: 10.1002/pro.4742] [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: 05/23/2023] [Revised: 07/18/2023] [Accepted: 07/27/2023] [Indexed: 07/30/2023]
Abstract
Cell signaling by receptor protein tyrosine kinases (RTKs) is tightly controlled by the counterbalancing actions of receptor protein tyrosine phosphatases (RPTPs). Due to their role in attenuating the signal-initiating potency of RTKs, RPTPs have long been viewed as therapeutic targets. However, the development of activators of RPTPs has remained limited. We previously reported that the homodimerization of a representative member of the RPTP family (protein tyrosine phosphatase receptor J or PTPRJ) is regulated by specific transmembrane (TM) residues. Disrupting this interaction by single point mutations promotes PTPRJ access to its RTK substrates (e.g., EGFR and FLT3), reduces RTK's phosphorylation and downstream signaling, and ultimately antagonizes RTK-driven cell phenotypes. Here, we designed and tested a series of first-in-class pH-responsive TM peptide agonists of PTPRJ that are soluble in aqueous solution but insert as a helical TM domain in lipid membranes when the pH is lowered to match that of the acidic microenvironment of tumors. The most promising peptide reduced EGFR's phosphorylation and inhibited cancer cell EGFR-driven migration and proliferation, similar to the PTPRJ's TM point mutations. Developing tumor-selective and TM-targeting peptide binders of critical RPTPs could afford a potentially transformative approach to studying RPTP's selectivity mechanism without requiring less specific inhibitors and represent a novel class of therapeutics against RTK-driven cancers.
Collapse
Affiliation(s)
- Sophie Rizzo
- Department of ChemistryLehigh UniversityBethlehemPennsylvaniaUSA
| | - Eden Sikorski
- Department of ChemistryLehigh UniversityBethlehemPennsylvaniaUSA
| | - Soohyung Park
- Department of Biological SciencesLehigh UniversityBethlehemPennsylvaniaUSA
| | - Wonpil Im
- Department of ChemistryLehigh UniversityBethlehemPennsylvaniaUSA
- Department of Biological SciencesLehigh UniversityBethlehemPennsylvaniaUSA
| | - Victor Vasquez‐Montes
- Department of Biochemistry and Molecular BiologyThe University of Kansas Medical CenterKansas CityKansasUSA
| | - Alexey S. Ladokhin
- Department of Biochemistry and Molecular BiologyThe University of Kansas Medical CenterKansas CityKansasUSA
| | - Damien Thévenin
- Department of ChemistryLehigh UniversityBethlehemPennsylvaniaUSA
| |
Collapse
|
19
|
Chen WC, Luo F, Wang T, Wang GX. 4'-(8-(4-Methylimidazole)-octyloxy)-arctigenin: The first inhibitor of fish rhabdovirus glycoprotein. FISH & SHELLFISH IMMUNOLOGY 2023; 139:108920. [PMID: 37385462 DOI: 10.1016/j.fsi.2023.108920] [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: 04/27/2023] [Revised: 06/10/2023] [Accepted: 06/26/2023] [Indexed: 07/01/2023]
Abstract
Spring viraemia of carp virus (SVCV), a highly pathogenic rhabdovirus, could cause spring viraemia of carp (SVC) with up to 90% lethality. Like other rhabdoviruses, the entry of SVCV into susceptible cells was mediated by a single envelope glycoprotein G. Specific inhibitors targeting the glycoprotein were the most effective means to alleviate the epidemic. The programs including SWISS-MODEL, I-TASSER, Phyre2 and AlphaFold2 were used to build a three-dimensional structural model of glycoprotein. The structural comparison between SVCV-G and homology protein VSV-G revealed that the SVCV glycoprotein ectodomain (residues 19 to 466) folded into four distinct domains. Based on the potential small molecule binding sites on glycoprotein surfaces, virtual screening of the anti-SVCV drug libraries was performed using Autodock software and 4'-(8-(4-Methylimidazole)-octyloxy)-arctigenin (MOA) with a high binding affinity was identified. The solubility enhancer tags including trigger factor and maltose binding protein were fused with the ectodomain of glycoprotein, and the target protein with a purity of about 90% was successfully obtained. The interaction confirmation tests revealed that the fluorescence intensity of a characteristic peak induced by the endogenous chromophores in glycoprotein was decreased with the addition of MOA, indicating changes in the microenvironment of glycoprotein. Moreover, the interaction could cause a slight conformational change in glycoprotein, as shown by the content of β-turn, β-folding, and random coil of protein all increased with the decrease of α-helix content after the addition of MOA compound. These results demonstrated that MOA could act as a novel drug against fish rhabdovirus via direct targeting of glycoprotein.
Collapse
Affiliation(s)
- Wei-Chao Chen
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province, 712100, China; College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, Fujian Province, 350002, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, Fujian Province, 350002, China
| | - Fei Luo
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province, 712100, China
| | - Tao Wang
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province, 712100, China
| | - Gao-Xue Wang
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province, 712100, China.
| |
Collapse
|
20
|
Modi R, McKee N, Zhang N, Alwali A, Nelson S, Lohar A, Ostafe R, Zhang DD, Parkinson EI. Stapled Peptides as Direct Inhibitors of Nrf2-sMAF Transcription Factors. J Med Chem 2023; 66:6184-6192. [PMID: 37097833 PMCID: PMC10184664 DOI: 10.1021/acs.jmedchem.2c02037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Indexed: 04/26/2023]
Abstract
Nuclear factor erythroid-related 2-factor 2 (Nrf2) is a transcription factor traditionally thought of as a cellular protector. However, in many cancers, Nrf2 is constitutively activated and correlated with therapeutic resistance. Nrf2 heterodimerizes with small musculoaponeurotic fibrosarcoma Maf (sMAF) transcription factors, allowing binding to the antioxidant responsive element (ARE) and induction of transcription of Nrf2 target genes. While transcription factors are historically challenging to target, stapled peptides have shown great promise for inhibiting these protein-protein interactions. Herein, we describe the first direct cell-permeable inhibitor of Nrf2/sMAF heterodimerization. N1S is a stapled peptide designed based on AlphaFold predictions of the interactions between Nrf2 and sMAF MafG. A cell-based reporter assay combined with in vitro biophysical assays demonstrates that N1S directly inhibits Nrf2/MafG heterodimerization. N1S treatment decreases the transcription of Nrf2-dependent genes and sensitizes Nrf2-dependent cancer cells to cisplatin. Overall, N1S is a promising lead for the sensitization of Nrf2-addicted cancers.
Collapse
Affiliation(s)
- Ramya Modi
- Department
of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Nick McKee
- Department
of Pharmacology and Toxicology, University
of Arizona, Tucson, Arizona 85721, United States
| | - Ning Zhang
- Department
of Pharmacology and Toxicology, University
of Arizona, Tucson, Arizona 85721, United States
| | - Amir Alwali
- Department
of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Samantha Nelson
- Department
of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Aditi Lohar
- Department
of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Raluca Ostafe
- Molecular
Evolution Protein Engineering and Production, Purdue University, West Lafayette, Indiana 47907, United States
| | - Donna D. Zhang
- Department
of Pharmacology and Toxicology, University
of Arizona, Tucson, Arizona 85721, United States
| | - Elizabeth I. Parkinson
- Department
of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
- Department
of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
21
|
Rončević T, Maleš M, Sonavane Y, Guida F, Pacor S, Tossi A, Zoranić L. Relating Molecular Dynamics Simulations to Functional Activity for Gly-Rich Membranolytic Helical Kiadin Peptides. Pharmaceutics 2023; 15:pharmaceutics15051433. [PMID: 37242675 DOI: 10.3390/pharmaceutics15051433] [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: 03/26/2023] [Revised: 04/27/2023] [Accepted: 04/30/2023] [Indexed: 05/28/2023] Open
Abstract
Kiadins are in silico designed peptides with a strong similarity to diPGLa-H, a tandem sequence of PGLa-H (KIAKVALKAL) and with single, double or quadruple glycine substitutions. They were found to show high variability in their activity and selectivity against Gram-negative and Gram-positive bacteria, as well as cytotoxicity against host cells, which are influenced by the number and placing of glycine residues along the sequence. The conformational flexibility introduced by these substitutions contributes differently peptide structuring and to their interactions with the model membranes, as observed by molecular dynamics simulations. We relate these results to experimentally determined data on the structure of kiadins and their interactions with liposomes having a phospholipid membrane composition similar to simulation membrane models, as well as to their antibacterial and cytotoxic activities, and also discuss the challenges in interpreting these multiscale experiments and understanding why the presence of glycine residues in the sequence affected the antibacterial potency and toxicity towards host cells in a different manner.
Collapse
Affiliation(s)
- Tomislav Rončević
- Department of Biology, Faculty of Science, University of Split, 21000 Split, Croatia
| | - Matko Maleš
- Faculty of Maritime Studies, University of Split, 21000 Split, Croatia
| | - Yogesh Sonavane
- Department of Physics, Faculty of Science, University of Split, 21000 Split, Croatia
| | - Filomena Guida
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Sabrina Pacor
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Alessandro Tossi
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Larisa Zoranić
- Department of Physics, Faculty of Science, University of Split, 21000 Split, Croatia
| |
Collapse
|
22
|
Zhao H, Wu D, Hassan SA, Nguyen A, Chen J, Piszczek G, Schuck P. A conserved oligomerization domain in the disordered linker of coronavirus nucleocapsid proteins. SCIENCE ADVANCES 2023; 9:eadg6473. [PMID: 37018390 PMCID: PMC10075959 DOI: 10.1126/sciadv.adg6473] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/07/2023] [Indexed: 06/01/2023]
Abstract
The nucleocapsid (N-)protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has a key role in viral assembly and scaffolding of the viral RNA. It promotes liquid-liquid phase separation (LLPS), forming dense droplets that support the assembly of ribonucleoprotein particles with as-of-yet unknown macromolecular architecture. Combining biophysical experiments, molecular dynamics simulations, and analysis of the mutational landscape, we describe a heretofore unknown oligomerization site that contributes to LLPS, is required for the assembly of higher-order protein-nucleic acid complexes, and is coupled to large-scale conformational changes of N-protein upon nucleic acid binding. The self-association interface is located in a leucine-rich sequence of the intrinsically disordered linker between N-protein folded domains and formed by transient helices assembling into trimeric coiled-coils. Critical residues stabilizing hydrophobic and electrostatic interactions between adjacent helices are highly protected against mutations in viable SARS-CoV-2 genomes, and the oligomerization motif is conserved across related coronaviruses, thus presenting a target for antiviral therapeutics.
Collapse
Affiliation(s)
- Huaying Zhao
- Laboratory of Dynamics of Macromolecular Assembly, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | - Di Wu
- Biophysics Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sergio A. Hassan
- Bioinformatics and Computational Biosciences Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ai Nguyen
- Laboratory of Dynamics of Macromolecular Assembly, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jiji Chen
- Advanced Imaging and Microscopy Resource, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | - Grzegorz Piszczek
- Biophysics Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter Schuck
- Laboratory of Dynamics of Macromolecular Assembly, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
23
|
Viana de Freitas T, Karmakar U, Vasconcelos AG, Santos MA, Oliveira do Vale Lira B, Costa SR, Barbosa EA, Cardozo-Fh J, Correa R, Ribeiro DJS, Prates MV, Magalhães KG, Soller Ramada MH, Roberto de Souza Almeida Leite J, Bloch C, Lima de Oliveira A, Vendrell M, Brand GD. Release of immunomodulatory peptides at bacterial membrane interfaces as a novel strategy to fight microorganisms. J Biol Chem 2023; 299:103056. [PMID: 36822328 PMCID: PMC10074799 DOI: 10.1016/j.jbc.2023.103056] [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: 09/23/2022] [Revised: 02/18/2023] [Accepted: 02/20/2023] [Indexed: 02/23/2023] Open
Abstract
Cationic and amphiphilic peptides can be used as homing devices to accumulate conjugated antibiotics to bacteria-enriched sites and promote efficient microbial killing. However, just as important as tackling bacterial infections, is the modulation of the immune response in this complex microenvironment. In the present report, we designed a peptide chimaera called Chim2, formed by a membrane-active module, an enzyme hydrolysis site and a formyl peptide receptor 2 (FPR2) agonist. This molecule was designed to adsorb onto bacterial membranes, promote their lysis, and upon hydrolysis by local enzymes, release the FPR2 agonist sequence for activation and recruitment of immune cells. We synthesized the isolated peptide modules of Chim2 and characterized their biological activities independently and as a single polypeptide chain. We conducted antimicrobial assays, along with other tests aiming at the analyses of the cellular and immunological responses. In addition, assays using vesicles as models of eukaryotic and prokaryotic membranes were conducted and solution structures of Chim2 were generated by 1H NMR. Chim2 is antimicrobial, adsorbs preferentially to negatively charged vesicles while adopting an α-helix structure and exposes its disorganized tail to the solvent, which facilitates hydrolysis by tryptase-like enzymes, allowing the release of the FPR2 agonist fragment. This fragment was shown to induce accumulation of the cellular activation marker, lipid bodies, in mouse macrophages and the release of immunomodulatory interleukins. In conclusion, these data demonstrate that peptides with antimicrobial and immunomodulatory activities can be considered for further development as drugs.
Collapse
Affiliation(s)
- Thiago Viana de Freitas
- Universidade de Brasília, Instituto de Química, Laboratório de Síntese e Análise de Biomoléculas, LSAB, Brasília, Distrito Federal, Brasil
| | - Utsa Karmakar
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, UK
| | - Andreanne G Vasconcelos
- Universidade de Brasília, Faculdade de Medicina, Núcleo de Pesquisa em Morfologia e Imunologia Aplicada, NuPMIA, Brasília, Distrito Federal, Brasil
| | - Michele A Santos
- Universidade de Brasília, Instituto de Química, Laboratório de Síntese e Análise de Biomoléculas, LSAB, Brasília, Distrito Federal, Brasil; Universidade de Brasília, Instituto de Química, Laboratório de Ressonância Magnética Nuclear, LRMN, Brasília, Distrito Federal, Brasil
| | - Bianca Oliveira do Vale Lira
- Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Distrito Federal, Brasil; Programa de Pós-Graduação em Gerontologia, Universidade Católica de Brasília, Brasília, Distrito Federal, Brasil
| | - Samuel Ribeiro Costa
- Universidade de Brasília, Instituto de Química, Laboratório de Síntese e Análise de Biomoléculas, LSAB, Brasília, Distrito Federal, Brasil
| | - Eder Alves Barbosa
- Universidade de Brasília, Instituto de Química, Laboratório de Síntese e Análise de Biomoléculas, LSAB, Brasília, Distrito Federal, Brasil
| | - José Cardozo-Fh
- Laboratório de Espectrometria de Massa, LEM, Embrapa Recursos Genéticos e Biotecnologia, Brasília, Distrito Federal, Brasil
| | - Rafael Correa
- Universidade de Brasília, Instituto de Biologia, Laboratório de Imunologia e Inflamação, LIMI, Brasília, Distrito Federal, Brasil
| | - Dalila J S Ribeiro
- Universidade de Brasília, Instituto de Biologia, Laboratório de Imunologia e Inflamação, LIMI, Brasília, Distrito Federal, Brasil
| | - Maura Vianna Prates
- Laboratório de Espectrometria de Massa, LEM, Embrapa Recursos Genéticos e Biotecnologia, Brasília, Distrito Federal, Brasil
| | - Kelly G Magalhães
- Universidade de Brasília, Instituto de Biologia, Laboratório de Imunologia e Inflamação, LIMI, Brasília, Distrito Federal, Brasil
| | - Marcelo Henrique Soller Ramada
- Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Distrito Federal, Brasil; Programa de Pós-Graduação em Gerontologia, Universidade Católica de Brasília, Brasília, Distrito Federal, Brasil
| | - José Roberto de Souza Almeida Leite
- Universidade de Brasília, Faculdade de Medicina, Núcleo de Pesquisa em Morfologia e Imunologia Aplicada, NuPMIA, Brasília, Distrito Federal, Brasil
| | - Carlos Bloch
- Laboratório de Espectrometria de Massa, LEM, Embrapa Recursos Genéticos e Biotecnologia, Brasília, Distrito Federal, Brasil
| | - Aline Lima de Oliveira
- Universidade de Brasília, Instituto de Química, Laboratório de Ressonância Magnética Nuclear, LRMN, Brasília, Distrito Federal, Brasil
| | - Marc Vendrell
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, UK
| | - Guilherme Dotto Brand
- Universidade de Brasília, Instituto de Química, Laboratório de Síntese e Análise de Biomoléculas, LSAB, Brasília, Distrito Federal, Brasil.
| |
Collapse
|
24
|
Yanaka S, Yagi-Utsumi M, Kato K, Kuwajima K. The B domain of protein A retains residual structures in 6 M guanidinium chloride as revealed by hydrogen/deuterium-exchange NMR spectroscopy. Protein Sci 2023; 32:e4569. [PMID: 36659853 PMCID: PMC9926473 DOI: 10.1002/pro.4569] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/10/2023] [Accepted: 01/13/2023] [Indexed: 01/21/2023]
Abstract
The characterization of residual structures persistent in unfolded proteins is an important issue in studies of protein folding, because the residual structures present, if any, may form a folding initiation site and guide the subsequent folding reactions. Here, we studied the residual structures of the isolated B domain (BDPA) of staphylococcal protein A in 6 M guanidinium chloride. BDPA is a small three-helix-bundle protein, and until recently its folding/unfolding reaction has been treated as a simple two-state process between the native and the fully unfolded states. We employed a dimethylsulfoxide (DMSO)-quenched hydrogen/deuterium (H/D)-exchange 2D NMR techniques with the use of spin desalting columns, which allowed us to investigate the H/D-exchange behavior of individually identified peptide amide (NH) protons. We obtained H/D-exchange protection factors of the 21 NH protons that form an α-helical hydrogen bond in the native structure, and the majority of these NH protons were significantly protected with a protection factor of 2.0-5.2 in 6 M guanidinium chloride, strongly suggesting that these weakly protected NH protons form much stronger hydrogen bonds under native folding conditions. The results can be used to deduce the structure of an early folding intermediate, when such an intermediate is shown by other methods. Among three native helical regions, the third helix in the C-terminal side was highly protected and stabilized by side-chain salt bridges, probably acting as the folding initiation site of BDPA. The present results are discussed in relation to previous experimental and computational findings on the folding mechanisms of BDPA.
Collapse
Affiliation(s)
- Saeko Yanaka
- Exploratory Research Center on Life and Living Systems (ExCELLS) and Institute for Molecular Science (IMS), National Institutes of Natural Sciences, Myodaiji, Okazaki, Aichi, Japan.,Department of Functional Molecular Science, School of Physical Sciences, SOKENDAI (the Graduate University for Advanced Studies), Myodaiji, Okazaki, Aichi, Japan.,Graduate School of Pharmaceutical Sciences, Nagoya City University, Mizuho-ku, Nagoya, Aichi, Japan
| | - Maho Yagi-Utsumi
- Exploratory Research Center on Life and Living Systems (ExCELLS) and Institute for Molecular Science (IMS), National Institutes of Natural Sciences, Myodaiji, Okazaki, Aichi, Japan.,Department of Functional Molecular Science, School of Physical Sciences, SOKENDAI (the Graduate University for Advanced Studies), Myodaiji, Okazaki, Aichi, Japan.,Graduate School of Pharmaceutical Sciences, Nagoya City University, Mizuho-ku, Nagoya, Aichi, Japan
| | - Koichi Kato
- Exploratory Research Center on Life and Living Systems (ExCELLS) and Institute for Molecular Science (IMS), National Institutes of Natural Sciences, Myodaiji, Okazaki, Aichi, Japan.,Department of Functional Molecular Science, School of Physical Sciences, SOKENDAI (the Graduate University for Advanced Studies), Myodaiji, Okazaki, Aichi, Japan.,Graduate School of Pharmaceutical Sciences, Nagoya City University, Mizuho-ku, Nagoya, Aichi, Japan
| | - Kunihiro Kuwajima
- Department of Physics, School of Science, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
25
|
Qiu C, Whittaker GR, Gellman SH, Daniel S, Abbott NL. Interactions of SARS-CoV-2 and MERS-CoV fusion peptides measured using single-molecule force methods. Biophys J 2023; 122:646-660. [PMID: 36650897 PMCID: PMC9841730 DOI: 10.1016/j.bpj.2023.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 08/07/2022] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
We address the challenge of understanding how hydrophobic interactions are encoded by fusion peptide (FP) sequences within coronavirus (CoV) spike proteins. Within the FPs of severe acute respiratory syndrome CoV 2 and Middle East respiratory syndrome CoV (MERS-CoV), a largely conserved peptide sequence called FP1 (SFIEDLLFNK and SAIEDLLFDK in SARS-2 and MERS, respectively) has been proposed to play a key role in encoding hydrophobic interactions that drive viral-host cell membrane fusion. Although a non-polar triad (Leu-Leu-Phe (LLF)) is common to both FP1 sequences, and thought to dominate the encoding of hydrophobic interactions, FP1 from SARS-2 and MERS differ in two residues (Phe 2 versus Ala 2 and Asn 9 versus Asp 9, respectively). Here we explore whether single-molecule force measurements can quantify hydrophobic interactions encoded by FP1 sequences, and then ask whether sequence variations between FP1 from SARS-2 and MERS lead to significant differences in hydrophobic interactions. We find that both SARS-2 and MERS wild-type FP1 generate measurable hydrophobic interactions at the single-molecule level, but that SARS-2 FP1 encodes a substantially stronger hydrophobic interaction than its MERS counterpart (1.91 ± 0.03 nN versus 0.68 ± 0.03 nN, respectively). By performing force measurements with FP1 sequences with single amino acid substitutions, we determine that a single-residue mutation (Phe 2 versus Ala 2) causes the almost threefold difference in the hydrophobic interaction strength generated by the FP1 of SARS-2 versus MERS, despite the presence of LLF in both sequences. Infrared spectroscopy and circular dichroism measurements support the proposal that the outsized influence of Phe 2 versus Ala 2 on the hydrophobic interaction arises from variation in the secondary structure adopted by FP1. Overall, these insights reveal how single-residue diversity in viral FPs, including FP1 of SARS-CoV-2 and MERS-CoV, can lead to substantial changes in intermolecular interactions proposed to play a key role in viral fusion, and hint at strategies for regulating hydrophobic interactions of peptides in a range of contexts.
Collapse
Affiliation(s)
- Cindy Qiu
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York; Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York
| | - Gary R Whittaker
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York
| | - Samuel H Gellman
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin
| | - Susan Daniel
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York; Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York
| | - Nicholas L Abbott
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York; Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York.
| |
Collapse
|
26
|
He Q, Xu S, Ma X, Ling T, Feng W, Lu X, Liu W, Chen Z. Coupled folding-upon-binding of human tumor suppressor MIG6 to lung cancer EGFR kinase domain and molecular trimming/stapling of MIG6-derived β-hairpins to target the coupling event. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2023; 52:17-25. [PMID: 36547692 DOI: 10.1007/s00249-022-01624-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
Human epidermal growth factor receptor (EGFR) is involved in strong association with malignant proliferation, which has been shown to play a central role in the development and progression of non-small cell lung cancer and other solid tumors. The tumor-suppressor protein MIG6 is a negative regulator of EGFR kinase activity by binding at the activation interface of asymmetric dimer of EGFR kinase domain to disrupt EGFR dimerization and then inactivate the kinase. The protein adopts two discrete fragments 1 and 2 to directly interact with EGFR. It is revealed that the MIG6 fragment 2 is intrinsically disordered in free unbound state, but would fold into a well-structured β-hairpin when binding to EGFR, thus characterized by a so-called coupled folding-upon-binding process, which can be regarded as a compromise between favorable direct readout and unfavorable indirect readout. Here, a 23-mer F2P peptide was derived from MIG6 fragment 2, trimmed into a 17-mer tF2P peptide that contains the binding hotspot region of the fragment 2, and then constrained with an ordered hairpin conformation in free unbound state by disulfide stapling, finally resulting in a rationally stapled/trimmed stF2P peptide that largely minimizes the unfavorable indirect readout effect upon its binding to EGFR kinase domain, with affinity improved considerably upon the trimming and stapling/trimming. These rationally designed β-hairpin peptides may be further exploited as potent anti-lung cancer agents to target the activation event of EGFR dimerization.
Collapse
Affiliation(s)
- Quan He
- Department of Respiratory and Critical Care Medicine, Zhenjiang Hospital of Integrated Traditional Chinese and Western Medicine, Zhenjiang, 212000, China
| | - Shuanglan Xu
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Yunnan University, The Second People's Hospital of Yunnan Province, Kunming, 650021, China
| | - Xiaomei Ma
- Department of Respiratory and Critical Care Medicine, Zhenjiang Hospital of Integrated Traditional Chinese and Western Medicine, Zhenjiang, 212000, China
| | - Ting Ling
- Department of Respiratory and Critical Care Medicine, Zhenjiang Hospital of Integrated Traditional Chinese and Western Medicine, Zhenjiang, 212000, China
| | - Weiqi Feng
- Department of Respiratory and Critical Care Medicine, Zhenjiang Hospital of Integrated Traditional Chinese and Western Medicine, Zhenjiang, 212000, China
| | - Xuzhi Lu
- Department of Respiratory and Critical Care Medicine, Zhenjiang Hospital of Integrated Traditional Chinese and Western Medicine, Zhenjiang, 212000, China
| | - Weihua Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Zi Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
27
|
Rokonujjaman M, Sahyouni A, Wolfe R, Jia L, Ghosh U, Weliky DP. A large HIV gp41 construct with trimer-of-hairpins structure exhibits V2E mutation-dominant attenuation of vesicle fusion and helicity very similar to V2E attenuation of HIV fusion and infection and supports: (1) hairpin stabilization of membrane apposition with larger distance for V2E; and (2) V2E dominance by an antiparallel β sheet with interleaved fusion peptide strands from two gp41 trimers. Biophys Chem 2023; 293:106933. [PMID: 36508984 PMCID: PMC9879285 DOI: 10.1016/j.bpc.2022.106933] [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: 09/29/2022] [Revised: 11/13/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022]
Abstract
There is complete attenuation of fusion and infection mediated by HIV gp160 with gp41 subunit with V2E mutation, and also V2E dominance with WT/V2E mixtures. V2E is at the N-terminus of the ∼25-residue fusion peptide (Fp) which likely binds the target membrane. In this study, large V2E attenuation and dominance were observed for vesicle fusion induced by FP_HM, a large gp41 ectodomain construct with Fp followed by hyperthermostable hairpin with N- and C-helices, and membrane-proximal external region (Mper). FP_HM is a trimer-of-hairpins, the final gp41 structure during fusion. Vesicle fusion and helicity were measured for FP_HM using trimers with different fractions (f's) of WT and V2E proteins. Reductions in FP_HM fusion and helicity vs. fV2E were quantitatively-similar to those for gp160-mediated fusion and infection. Global fitting of all V2E data supports 6 WT gp41 (2 trimers) required for fusion. These data are understood by a model in which the ∼25 kcal/mol free energy for initial membrane apposition is compensated by the thermostable hairpin between the Fp in target membrane and Mper/transmembrane domain in virus membrane. The data support a structural model for V2E dominance with a membrane-bound Fp with antiparallel β sheet and interleaved strands from the two trimers. Relative to fV2E = 0, a longer Fp sheet is stabilized with small fV2E because of salt-bridge and/or hydrogen bonds between E2 on one strand and C-terminal Fp residues on adjacent strands, like R22. A longer Fp sheet results in shorter N- and C-helices, and larger separation during membrane apposition which hinders fusion.
Collapse
Affiliation(s)
- Md Rokonujjaman
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
| | - Abdulrazak Sahyouni
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
| | - Robert Wolfe
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
| | - Lihui Jia
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
| | - Ujjayini Ghosh
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
| | - David P Weliky
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
28
|
Aguilar S, Brunetti AE, Garay AV, Santos LC, Perez LO, Moreira D, Cancelarich NL, Barbosa EA, Basso NG, de Freitas SM, Faivovich J, Brand G, Cabrera GM, Leite JRSA, Marani MM. Structure and function of cationic hylin bioactive peptides from the tree frog Boana pulchella in interaction with lipid membranes. Peptides 2023; 159:170900. [PMID: 36336169 DOI: 10.1016/j.peptides.2022.170900] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/05/2022]
Abstract
Amphibians have a great diversity of bioactive peptides in their skin. The cDNA prepro-peptide sequencing allowed the identification of five novel mature peptides expressed in the skin of Boana pulchella, four with similar sequences to hylin peptides having a cationic amphipathic-helical structure. Whole mature peptides and some of their fragments were chemically-synthesized and tested against Gram-positive and Gram-negative bacterial strains. The mature peptide hylin-Pul3 was the most active, with a MIC= 14 µM against Staphylococcus aureus. Circular dichroism assays indicated that peptides are mostly unstructured in buffer solutions. Still, adding large unilamellar vesicles composed of dimyristoyl phosphatidylcholine and dimyristoylphosphatidylglycerol increased the α-helix content of novel hylins. These results demonstrate the strong influence of the environment on peptide conformation and highlight its significance while addressing the pharmacology of peptides and their biological function in frogs.
Collapse
Affiliation(s)
- Silvana Aguilar
- IPEEC-CONICET, Consejo Nacional de Investigaciones Científicas y Técnicas, U9120ACD Puerto Madryn, Argentina
| | - Andrés E Brunetti
- Laboratorio de Genética Evolutiva, Instituto de Biología Subtropical (CONICET-UNaM), Facultad de Ciencias Exactas Químicas y Naturales, Universidad Nacional de Misiones, N3300LQH Posadas, Argentina; Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Departamento de Ciências Biomoleculares, Universidade de São Paulo, 14040-903 Ribeirão Preto, Brazil
| | - Aisel Valle Garay
- Instituto de Ciências Biológicas, Departamento de Biologia Celular, Laboratório de Biofísica Molecular, Universidade de Brasília (UnB), Brasília, DF 70910-900, Brazil
| | - Liem Canet Santos
- Instituto de Ciências Biológicas, Departamento de Biologia Celular, Laboratório de Biofísica Molecular, Universidade de Brasília (UnB), Brasília, DF 70910-900, Brazil
| | - Luis O Perez
- IPCSH-CONICET, Consejo Nacional de Investigaciones Científicas y Técnicas, Puerto Madryn, Argentina
| | - Daniel Moreira
- Núcleo de Pesquisa em Morfologia e Imunologia Aplicada, NuPMIA, Faculdade de Medicina, Universidade de Brasília, UnB, Brasília, 70910-900, Brazil
| | - Natalia L Cancelarich
- IPEEC-CONICET, Consejo Nacional de Investigaciones Científicas y Técnicas, U9120ACD Puerto Madryn, Argentina
| | - Eder Alves Barbosa
- Núcleo de Pesquisa em Morfologia e Imunologia Aplicada, NuPMIA, Faculdade de Medicina, Universidade de Brasília, UnB, Brasília, 70910-900, Brazil
| | - Néstor G Basso
- IDEAus-CONICET, Consejo Nacional de Investigaciones Científicas y Técnicas, U9120ACD Puerto Madryn, Argentina
| | - Sonia Maria de Freitas
- Instituto de Ciências Biológicas, Departamento de Biologia Celular, Laboratório de Biofísica Molecular, Universidade de Brasília (UnB), Brasília, DF 70910-900, Brazil
| | - Julián Faivovich
- División Herpetología, Museo Argentino de Ciencias Naturales 'Bernardino Rivadavia' (CONICET), Buenos Aires, Argentina
| | - Guilherme Brand
- Laboratório de Síntese e Análise de Biomoléculas, LSAB, Instituto de Química, Universidade de Brasília (UnB), Brasília, DF 70910-900, Brazil
| | - Gabriela M Cabrera
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Orgánica, Universidad de Buenos Aires, Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
| | - José R S A Leite
- Núcleo de Pesquisa em Morfologia e Imunologia Aplicada, NuPMIA, Faculdade de Medicina, Universidade de Brasília, UnB, Brasília, 70910-900, Brazil; Laboratorio de Síntese e Análise de Biomolećulas, Instituto de Química, Universidade de Brasília, Brazil; Laboratorio de Espectrometria de Massa, EMBRAPA Recursos Genéticos e Biotecnología, Brasil, Instituto de Química, Universidade de Brasília, Brazil
| | - Mariela M Marani
- IPEEC-CONICET, Consejo Nacional de Investigaciones Científicas y Técnicas, U9120ACD Puerto Madryn, Argentina.
| |
Collapse
|
29
|
Zhang L, Gong Y, Shen L. Molecular Stapling of Human Pediatric RSV Phosphoprotein’s C-terminal Tail-Derived Peptides to Target the Coupled Folding-Upon-Binding Event Between Phosphoprotein and Nucleocapsid. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10483-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
30
|
Mitchell CJ, Johnson TS, Deber CM. Transmembrane peptide effects on bacterial membrane integrity and organization. Biophys J 2022; 121:3253-3262. [PMID: 35923102 PMCID: PMC9463641 DOI: 10.1016/j.bpj.2022.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 07/10/2022] [Accepted: 07/21/2022] [Indexed: 11/02/2022] Open
Abstract
As the bacterial multidrug resistance crisis continues, membrane-active antimicrobial peptides are being explored as an alternate treatment to conventional antibiotics. In contrast to antimicrobial peptides, which function by a nonspecific membrane disruption mechanism, here we describe a series of transmembrane (TM) peptides that are designed to act as drug efflux inhibitors by aligning with and out-competing a conserved TM4-TM4 homodimerization motif within bacterial small multidrug resistance proteins. The peptides contain two terminal tags: a C-terminal lysine tag to direct the peptides toward the negatively charged bacterial membrane, and an uncharged N-terminal sarcosine (N-methyl-glycine) tag to promote membrane insertion. While effective at inhibiting efflux activity, ostensibly through their designed mechanism of action, the impact of the peptides on the bacterial inner membrane remains undetermined. To evaluate the extant peptide-membrane interactions, we performed a series of biophysical measurements. Circular dichroism spectroscopy and Trp fluorescence showed that the peptides insert into the membrane generally in helical form. Interestingly, differential scanning calorimetry of the peptides added to bacterial-like membranes (POPE:POPG 3:1) revealed the peptides' ability to demix the POPE and POPG lipids, creating two pools, one of which is likely a peptide-POPG conglomerate, and the other a POPE-rich component where the native POPG content has been depleted. However, dye leakage assays confirmed that these events occur without causing significant membrane disruption both in vitro and in vivo, indicating that the peptides can target the small multidrug resistance TM4-TM4 motif without nonspecific membrane disruption. In related studies, DiOC2(3) fluorescence indicated moderate peptide-mediated reduction of the proton motive force for all peptides, including control peptides that did not display inhibitory activity. The overall findings suggest that peptides designed with suitable tags, sequence hydrophobicity, and charge distribution can be directed more generally to impact proteins whose function involves membrane-embedded protein-protein interactions.
Collapse
Affiliation(s)
- Chloe J Mitchell
- Program in Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto M5G 0A4, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto M5S 1A8, Ontario, Canada
| | - Tyler S Johnson
- Program in Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto M5G 0A4, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto M5S 1A8, Ontario, Canada
| | - Charles M Deber
- Program in Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto M5G 0A4, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto M5S 1A8, Ontario, Canada.
| |
Collapse
|
31
|
Zheng Q, Lee B, Kebede MT, Ivancic VA, Kemeh MM, Brito HL, Spratt DE, Lazo ND. Exchange Broadening Underlies the Enhancement of IDE-Dependent Degradation of Insulin by Anionic Membranes. ACS OMEGA 2022; 7:24757-24765. [PMID: 35874268 PMCID: PMC9301717 DOI: 10.1021/acsomega.2c02747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Insulin-degrading enzyme (IDE) is an evolutionarily conserved ubiquitous zinc metalloprotease implicated in the efficient degradation of insulin monomer. However, IDE also degrades monomers of amyloidogenic peptides associated with disease, complicating the development of IDE inhibitors. In this work, we investigated the effects of the lipid composition of membranes on the IDE-dependent degradation of insulin. Kinetic analysis based on chromatography and insulin's helical circular dichroic signal showed that the presence of anionic lipids in membranes enhances IDE's activity toward insulin. Using NMR spectroscopy, we discovered that exchange broadening underlies the enhancement of IDE's activity. These findings, together with the adverse effects of anionic membranes in the self-assembly of IDE's amyloidogenic substrates, suggest that the lipid composition of membranes is a key determinant of IDE's ability to balance the levels of its physiologically and pathologically relevant substrates and achieve proteostasis.
Collapse
Affiliation(s)
| | | | | | - Valerie A. Ivancic
- Gustaf H. Carlson School
of Chemistry and Biochemistry, Clark University, Worcester, Massachusetts 01610, United States
| | - Merc M. Kemeh
- Gustaf H. Carlson School
of Chemistry and Biochemistry, Clark University, Worcester, Massachusetts 01610, United States
| | - Henrique Lemos Brito
- Gustaf H. Carlson School
of Chemistry and Biochemistry, Clark University, Worcester, Massachusetts 01610, United States
| | - Donald E. Spratt
- Gustaf H. Carlson School
of Chemistry and Biochemistry, Clark University, Worcester, Massachusetts 01610, United States
| | - Noel D. Lazo
- Gustaf H. Carlson School
of Chemistry and Biochemistry, Clark University, Worcester, Massachusetts 01610, United States
| |
Collapse
|
32
|
Rončević T, Gerdol M, Mardirossian M, Maleš M, Cvjetan S, Benincasa M, Maravić A, Gajski G, Krce L, Aviani I, Hrabar J, Trumbić Ž, Derks M, Pallavicini A, Weingarth M, Zoranić L, Tossi A, Mladineo I. Anisaxins, helical antimicrobial peptides from marine parasites, kill resistant bacteria by lipid extraction and membrane disruption. Acta Biomater 2022; 146:131-144. [PMID: 35470073 DOI: 10.1016/j.actbio.2022.04.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 04/12/2022] [Accepted: 04/14/2022] [Indexed: 12/21/2022]
Abstract
An infecting and propagating parasite relies on its innate defense system to evade the host's immune response and to survive challenges from commensal bacteria. More so for the nematode Anisakis, a marine parasite that during its life cycle encounters both vertebrate and invertebrate hosts and their highly diverse microbiotas. Although much is still unknown about how the nematode mitigates the effects of these microbiota, its antimicrobial peptides likely play an important role in its survival. We identified anisaxins, the first cecropin-like helical antimicrobial peptides originating from a marine parasite, by mining available genomic and transcriptomic data for Anisakis spp. These peptides are potent bactericidal agents in vitro, selectively active against Gram-negative bacteria, including multi-drug resistant strains, at sub-micromolar concentrations. Their interaction with bacterial membranes was confirmed by solid state NMR (ssNMR) and is highly dependent on the peptide concentration as well as peptide to lipid ratio, as evidenced by molecular dynamics (MD) simulations. MD results indicated that an initial step in the membranolytic mode of action involves membrane bulging and lipid extraction; a novel mechanism which may underline the peptides' potency. Subsequent steps include membrane permeabilization leading to leakage of molecules and eventually cell death, but without visible macroscopic damage, as shown by atomic force microscopy and flow cytometry. This membranolytic antibacterial activity does not translate to cytotoxicity towards human peripheral blood mononuclear cells (HPBMCs), which was minimal at well above bactericidal concentrations, making anisaxins promising candidates for further drug development. STATEMENT OF SIGNIFICANCE: Witnessing the rapid spread of antibiotic resistance resulting in millions of infected and dozens of thousands dying worldwide every year, we identified anisaxins, antimicrobial peptides (AMPs) from marine parasites, Anisakis spp., with potent bactericidal activity and selectivity towards multi-drug resistant Gram-negative bacteria. Anisaxins are membrane-active peptides, whose activity, very sensitive to local peptide concentrations, involves membrane bulging and lipid extraction, leading to membrane permeabilization and bacterial cell death. At the same time, their toxicity towards host cells is negligible, which is often not the case for membrane-active AMPs, therefore making them suitable drug candidates. Membrane bulging and lipid extraction are novel concepts that broaden our understanding of peptide interactions with bacterial functional structures, essential for future design of such biomaterials.
Collapse
Affiliation(s)
- Tomislav Rončević
- Department of Biology, Faculty of Science, University of Split, Ruđera Boškovića 33, Split 21000, Croatia.
| | - Marco Gerdol
- Department of Life Sciences, University of Trieste, Trieste 34127, Italy
| | - Mario Mardirossian
- Department of Medical Sciences, University of Trieste, Trieste 34125, Italy
| | - Matko Maleš
- Faculty of Maritime Studies, University of Split, Split 21000, Croatia
| | - Svjetlana Cvjetan
- Laboratory for Aquaculture, Institute of Oceanography and Fisheries, Split 21000, Croatia
| | - Monica Benincasa
- Department of Life Sciences, University of Trieste, Trieste 34127, Italy
| | - Ana Maravić
- Department of Biology, Faculty of Science, University of Split, Ruđera Boškovića 33, Split 21000, Croatia
| | - Goran Gajski
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, Zagreb 10000, Croatia
| | - Lucija Krce
- Department of Physics, Faculty of Science, University of Split, Split 21000, Croatia
| | - Ivica Aviani
- Department of Physics, Faculty of Science, University of Split, Split 21000, Croatia
| | - Jerko Hrabar
- Laboratory for Aquaculture, Institute of Oceanography and Fisheries, Split 21000, Croatia
| | - Željka Trumbić
- University Department of Marine Studies, University of Split, Split 21000, Croatia
| | - Maik Derks
- NMR spectroscopy, Bijvoet Centre for Biomolecular Research, University of Utrecht, Utrecht 3584CH, The Netherlands; Membrane Biochemistry and Biophysics, Bijvoet Centre for Biomolecular Research, Department of Chemistry, Utrecht University, Padualaan 8, Utrecht 3584 CH, The Netherlands
| | - Alberto Pallavicini
- Department of Life Sciences, University of Trieste, Trieste 34127, Italy; Oceanography Division, Istituto Nazionale di Oceanografia e di Geofisica Sperimentale - OGS, Trieste, Italy
| | - Markus Weingarth
- NMR spectroscopy, Bijvoet Centre for Biomolecular Research, University of Utrecht, Utrecht 3584CH, The Netherlands
| | - Larisa Zoranić
- Department of Physics, Faculty of Science, University of Split, Split 21000, Croatia
| | - Alessandro Tossi
- Department of Life Sciences, University of Trieste, Trieste 34127, Italy
| | - Ivona Mladineo
- Laboratory of Functional Helminthology, Biology Centre Czech Academy of Sciences, Institute of Parasitology BC CAS, Branisovska 31, Ceske Budejovice 37005, Czech Republic.
| |
Collapse
|
33
|
Aupič J, Lapenta F, Strmšek Ž, Merljak E, Plaper T, Jerala R. Metal ion-regulated assembly of designed modular protein cages. SCIENCE ADVANCES 2022; 8:eabm8243. [PMID: 35714197 PMCID: PMC9205593 DOI: 10.1126/sciadv.abm8243] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 05/04/2022] [Indexed: 06/15/2023]
Abstract
Coiled-coil (CC) dimers are versatile, customizable building modules for the design of diverse protein architectures unknown in nature. Incorporation of dynamic self-assembly, regulated by a selected chemical signal, represents an important challenge in the construction of functional polypeptide nanostructures. Here, we engineered metal binding sites to render an orthogonal set of CC heterodimers Zn(II)-responsive as a generally applicable principle. The designed peptides assemble into CC heterodimers only in the presence of Zn(II) ions, reversibly dissociate by metal ion sequestration, and additionally act as pH switches, with low pH triggering disassembly. The developed Zn(II)-responsive CC set is used to construct programmable folding of CC-based nanostructures, from protein triangles to a two-chain bipyramidal protein cage that closes and opens depending on the metal ion. This demonstrates that dynamic self-assembly can be designed into CC-based protein cages by incorporation of metal ion-responsive CC building modules that act as conformational switches and that could also be used in other contexts.
Collapse
Affiliation(s)
- Jana Aupič
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
| | - Fabio Lapenta
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
- EN-FIST Centre of Excellence, Trg OF 13, SI-1000 Ljubljana, Slovenia
| | - Žiga Strmšek
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
| | - Estera Merljak
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
- Interdisciplinary Doctoral Programme in Biomedicine, University of Ljubljana, Kongresni trg 12, SI-1000 Ljubljana, Slovenia
| | - Tjaša Plaper
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
- Interdisciplinary Doctoral Programme in Biomedicine, University of Ljubljana, Kongresni trg 12, SI-1000 Ljubljana, Slovenia
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
- EN-FIST Centre of Excellence, Trg OF 13, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
34
|
Jiang B, Murray C, Cole BL, Glover JNM, Chan GK, Deschenes J, Mani RS, Subedi S, Nerva JD, Wang AC, Lockwood CM, Mefford HC, Leary SES, Ojemann JG, Weinfeld M, Ene CI. Mutations of the DNA repair gene PNKP in a patient with microcephaly, seizures, and developmental delay (MCSZ) presenting with a high-grade brain tumor. Sci Rep 2022; 12:5386. [PMID: 35354845 PMCID: PMC8967877 DOI: 10.1038/s41598-022-09097-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 03/04/2022] [Indexed: 11/08/2022] Open
Abstract
Polynucleotide Kinase-Phosphatase (PNKP) is a bifunctional enzyme that possesses both DNA 3'-phosphatase and DNA 5'-kinase activities, which are required for processing termini of single- and double-strand breaks generated by reactive oxygen species (ROS), ionizing radiation and topoisomerase I poisons. Even though PNKP is central to DNA repair, there have been no reports linking PNKP mutations in a Microcephaly, Seizures, and Developmental Delay (MSCZ) patient to cancer. Here, we characterized the biochemical significance of 2 germ-line point mutations in the PNKP gene of a 3-year old male with MSCZ who presented with a high-grade brain tumor (glioblastoma multiforme) within the cerebellum. Functional and biochemical studies demonstrated these PNKP mutations significantly diminished DNA kinase/phosphatase activities, altered its cellular distribution, caused defective repair of DNA single/double stranded breaks, and were associated with a higher propensity for oncogenic transformation. Our findings indicate that specific PNKP mutations may contribute to tumor initiation within susceptible cells in the CNS by limiting DNA damage repair and increasing rates of spontaneous mutations resulting in pediatric glioma associated driver mutations such as ATRX and TP53.
Collapse
Affiliation(s)
- Bingcheng Jiang
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11560 University Ave., Edmonton, AB, T6G 1Z2, Canada
| | - Cameron Murray
- Department of Biochemistry, University of Alberta, Medical Sciences Building, Edmonton, AB, T6G 2H7, Canada
| | - Bonnie L Cole
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - J N Mark Glover
- Department of Biochemistry, University of Alberta, Medical Sciences Building, Edmonton, AB, T6G 2H7, Canada
| | - Gordon K Chan
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11560 University Ave., Edmonton, AB, T6G 1Z2, Canada
| | - Jean Deschenes
- Department of Laboratory Medicine and Pathology, University of Alberta, Cross Cancer Institute, 11560 University Ave., Edmonton, AB, T6G 1Z2, Canada
| | - Rajam S Mani
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11560 University Ave., Edmonton, AB, T6G 1Z2, Canada
| | - Sudip Subedi
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11560 University Ave., Edmonton, AB, T6G 1Z2, Canada
| | - John D Nerva
- Department of Neurological Surgery, Tulane University, New Orleans, LA, USA
| | - Anthony C Wang
- Department of Neurological Surgery, University of California Los Angeles, Los Angeles, CA, USA
| | | | - Heather C Mefford
- Division of Genetics Medicine, University of Washington, Seattle, WA, USA
| | - Sarah E S Leary
- Division of Pediatric Hematology/Oncology, Seattle Children's Hospital, Seattle, WA, USA
| | - Jeffery G Ojemann
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Michael Weinfeld
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11560 University Ave., Edmonton, AB, T6G 1Z2, Canada.
| | - Chibawanye I Ene
- Department of Neurological Surgery, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
35
|
Neira JL, Araujo-Abad S, Cámara-Artigas A, Rizzuti B, Abian O, Giudici AM, Velazquez-Campoy A, de Juan Romero C. Biochemical and biophysical characterization of PADI4 supports its involvement in cancer. Arch Biochem Biophys 2022; 717:109125. [DOI: 10.1016/j.abb.2022.109125] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 12/12/2022]
|
36
|
Shiratori T, Goto S, Sakaguchi T, Kasai T, Otsuka Y, Higashi K, Makino K, Takahashi H, Komatsu K. Singular value decomposition analysis of the secondary structure features contributing to the circular dichroism spectra of model proteins. Biochem Biophys Rep 2021; 28:101153. [PMID: 34712848 PMCID: PMC8528683 DOI: 10.1016/j.bbrep.2021.101153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
Amyloid fibril formation occurs in restricted environment, such as the interface between intercellular fluids and bio-membranes. Conformational interconversion from α-helix to β-structure does not progress in fluids; however, it can occur after sedimentary aggregation during amyloid fibril formation induced by heat treatment of hen egg white lysozyme (HEWL). Secondary structures of various proteins and denatured proteins titrated with 2,2,2-trifluoroethanol (TFE) were examined using their CD spectra. Gaussian peak/trough and singular value decompositions (SVD) showed that the spectral pattern of the α-helix comprised a sharp trough at wavelength 207 nm and a broad trough at 220 nm. Conversely, we distinguished two patterns for β-sheet-a spread barrel type, corresponding to ConA, and a tightly weaved type, corresponding to the soybean trypsin inhibitor. Herein, we confirmed that the spectral/conformational interconversion of the heat-treated HEWL was not observed in the dissolved fluid.
Collapse
Affiliation(s)
- Tomoki Shiratori
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Satoru Goto
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Tomoyo Sakaguchi
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Takahiro Kasai
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Yuta Otsuka
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Kyohei Higashi
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Kosho Makino
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Hideyo Takahashi
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Kazushi Komatsu
- Department of Mathematics, Faculty of Science, Kochi University, 2-5-1 Akebonocho, Kochi, 780-8520, Japan
| |
Collapse
|
37
|
Characterization of Conjugates between α-Lactalbumin and Benzyl Isothiocyanate-Effects on Molecular Structure and Proteolytic Stability. Molecules 2021; 26:molecules26206247. [PMID: 34684828 PMCID: PMC8539348 DOI: 10.3390/molecules26206247] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/12/2021] [Accepted: 10/12/2021] [Indexed: 11/27/2022] Open
Abstract
In complex foods, bioactive secondary plant metabolites (SPM) can bind to food proteins. Especially when being covalently bound, such modifications can alter the structure and, thus, the functional and biological properties of the proteins. Additionally, the bioactivity of the SPM can be affected as well. Consequently, knowledge of the influence of chemical modifications on these properties is particularly important for food processing, food safety, and nutritional physiology. As a model, the molecular structure of conjugates between the bioactive metabolite benzyl isothiocyanate (BITC, a hydrolysis product of the glucosinolate glucotropaeolin) and the whey protein α-lactalbumin (α-LA) was investigated using circular dichroism spectroscopy, anilino-1-naphthalenesulfonic acid fluorescence, and dynamic light scattering. Free amino groups were determined before and after the BITC conjugation. Finally, mass spectrometric analysis of the BITC-α-LA protein hydrolysates was performed. As a result of the chemical modifications, a change in the secondary structure of α-LA and an increase in surface hydrophobicity and hydrodynamic radii were documented. BITC modification at the ε-amino group of certain lysine side chains inhibited tryptic hydrolysis. Furthermore, two BITC-modified amino acids were identified, located at two lysine side chains (K32 and K113) in the amino acid sequence of α-LA.
Collapse
|
38
|
Residual Helicity at the Active Site of the Histidine Phosphocarrier, HPr, Modulates Binding Affinity to Its Natural Partners. Int J Mol Sci 2021; 22:ijms221910805. [PMID: 34639146 PMCID: PMC8509676 DOI: 10.3390/ijms221910805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/01/2021] [Accepted: 10/02/2021] [Indexed: 11/16/2022] Open
Abstract
The phosphoenolpyruvate-dependent phosphotransferase system (PTS) modulates the preferential use of sugars in bacteria. The first proteins in the cascade are common to all organisms (EI and HPr). The active site of HPr involves a histidine (His15) located immediately before the beginning of the first α-helix. The regulator of sigma D (Rsd) protein also binds to HPr. The region of HPr comprising residues Gly9-Ala30 (HPr9–30), involving the first α-helix (Ala16-Thr27) and the preceding active site loop, binds to both the N-terminal region of EI and intact Rsd. HPr9–30 is mainly disordered. We attempted to improve the affinity of HPr9–30 to both proteins by mutating its sequence to increase its helicity. We designed peptides that led to a marginally larger population in solution of the helical structure of HPr9–30. Molecular simulations also suggested a modest increment in the helical population of mutants, when compared to the wild-type. The mutants, however, were bound with a less favorable affinity than the wild-type to both the N-terminal of EI (EIN) or Rsd, as tested by isothermal titration calorimetry and fluorescence. Furthermore, mutants showed lower antibacterial properties against Staphylococcus aureus than the wild-type peptide. Therefore, we concluded that in HPr, a compromise between binding to its partners and residual structure at the active site must exist to carry out its function.
Collapse
|
39
|
Structural definition of the discrete hotspot sites of BMP-2 conformational wrist epitope and rational design of the hotspot-derived osteogenic peptides against chondrocyte senescence. Bioorg Chem 2021; 116:105382. [PMID: 34598087 DOI: 10.1016/j.bioorg.2021.105382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/10/2021] [Accepted: 09/20/2021] [Indexed: 01/05/2023]
Abstract
The bone morphogenetic protein-2 (BMP-2) is an essential regulator of bone formation and remodeling, which has also been implicated in the pathogenesis of osteoarthritis and its closely related chondrocyte senescence. The BMP-2 uses a conformational wrist epitope and a linear knuckle epitope to interact with type-I (BMPR-I) and type-II (BMPR-II) receptors, respectively. Previously, the knuckle epitope has been intensely studied, but the wrist epitope still remains largely unexplored due to its discontinuous nature. In the present work, the intermolecular interaction of BMP-2 with BMPR-I was investigated systematically at structural, energetic and dynamic levels. Three discrete hotspots that represent the key BMPR-I recognition sites of BMP-2 were identified; they are spatially dispersed over the two monomers of BMP-2 dimer and totally account for 83.5 % binding potency of BMP-2 to BMPR-I (hotspot 1: residues 49-70 in monomer 1; hotspot 2: residues 24-31 in monomer 2; hotspot 3: residues 88-107 in monomer 2). Therefore, we defined the three discrete hotspot sites as the core region of wrist epitope; their contribution to the binding increases in the order: hotspot 2 < hotspot 3 < hotspot 1. We demonstrated that the primary hotspot 1 site has a native U-shaped conformation in the full-length BMP-2 protein context, but it cannot maintain in the native conformation when split from the context to obtain a free hotspot-1 peptide, thus largely impairing its binding potency to BMPR-I. We further employed disulfide-bonded cyclization and head-to-tail cyclization to constrain the peptide conformation, and found that only the former can effectively constrain the peptide into native conformation, thus considerably improving its binding affinity to BMPR-I, whereas the latter totally disorders the native conformation, thus rendering the peptide as a full nonbinder of BMPR-I.
Collapse
|
40
|
Eller MW, Siaw HMH, Dyer RB. Stability of HA2 Prefusion Structure and pH-Induced Conformational Changes in the HA2 Domain of H3N2 Hemagglutinin. Biochemistry 2021; 60:2623-2636. [PMID: 34435771 PMCID: PMC8485334 DOI: 10.1021/acs.biochem.1c00551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Influenza hemagglutinin is the fusion protein that mediates fusion of the viral and host membranes through a large conformational change upon acidification in the developing endosome. The "spring-loaded" model has long been used to describe the mechanism of hemagglutinin and other type 1 viral glycoproteins. This model postulates a metastable conformation of the HA2 subunit, caged from adopting a lower-free energy conformation by the HA1 subunit. Here, using a combination of biochemical and spectroscopic methods, we study a truncated construct of HA2 (HA2*, lacking the transmembrane domain) recombinantly expressed in Escherichia coli as a model for HA2 without the influence of HA1. Our data show that HA2* folds into a conformation like that of HA2 in full length HA and forms trimers. Upon acidification, HA2* undergoes a conformational change that is consistent with the change from pre- to postfusion HA2 in HA. This conformational change is fast and occurs on a time scale that is not consistent with aggregation. These results suggest that the prefusion conformation of HA2 is stable and the change to the postfusion conformation is due to protonation of HA2 itself and not merely uncaging by HA1.
Collapse
Affiliation(s)
- Micah W Eller
- Department of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Hew Ming Helen Siaw
- Department of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - R Brian Dyer
- Department of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
41
|
Anti-biofilm and anti-inflammatory effects of Lycosin-II isolated from spiders against multi-drug resistant bacteria. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1864:183769. [PMID: 34506798 DOI: 10.1016/j.bbamem.2021.183769] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 08/26/2021] [Accepted: 09/01/2021] [Indexed: 12/14/2022]
Abstract
Currently, multidrug-resistant bacteria are rapidly increasing worldwide because of the misuse or overuse of antibiotics. In particular, few options exist for treating infections caused by long-persisting oxacillin-resistant strains and recently proliferating carbapenem-resistant strains. Therefore, alternative treatments are urgently needed. The antimicrobial peptide (AMP) Lycosin-II is a peptide consisting of 21 amino acids isolated from the venom of the spider Lycosa singoriensis. Lycosin-II showed strong antibacterial activity and biofilm inhibition effects against gram-positive and gram-negative bacteria including oxacillin-resistant Staphylococcus aureus (S. aureus) and meropenem-resistant Pseudomonas aeruginosa (P. aeruginosa) isolated from patients. In addition, Lycosin-II was not cytotoxic against human foreskin fibroblast Hs27 or hemolytic against sheep red blood cells at the concentration of which exerted antibacterial activity. The mechanism of action of Lycosin-II involves binding to lipoteichoic acid and lipopolysaccharide of gram-positive and gram-negative bacterial membranes, respectively, to destroy the bacterial membrane. Moreover, Lycosin-II showed anti-inflammatory effects by inhibiting the expression of pro-inflammatory cytokines that are increased during bacterial infection in Hs27 cells. These results suggest that Lycosin-II can serve as a therapeutic agent against infections with multidrug-resistant strains.
Collapse
|
42
|
Synthesis and characterization of Pd(II) antitumor complex, DFT calculation and DNA/BSA binding insight through the combined experimental and theoretical aspects. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130535] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
43
|
de Souza Von Zuben E, Eloy JO, Araujo VHS, Gremião MPD, Chorilli M. Insulin-loaded liposomes functionalized with cell-penetrating peptides: influence on drug release and permeation through porcine nasal mucosa. Colloids Surf A Physicochem Eng Asp 2021. [DOI: 10.1016/j.colsurfa.2021.126624] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
44
|
Mosher CZ, Brudnicki PAP, Gong Z, Childs HR, Lee SW, Antrobus RM, Fang EC, Schiros TN, Lu HH. Green electrospinning for biomaterials and biofabrication. Biofabrication 2021; 13. [PMID: 34102612 DOI: 10.1088/1758-5090/ac0964] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/08/2021] [Indexed: 11/12/2022]
Abstract
Green manufacturing has emerged across industries, propelled by a growing awareness of the negative environmental and health impacts associated with traditional practices. In the biomaterials industry, electrospinning is a ubiquitous fabrication method for producing nano- to micro-scale fibrous meshes that resemble native tissues, but this process traditionally utilizes solvents that are environmentally hazardous and pose a significant barrier to industrial scale-up and clinical translation. Applying sustainability principles to biomaterial production, we have developed a 'green electrospinning' process by systematically testing biologically benign solvents (U.S. Food and Drug Administration Q3C Class 3), and have identified acetic acid as a green solvent that exhibits low ecological impact (global warming potential (GWP) = 1.40 CO2eq. kg/L) and supports a stable electrospinning jet under routine fabrication conditions. By tuning electrospinning parameters, such as needle-plate distance and flow rate, we updated the fabrication of widely utilized biomedical polymers (e.g. poly-α-hydroxyesters, collagen), polymer blends, polymer-ceramic composites, and growth factor delivery systems. Resulting 'green' fibers and composites are comparable to traditional meshes in terms of composition, chemistry, architecture, mechanical properties, and biocompatibility. Interestingly, material properties of green synthetic fibers are more biomimetic than those of traditionally electrospun fibers, doubling in ductility (91.86 ± 35.65 vs. 45 ± 15.07%,n= 10,p< 0.05) without compromising yield strength (1.32 ± 0.26 vs. 1.38 ± 0.32 MPa) or ultimate tensile strength (2.49 ± 0.55 vs. 2.36 ± 0.45 MPa). Most importantly, green electrospinning proves advantageous for biofabrication, rendering a greater protection of growth factors during fiber formation (72.30 ± 1.94 vs. 62.87 ± 2.49% alpha helical content,n= 3,p< 0.05) and recapitulating native ECM mechanics in the fabrication of biopolymer-based meshes (16.57 ± 3.92% ductility, 33.38 ± 30.26 MPa elastic modulus, 1.30 ± 0.19 MPa yield strength, and 2.13 ± 0.36 MPa ultimate tensile strength,n= 10). The eco-conscious approach demonstrated here represents a paradigm shift in biofabrication, and will accelerate the translation of scalable biomaterials and biomimetic scaffolds for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Christopher Z Mosher
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Philip A P Brudnicki
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Zhengxiang Gong
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Hannah R Childs
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Sang Won Lee
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Romare M Antrobus
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Elisa C Fang
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Theanne N Schiros
- Materials Research Science and Engineering Center, Columbia University, New York, NY 10027, United States of America.,Science and Mathematics Department, Fashion Institute of Technology, New York, NY 10001, United States of America
| | - Helen H Lu
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America.,Materials Research Science and Engineering Center, Columbia University, New York, NY 10027, United States of America
| |
Collapse
|
45
|
Choi SR, Wang HM, Shin MH, Lim HS. Hydrophobic Tagging-Mediated Degradation of Transcription Coactivator SRC-1. Int J Mol Sci 2021; 22:6407. [PMID: 34203850 PMCID: PMC8232704 DOI: 10.3390/ijms22126407] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/11/2021] [Accepted: 06/12/2021] [Indexed: 01/19/2023] Open
Abstract
Steroid receptor coactivator-1 (SRC-1) is a transcription coactivator playing a pivotal role in mediating a wide range of signaling pathways by interacting with related transcription factors and nuclear receptors. Aberrantly elevated SRC-1 activity is associated with cancer metastasis and progression, and therefore, suppression of SRC-1 is emerging as a promising therapeutic strategy. In this study, we developed a novel SRC-1 degrader for targeted degradation of cellular SRC-1. This molecule consists of a selective ligand for SRC-1 and a bulky hydrophobic group. Since the hydrophobic moiety on the protein surface could mimic a partially denatured hydrophobic region of a protein, SRC-1 could be recognized as an unfolded protein and experience the chaperone-mediated degradation in the cells through the ubiquitin-proteasome system (UPS). Our results demonstrate that a hydrophobic-tagged chimeric molecule is shown to significantly reduce cellular levels of SRC-1 and suppress cancer cell migration and invasion. Together, these results highlight that our SRC-1 degrader represents a novel class of therapeutic candidates for targeting cancer metastasis. Moreover, we believe that the hydrophobic tagging strategy would be widely applicable to develop peptide-based protein degraders with enhanced cellular activity.
Collapse
Affiliation(s)
- So Ra Choi
- Department of Chemistry and Division of Advanced Material Science, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (S.R.C.); (H.M.W.)
| | - Hee Myeong Wang
- Department of Chemistry and Division of Advanced Material Science, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (S.R.C.); (H.M.W.)
| | - Min Hyeon Shin
- Department of Chemistry and Division of Advanced Material Science, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (S.R.C.); (H.M.W.)
- POSTECH Biotech Center, Pohang 37673, Korea
| | - Hyun-Suk Lim
- Department of Chemistry and Division of Advanced Material Science, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (S.R.C.); (H.M.W.)
| |
Collapse
|
46
|
Liu Q, Zhou J, Gao J, Zhang X, Yang J, Hu C, Chu W, Yao M. Targeting the membrane fusion event of human respiratory syncytial virus with rationally designed α-helical hairpin traps. Life Sci 2021; 280:119695. [PMID: 34111463 DOI: 10.1016/j.lfs.2021.119695] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 12/25/2022]
Abstract
AIMS Rational design of protein scaffolds with specific biological functions/activities has attracted much attention over the past decades. In the present study, we systematically examine the trimer-of-hairpins (TOH) motif of human respiratory syncytial virus (RSV) F protein, which plays a central role in viral membrane fusion and is a coiled-coil six-helix bundle formed by the antiparallel intermolecular interaction between three N-terminal heptad-repeat (HRN) helices and three C-terminal heptad-repeat (HRC) helices. MAIN METHODS A rational strategy that integrates dynamics simulation, thermodynamics calculation, fluorescence polarization and circular dichroism is proposed to design HRC-targeted α-helical hairpin traps based on the crystal template of HRN core. KEY FINDINGS The designed hairpin traps possess a typical helix-turn-helix scaffold that can be stabilized by stapling a disulfide bridge across its helical arms, which are highly structured (helicity >60%) and can mimic the native spatial arrangement of HRN helices in TOH motif to trap the hotspot sites of HRC with effective affinity (Kd is up to 6.4 μM). SIGNIFICANCE The designed α-helical hairpin traps can be used as lead entities for further developing TOH-disrupting agents to target RSV membrane fusion event and the proposed rational design strategy can be readily modified to apply for other type I viruses.
Collapse
Affiliation(s)
- Qiuhong Liu
- Department of Respiratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jinqiao Zhou
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jing Gao
- Department of Respiratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Xiaoqin Zhang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China.
| | - Jingrui Yang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China
| | - Chunling Hu
- Department of Respiratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Weili Chu
- Department of Respiratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Mengying Yao
- Department of Respiratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
47
|
Robustelli J, Baumgart T. Membrane partitioning and lipid selectivity of the N-terminal amphipathic H0 helices of endophilin isoforms. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1863:183660. [PMID: 34090873 DOI: 10.1016/j.bbamem.2021.183660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 11/26/2022]
Abstract
Endophilin is an N-BAR protein, which is characterized by a crescent-shaped BAR domain and an amphipathic helix that contributes to the membrane binding of these proteins. The exact function of that H0 helix has been a topic of debate. In mammals, there are five different endophilin isoforms, grouped into A (three members) and B (two members) subclasses, which have been described to differ in their subcellular localization and function. We asked to what extent molecular properties of the H0 helices of these members affect their membrane targeting behavior. We found that all H0 helices of the endophilin isoforms display a two-state equilibrium between disordered and α-helical states in which the helical secondary structure can be stabilized through trifluoroethanol. The helicities in high TFE were strikingly different among the H0 peptides. We investigated H0-membrane partitioning by the monitoring of secondary structure changes via CD spectroscopy. We found that the presence of anionic phospholipids is critical for all H0 helices partitioning into membranes. Membrane partitioning is found to be sensitive to variations in membrane complexity. Overall, the H0 B subfamily displays stronger membrane partitioning than the H0 A subfamily. The H0 A peptide-membrane binding occurs predominantly through electrostatic interactions. Variation among the H0 A subfamily may be attributed to slight alterations in the amino acid sequence. Meanwhile, the H0 B subfamily displays greater specificity for certain membrane compositions, and this may link H0 B peptide binding to endophilin B's cellular function.
Collapse
Affiliation(s)
- Jaclyn Robustelli
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Tobias Baumgart
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
48
|
Kuroki K, Sakai T, Masuda T, Kawano K, Futaki S. Membrane anchoring of a curvature-inducing peptide, EpN18, promotes membrane translocation of octaarginine. Bioorg Med Chem Lett 2021; 43:128103. [PMID: 33984474 DOI: 10.1016/j.bmcl.2021.128103] [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: 03/19/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 11/25/2022]
Abstract
EpN18 is a curvature-inducing peptide, which loosens lipid packing upon interaction with the cell membrane, and facilitates cell-membrane penetration by arginine-rich cell-penetrating peptides, including octaarginine (R8). In the present study, we conjugated the N-terminal of EpN18 with a pyrenebutyryl (pBu) moiety, which acts as an anchoring unit that increases membrane interactions. Enhanced lipid-packing loosening and cytosolic translocation of R8 were observed by the pBu anchoring of EpN18.
Collapse
Affiliation(s)
- Kakeru Kuroki
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Takayuki Sakai
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Toshihiro Masuda
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Kenichi Kawano
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Shiroh Futaki
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan.
| |
Collapse
|
49
|
A novel peptide design aids in the expression and its simplified process of manufacturing of Insulin Glargine in Pichia pastoris. Appl Microbiol Biotechnol 2021; 105:3061-3074. [PMID: 33821296 DOI: 10.1007/s00253-021-11224-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 02/15/2021] [Accepted: 03/07/2021] [Indexed: 10/21/2022]
Abstract
Manufacturing of insulin and its analogues relied upon in vitro enzymatic cleavages of its precursor forms (single chain precursor, SCP) at both ends of a connecting peptide (C-peptide) that links the respective B-chain and A-chains to corresponding final forms. We have demonstrated a simplified approach of cleaving P. pastoris expressed SCP, distinctly at one site for conversion to insulin glargine. The design of the precursor was made in such a way that there is no C-peptide in the precursor which needs to be removed in the final product. Instead of traditional both side cleavage of the C-peptide and removing the C-peptide (by trypsin), followed by 2nd enzyme reaction (typically carboxipeptidase B), present work established only one side cleavage of the sequence by only trypsin converts the precursor to final insulin glargine product. The novel design of the precursor helped in producing insulin glargine in a single step with an application of single enzyme brought high degree of process efficiencies. Highly purified product was generated through two reversed phase high pressure chromatographic steps. Purified product was compared with the reference product Lantus®, for various physico-chemical and biological properties. Primary, secondary and tertiary structures as well as biological pharmaco-dynamic effects were found comparable. High cell density fermentation that gave a good yield of the SCP, a single step conversion to insulin glargine, enabled by a unique design of SCP and a distinct purification approach, has led to a simplified and economical manufacturing process of this important drug used to treat diabetes. KEY POINTS: • Novel concept for processing single chain precursor of insulin glargine • Simple and economic process for insulin glargine • Physicochemical characterization and animal Pharmacodynamics show similarity to Lantus.
Collapse
|
50
|
Evaluation of α-Chitosan from Crab Shell and β-Chitosan from Squid Gladius Based on Biochemistry Performance. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11073183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The objective of this study is to innovatively evaluate the biochemistry performance of α-chitosan from Portunus trituberculatus shell and β-chitosan from Illex argentinus squid gladius by using the weighted composite index method, and provide a theoretical basis for better development and utilization of chitosan biomedical materials. To build a composite evaluation system, seven key indicators, including molecular weight (Mw), deacetylation degree (DD), water binding capacity (WBC), fat binding capacity (FBC), thermal stability (TS), primary structure and secondary structure, which significantly affect chitosan biochemical characteristics, were determined and analyzed. The viscosity average Mw of chitosan was in the range of 22.5–377.1 kDa, and the DD was 83.4–97.8%. Thermogravimetric (TG) and differential scanning calorimetry (DSC) analyses of commercial chitosan (CS), crab chitosan (CSC) and squid chitosan (CSS) showed a downward trend in TS, while WBC and FBC showed an obvious upward trend. FT-IR had a similar profile in peak shape, but the peak position slightly shifted. CD indicated that chitosan maintained the double helix structure and multiple secondary structural elements. The composite weighted index values of CS, CSC and CSS were 0.85, 0.94 and 1.31 respectively, which indicated that the CSS biochemistry performance was significantly better than CSC, and β-chitosan has great potential in biomedical materials.
Collapse
|