1
|
Fung TS, Ryu KW, Thompson CB. Arginine: at the crossroads of nitrogen metabolism. EMBO J 2025:10.1038/s44318-025-00379-3. [PMID: 39920310 DOI: 10.1038/s44318-025-00379-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 02/09/2025] Open
Abstract
L-arginine is the most nitrogen-rich amino acid, acting as a key precursor for the synthesis of nitrogen-containing metabolites and an essential intermediate in the clearance of excess nitrogen. Arginine's side chain possesses a guanidino group which has unique biochemical properties, and plays a primary role in nitrogen excretion (urea), cellular signaling (nitric oxide) and energy buffering (phosphocreatine). The post-translational modification of protein-incorporated arginine by guanidino-group methylation also contributes to epigenetic gene control. Most human cells do not synthesize sufficient arginine to meet demand and are dependent on exogenous arginine. Thus, dietary arginine plays an important role in maintaining health, particularly upon physiologic stress. How cells adapt to changes in extracellular arginine availability is unclear, mostly because nearly all tissue culture media are supplemented with supraphysiologic levels of arginine. Evidence is emerging that arginine-deficiency can influence disease progression. Here, we review new insights into the importance of arginine as a metabolite, emphasizing the central role of mitochondria in arginine synthesis/catabolism and the recent discovery that arginine can act as a signaling molecule regulating gene expression and organelle dynamics.
Collapse
Affiliation(s)
- Tak Shun Fung
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Keun Woo Ryu
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Craig B Thompson
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
2
|
Kim H, Jang JH, Kim HR, Cho JH. Novel-designed antimicrobial peptides with dual antimicrobial and anti-inflammatory actions against Cutibacterium acnes for acne vulgaris therapy. Biochem Pharmacol 2025; 232:116708. [PMID: 39662606 DOI: 10.1016/j.bcp.2024.116708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/21/2024] [Accepted: 12/05/2024] [Indexed: 12/13/2024]
Abstract
Acne vulgaris is a prevalent skin condition among adolescents, primarily instigated by over-colonization and subsequent inflammation triggered by Cutibacterium acnes. Although topical and oral antibiotics are standard treatments, they often lead to the proliferation of antibiotic-resistant bacteria and are associated with undesirable side effects. Antimicrobial peptides (AMPs) are considered a promising solution to these challenges. In this study, we aimed to develop novel short AMPs to combat C. acnes. By comparing sequences and abstracting the distribution of residue types of established AMPs, we derived a sequence template. Using this template, we crafted novel anti-C. acnes peptides comprising 13 amino acid residues. To enhance their potential therapeutic application, we designed a series of peptides by varying the number and position of the tryptophan residues. Among these peptides, DAP-7 and DAP-10 demonstrated potent antimicrobial activity against both antibiotic-susceptible and -resistant strains of C. acnes, with minimal cytotoxicity. The antimicrobial action of these peptides was attributed to their ability to target the bacterial membrane, resulting in permeabilization and rupture. Moreover, DAP-7 and DAP-10 effectively reduced the expression of pro-inflammatory cytokines induced by C. acnes and remained stable for up to 12 h after exposure to proteases found in acne lesions. Notably, DAP-7 decreased the C. acnes colonies on the ears and significantly alleviated C. acnes-induced ear swelling in a mouse model. Our findings suggest that the DAP-7 and DAP-10 peptides hold promise as candidates for developing a new acne vulgaris treatment.
Collapse
Affiliation(s)
- Hyun Kim
- Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, South Korea
| | - Ju Hye Jang
- Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, South Korea
| | - Ha Rang Kim
- Division of Applied Life Science (BK21Four), Gyeongsang National University, Jinju 52828, South Korea
| | - Ju Hyun Cho
- Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, South Korea; Division of Applied Life Science (BK21Four), Gyeongsang National University, Jinju 52828, South Korea; Division of Life Science, Gyeongsang National University, Jinju 52828, South Korea.
| |
Collapse
|
3
|
Bosquetti LM, Oliveira CS, Cerchiaro G, Oliveira Junior VX. Synthesis and characterization of new antimicrobial peptides derived from Temporin F. J Pept Sci 2025; 31:e3655. [PMID: 39357847 DOI: 10.1002/psc.3655] [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: 05/25/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024]
Abstract
Antimicrobial peptides (AMPs) are a promising source of new compounds against resistant bacteria. Temporins are a class of AMPs found on the amphibian Rana temporaria and show activity against Gram-positive and Gram-negative bacteria. There are few studies on how these antimicrobials have been used, but new Temporin-F derivatives were engineered with Lys-substitutions to assess the impact of the net charge on antimicrobial activity and toxicity. We demonstrated through some assays that it is possible to increase the antibacterial activity while maintaining a reduced peptide hemolytic activity with specific substitutions. Our lead synthetic peptide, G6K-Temporin F, has shown higher antimicrobial activity against Gram-negative and Gram-positive bacteria in vitro (MIC range 2 to 32 μmol L-1), with low hemolytic activity maintained, resulting in an increase in the therapeutic window (TW), of 12.5. Also, it showed more resistant to enzymatic degradation. On the other hand, more significant increases in net charges, such as in P3K-G11K-Temporin F, result in a severe increase in toxicity with lower gains in antimicrobial activity (TW of 0.65). In conclusion, we demonstrated that a moderate increase in net charge can lead to a more active analog and G6K-Temporin F is revealed to be promising as a candidate for new AMP therapeutics.
Collapse
Affiliation(s)
- Lucas Melo Bosquetti
- Center of Human Sciences and Humanities, Federal University of ABC, Santo Andre, SP, Brazil
| | - Cyntia Silva Oliveira
- São Paulo School of Medicine, Postgraduate Program in Molecular Biology, Federal University of São Paulo, Sao Paulo, SP, Brazil
| | - Giselle Cerchiaro
- Center of Human Sciences and Humanities, Federal University of ABC, Santo Andre, SP, Brazil
| | - Vani Xavier Oliveira Junior
- Center of Human Sciences and Humanities, Federal University of ABC, Santo Andre, SP, Brazil
- São Paulo School of Medicine, Postgraduate Program in Molecular Biology, Federal University of São Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|
4
|
Hazrati M, Sukeník L, Vácha R. Split Membrane: A New Model to Accelerate All-Atom MD Simulation of Phospholipid Bilayers. J Chem Inf Model 2025; 65:845-856. [PMID: 39779296 PMCID: PMC11776049 DOI: 10.1021/acs.jcim.4c01664] [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: 09/16/2024] [Revised: 11/30/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
All-atom molecular dynamics simulations are powerful tools for studying cell membranes and their interactions with proteins and other molecules. However, these processes occur on time scales determined by the diffusion rate of phospholipids, which are challenging to achieve in all-atom models. Here, we present a new all-atom model that accelerates lipid diffusion by splitting phospholipid molecules into head and tail groups. The bilayer structure is maintained by using external lateral potentials, which compensate for the lipid split. This split model enhances lateral lipid diffusion more than ten times, allowing faster and cheaper equilibration of large systems with different phospholipid types. The current model has been tested on membranes containing PSM, POPC, POPS, POPE, POPA, and cholesterol. We have also evaluated the interaction of the split model membranes with the Disheveled DEP domain and amphiphilic helix motif of the transcriptional repressor Opi1 as representative of peripheral proteins as well as the dimeric fragment of the epidermal growth factor receptor transmembrane domain and the Human A2A Adenosine of G protein-coupled receptors as representative of transmembrane proteins. The split model can predict the interaction sites of proteins and their preferred phospholipid type. Thus, the model could be used to identify lipid binding sites and equilibrate large membranes at an affordable computational cost.
Collapse
Affiliation(s)
- Mehrnoosh
Khodam Hazrati
- CEITEC—Central
European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech
Republic
| | - Lukáš Sukeník
- CEITEC—Central
European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech
Republic
- Department
of Condensed Matter Physics, Faculty of Science, Masaryk University, Kotlářská 267/2, 611 37 Brno, Czech
Republic
| | - Robert Vácha
- CEITEC—Central
European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech
Republic
- Department
of Condensed Matter Physics, Faculty of Science, Masaryk University, Kotlářská 267/2, 611 37 Brno, Czech
Republic
- National
Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| |
Collapse
|
5
|
Wang C, Xue Y, Guo J, Ma Q, Lu X. From Antibacterial Activity to Molecular Mechanism: Case Study of Hexapeptide RWWRWW and Its Analogues. Chembiochem 2025:e202401065. [PMID: 39835531 DOI: 10.1002/cbic.202401065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 01/22/2025]
Abstract
In recent years, antimicrobial peptides (AMPs) have emerged as a potent weapon against the growing threat of antibiotic resistance. Among AMPs, the ones containing tryptophan (W) and arginine (R) exhibit enhanced antimicrobial properties, benefiting from the unique physicochemical features of the two amino acids. Herein, we designed three hexapeptides, including WR, DWR (D-isomer), and RF, derived from the original sequence, RWWRWW-NH2 (RW). By combining sum frequency generation vibrational spectroscopy (SFG-VS) and molecular dynamics (MD) simulation, we examined AMPs' interactions with model bacterial membrane at the molecular level. Our findings revealed the innate different structural features associated with molecular aggregation and membrane activity between L-(WR, RF and RW) and D-isomer. The D-isomer was demonstrated to aggregate via intermolecular hydrogen bonding, which reduced its membrane adsorption quantity and consequently weakened its disruptive effect on the model membrane; while L-isomers rarely aggregated and thus could fully interact with the model membrane. D-isomer was proven to lack a stable helical structure, while L-isomers adopted helical structures, which was believed to be the reason for DWR's tendency to aggregate easily. This study should contribute to designing novel short-chain AMPs with high efficiency, especially in the case that D-isomers will be used.
Collapse
Affiliation(s)
- Chu Wang
- School of Biological Science and Medical Engineering, State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Yunmo Xue
- School of Biological Science and Medical Engineering, State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Jingyao Guo
- Department of General Dentistry, State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210096, P. R. China
| | - Qian Ma
- Department of General Dentistry, State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210096, P. R. China
| | - Xiaolin Lu
- School of Biological Science and Medical Engineering, State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| |
Collapse
|
6
|
Markova E, Wolowczyk C, Mohamed A, Sofias AM, Martin-Armas M, Sundset R, Berndtsson J, Hak S, Škalko-Basnet N. Liposomal Nω-hydroxy-l-norarginine, a proof-of-concept: Arginase inhibitors can be incorporated in liposomes while retaining their therapeutic activity ex vivo. Eur J Pharm Sci 2025; 204:106959. [PMID: 39521192 DOI: 10.1016/j.ejps.2024.106959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 10/02/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Cancer immunotherapy has evolved significantly over the last decade, with therapeutics targeting the adaptive immune system showing exciting effects in clinics. Yet, the modulation of the innate immune system, particularly the tumor-associated innate immune cells which are an integral part of immune responses in cancer, remains less understood. The arginase 1 (Arg1) pathway is a pivotal metabolic pathway that tumor-associated innate immune cells exploit to create an immunosuppressive tumor microenvironment, leading to the evasion of immune surveillance. The inhibition of Arg1 presents a therapeutic opportunity to reverse this immunosuppression, and Nω‑hydroxy-l-norarginine (nor-NOHA) has emerged as a potent arginase inhibitor with promising in vivo efficacy. However, the rapid systemic clearance of nor-NOHA poses a significant challenge for its therapeutic application. This study pioneers the encapsulation of nor-NOHA in liposomes, aiming to enhance its bioavailability and prolong its inhibitory activity against Arg1. Historically, the extensive interaction between innate immune cells and nanoparticles has been one of the biggest drawbacks in nanomedicine. Here we seek to utilize this effect and deliver liposomal nor-NOHA to the arginase 1 expressing innate immune cells. We systematically investigated the effect of lipid composition, acyl chain length, manufacturing and loading methodology on the encapsulation efficiency (EE%) and release profile of nor-NOHA. Our results indicate that while the manufacturing method and lipid acyl chain length do not significantly impact EE%, they crucially influence the release kinetics of nor-NOHA, with longer acyl chains demonstrating a more sustained release of nor-NOHA from liposomes enabling continuous inhibition of Arg1. Our findings suggest that liposomal nor-NOHA retains its functional inhibitory activity and could offer improved pharmacokinetic properties, making it a compelling base for iterations for further innovative cancer immunotherapeutic strategies in preclinical and clinical evaluations.
Collapse
Affiliation(s)
- Elena Markova
- Nuclear Medicine and Radiation Biology Research Group, Department of Clinical Medicine, Faculty of Health Science, UiT The Arctic University of Norway, Tromsø, Norway; PET Imaging Center Tromsø, University Hospital of North Norway (UNN), Tromsø, Norway.
| | - Camilla Wolowczyk
- Department of Biomedical Laboratory Science, Faculty of Natural Sciences & Centre of Molecular Inflammation Research (CEMIR), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Aly Mohamed
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Science, UiT The Arctic University of Norway, Tromsø, Norway
| | - Alexandros Marios Sofias
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen, Germany
| | - Montserrat Martin-Armas
- Nuclear Medicine and Radiation Biology Research Group, Department of Clinical Medicine, Faculty of Health Science, UiT The Arctic University of Norway, Tromsø, Norway; PET Imaging Center Tromsø, University Hospital of North Norway (UNN), Tromsø, Norway
| | - Rune Sundset
- Nuclear Medicine and Radiation Biology Research Group, Department of Clinical Medicine, Faculty of Health Science, UiT The Arctic University of Norway, Tromsø, Norway; PET Imaging Center Tromsø, University Hospital of North Norway (UNN), Tromsø, Norway
| | - Jens Berndtsson
- Centre for Cellular Imaging, Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sjoerd Hak
- Department of Biotechnology and Nanomedicine, SINTEF Industry & Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Nataša Škalko-Basnet
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Science, UiT The Arctic University of Norway, Tromsø, Norway.
| |
Collapse
|
7
|
Zhu Y, Li B, Xu W, Wang Y, Li G, Bi C, Shao C, Shan A. Association of idealized amphiphiles and protease inhibitors: Conferring antimicrobial peptides with stable antibacterial activity under physiological conditions to combat multidrug-resistant bacteria. Drug Resist Updat 2024; 79:101183. [PMID: 39667175 DOI: 10.1016/j.drup.2024.101183] [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: 09/07/2024] [Revised: 11/20/2024] [Accepted: 12/07/2024] [Indexed: 12/14/2024]
Abstract
AIMS The unstable antimicrobial activity of antimicrobial peptides (AMPs) under physiological conditions (especially the degradation instigated proteases) seems to be a persistent impediment for their successful implementation in clinical trials. Consequently, our objective was to devise AMP engineering frameworks that could sustain robust antibacterial efficacy within physiological environments. METHODS In this work, we harvested AMPs with stable antimicrobial activity under the physiological barriers through the combination of idealized amphiphiles and trypsin inhibitors. RESULTS We screened and identified the lead peptides IK3-A and IK3-S, which showed potent activity against Gram-negative bacteria, including multidrug-resistant (MDR) bacteria, and exhibited promising biocompatibility with mammalian cells. Remarkably, IK3-A and IK3-S maintained sustained antibacterial potency under physiological salts, serum, and protease conditions. Furthermore, both IK3-A and IK3-S kill Gram-negative bacteria by attacking the bacterial cell membrane and inducing oxidative damage (at high concentrations). Crucially, IK3-A and IK3-S have optimal safety and efficacy in mice. CONCLUSIONS This is the first work to compare the effects of different trypsin inhibitors on the resistance of AMPs to protease hydrolysis on the same sequence platform. In conclusion, these findings provide guidance for the molecular design of AMPs with stable antibacterial activity under physiological conditions and facilitates the process of clinical translation of AMPs as antimicrobial biomaterials against MDR bacteria. Moreover, this may stimulate a more general interest in protease inhibitors as molecular scaffolds in the creation of highly stable peptide-based biomaterials.
Collapse
Affiliation(s)
- Yongjie Zhu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Bowen Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Wanying Xu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Yuanmengxue Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Guoyu Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Chongpeng Bi
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Changxuan Shao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China.
| | - Anshan Shan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
8
|
Quintal Bojórquez NDC, Segura Campos MR. Novel peptides derived from S. hispanica seeds induce selective cytotoxicity on human cancer cells. Food Chem 2024; 460:140470. [PMID: 39032303 DOI: 10.1016/j.foodchem.2024.140470] [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: 04/01/2024] [Revised: 07/04/2024] [Accepted: 07/13/2024] [Indexed: 07/23/2024]
Abstract
Cancer prevails as one of the major health concerns worldwide due to the consistent rise in incidence and lack of effective therapies. Previous studies identified the peptides KLKKNL, MLKSKR, and KKYRVF from Salvia hispanica seeds and stated their selective anticancer activity. Thus, this study aimed to determine the cell death pathway induced by these peptides on five cancer cell lines (MCF-7, Caco2, HepG2, DU145, and HeLa). Based on the results of this work, it is possible to suggest that KLKKNL primarily induces selective cancer cell death through the apoptotic pathway in the Caco2 and HeLa lines. On the other hand, the peptide KKYRVF reported the highest statistical (p < 0.05) selective cytotoxic effect on the MCF-7, Caco2, HepG2, and DU145 cancer cell lines by induction of the necrotic pathway. These findings offer some understanding of the selective anticancer effect of KLKKNL, MLKSKR, and KKYRVF.
Collapse
Affiliation(s)
- Nidia Del Carmen Quintal Bojórquez
- Facultad de Ingeniería Química, Universidad Autónoma de Yucatán, Periférico Norte Km. 33.5, Tablaje Catastral 13615, Colonia Chuburná de Hidalgo Inn. Mérida, Yucatán, C.P., Mexico
| | - Maira Rubi Segura Campos
- Facultad de Ingeniería Química, Universidad Autónoma de Yucatán, Periférico Norte Km. 33.5, Tablaje Catastral 13615, Colonia Chuburná de Hidalgo Inn. Mérida, Yucatán, C.P., Mexico.
| |
Collapse
|
9
|
Werner J, Umstätter F, Böhmann MB, Müller H, Beijer B, Hertlein T, Kaschnitz L, Bram V, Kleist C, Klika KD, Mühlberg E, Braune G, Wohlfart S, Gärtner M, Peter S, Zimmermann S, Haberkorn U, Ohlsen K, Brötz-Oesterhelt H, Mier W, Uhl P. Conjugation of Polycationic Peptides Extends the Efficacy Spectrum of β-Lactam Antibiotics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2411406. [PMID: 39499737 DOI: 10.1002/advs.202411406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/28/2024] [Indexed: 11/07/2024]
Abstract
Antibiotic-resistant enterococci represent a significant global health challenge. Unfortunately, most β-lactam antibiotics are not applicable for enterococcal infections due to intrinsic resistance. To extend their antimicrobial spectrum, polycationic peptides are conjugated to examples from each of the four classes of β-lactam antibiotics. Remarkably, the β-lactam-peptide conjugates gained an up to 1000-fold increase in antimicrobial activity against vancomycin-susceptible and vancomycin-resistant enterococci. Even against β-lactam-resistant Gram-negative strains, the conjugates are found to be effective despite their size exceeding the exclusion volume of porins. The extraordinary gain of activity can be explained by an altered mode of killing. Of note, the conjugates showed a concentration-dependent activity in contrast to the parent β-lactam antibiotics that exhibited a time-dependent mode of action. In comparison to the parent β-lactams, the conjugates showed altered affinities to the penicillin-binding proteins. Furthermore, it is found that peptide conjugation also resulted in a different elimination route of the compounds when administered to rodents. In mice systemically infected with vancomycin-resistant enterococci, treatment with a β-lactam-peptide conjugate reduced bacterial burden in the liver compared to its originator. Therefore, peptide modification of β-lactam antibiotics represents a promising platform strategy to broaden their efficacy spectrum, particularly against enterococci.
Collapse
Affiliation(s)
- Julia Werner
- Department of Nuclear Medicine, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Florian Umstätter
- Department of Nuclear Medicine, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Manuel B Böhmann
- Department of Pharmaceutical Technology, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, 69120, Heidelberg, Germany
| | - Hannah Müller
- Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076, Tübingen, Germany
| | - Barbro Beijer
- Department of Nuclear Medicine, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Tobias Hertlein
- Institute of Molecular Infection Biology, University of Würzburg, 97080, Würzburg, Germany
| | - Laura Kaschnitz
- Department of Nuclear Medicine, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Veronika Bram
- Department of Nuclear Medicine, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Christian Kleist
- Department of Nuclear Medicine, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Karel D Klika
- NMR Spectroscopy Analysis Unit, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Eric Mühlberg
- Department of Pharmaceutical Technology, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, 69120, Heidelberg, Germany
| | - Gabriel Braune
- Institute of Molecular Infection Biology, University of Würzburg, 97080, Würzburg, Germany
| | - Sabrina Wohlfart
- Department of Nuclear Medicine, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Martin Gärtner
- Department of Pharmaceutical and Bioorganic Chemistry, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, 69120, Heidelberg, Germany
| | - Silke Peter
- Medical Microbiology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076, Tübingen, Germany
| | - Stefan Zimmermann
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Uwe Haberkorn
- Department of Nuclear Medicine, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Knut Ohlsen
- Institute of Molecular Infection Biology, University of Würzburg, 97080, Würzburg, Germany
| | - Heike Brötz-Oesterhelt
- Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076, Tübingen, Germany
| | - Walter Mier
- Department of Nuclear Medicine, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Philipp Uhl
- Department of Pharmaceutical Technology, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, 69120, Heidelberg, Germany
| |
Collapse
|
10
|
Narang K, Nath A, Hemstrom W, Chu SKS. HaloClass: Salt-Tolerant Protein Classification with Protein Language Models. Protein J 2024; 43:1035-1044. [PMID: 39432175 PMCID: PMC11543744 DOI: 10.1007/s10930-024-10236-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
Salt-tolerant proteins, also known as halophilic proteins, have unique adaptations to function in high-salinity environments. These proteins have naturally evolved in extremophilic organisms, and more recently, are being increasingly applied as enzymes in industrial processes. Due to an abundance of salt-tolerant sequences and a simultaneous lack of experimental structures, most computational methods to predict stability are sequence-based only. These approaches, however, are hindered by a lack of structural understanding of these proteins. Here, we present HaloClass, an SVM classifier that leverages ESM-2 protein language model embeddings to accurately identify salt-tolerant proteins. On a newer and larger test dataset, HaloClass outperforms existing approaches when predicting the stability of never-before-seen proteins that are distal to its training set. Finally, on a mutation study that evaluated changes in salt tolerance based on single- and multiple-point mutants, HaloClass outperforms existing approaches, suggesting applications in the guided design of salt-tolerant enzymes.
Collapse
Affiliation(s)
- Kush Narang
- College of Biological Sciences, University of California, Davis, USA.
| | - Abhigyan Nath
- Department of Biochemistry, Pt. Jawahar Lal Nehru Memorial Medical College, Raipur, India
| | - William Hemstrom
- Department of Biological Sciences, Purdue University, West Lafayette, USA
| | - Simon K S Chu
- Biophysics Graduate Program, University of California, Davis, USA
| |
Collapse
|
11
|
Kasahara K, Kawade R, Nakakido M, Matsunaga R, Akiba H, Entzminger KC, Maruyama T, Okumura SCJ, Caaveiro JMM, Kuroda D, Tsumoto K. Unveiling the structural mechanisms behind high affinity and selectivity in phosphorylated epitope-specific rabbit antibodies. J Biol Chem 2024; 300:107989. [PMID: 39542251 DOI: 10.1016/j.jbc.2024.107989] [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: 07/16/2024] [Revised: 10/14/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
Protein phosphorylation is a crucial process in various cellular functions, and its irregularities have been implicated in several diseases, including cancer. Antibodies are commonly employed to detect protein phosphorylation in research. However, unlike the extensive studies on recognition mechanisms of the phosphate group by proteins such as kinases and phosphatases, only a few studies have explored antibody mechanisms. In this study, we produced and characterized two rabbit monoclonal antibodies that recognize a monophosphorylated Akt peptide. Through crystallography, thermodynamic mutational analyses, and molecular dynamics simulations, we investigated the unique recognition mechanism that enables higher binding affinity and selectivity of the antibodies compared to other generic proteins with lower binding affinity to phosphorylated epitopes. Our results demonstrate that molecular dynamics simulations provide novel insights into the dynamic aspects of molecular recognition of posttranslational modifications by proteins beyond static crystal structures, highlighting how specific atomic level interactions drive the exceptional affinity and selectivity of antibodies.
Collapse
Affiliation(s)
- Keisuke Kasahara
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Raiji Kawade
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Makoto Nakakido
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Ryo Matsunaga
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hiroki Akiba
- Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki City, Osaka, Japan; Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | | | | | | | - Jose M M Caaveiro
- Laboratory of Protein Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka-shi, Japan
| | - Daisuke Kuroda
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan.
| | - Kouhei Tsumoto
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki City, Osaka, Japan; Abwiz Bio Inc., San Diego, California, USA; Medical Proteomics Laboratory, Institute of Medical Sciences, The University of Tokyo, Minato-ku, Tokyo, Japan.
| |
Collapse
|
12
|
Berkowitz N, MacMillan A, Simmons MB, Shinde U, Purdy GE. Structural modeling and characterization of the Mycobacterium tuberculosis MmpL3 C-terminal domain. FEBS Lett 2024; 598:2734-2747. [PMID: 39198717 PMCID: PMC11560685 DOI: 10.1002/1873-3468.15007] [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: 03/21/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024]
Abstract
The Mycobacterium tuberculosis (Mtb) cell envelope provides a protective barrier against the immune response and antibiotics. The mycobacterial membrane protein large (MmpL) family of proteins export cell envelope lipids and siderophores; therefore, these proteins are important for the basic biology and pathogenicity of Mtb. In particular, MmpL3 is essential and a known drug target. Despite interest in MmpL3, the structural data in the field are incomplete. Utilizing homology modeling, AlphaFold, and biophysical techniques, we characterized the cytoplasmic C-terminal domain (CTD) of MmpL3 to better understand its structure and function. Our in silico models of the MmpL11TB and MmpL3TB CTD reveal notable features including a long unstructured linker that connects the globular domain to the last transmembrane (TM) in each transporter, charged lysine and arginine residues facing the membrane, and a C-terminal alpha helix. Our predicted overall structure enables a better understanding of these transporters.
Collapse
Affiliation(s)
- Naomi Berkowitz
- Oregon Health & Science University, Department of Molecular Microbiology & Immunology, Portland, OR, 97239, United States
| | - Allison MacMillan
- Oregon Health & Science University, Department of Molecular Microbiology & Immunology, Portland, OR, 97239, United States
| | - Marit B. Simmons
- Oregon Health & Science University, Department of Molecular Microbiology & Immunology, Portland, OR, 97239, United States
| | - Ujwal Shinde
- Oregon Health & Science University, Biophysics Core Facility, Portland, OR, 97239, United States
| | - Georgiana E. Purdy
- Oregon Health & Science University, Department of Molecular Microbiology & Immunology, Portland, OR, 97239, United States
| |
Collapse
|
13
|
Wong CWY, Burton T, Carrera Montoya J, Birje N, Zhou X, Salazar JK, Mackenzie JM, Rau TF, Teplitski M, Zhang W. Antimicrobial Efficacy of GS-2 on Reusable Food Packaging Materials for Specialty Crops. Foods 2024; 13:3490. [PMID: 39517274 PMCID: PMC11545833 DOI: 10.3390/foods13213490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/17/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
The European Union (EU) regulations mandate 10% of all food packaging to be reusable by 2030. United States (U.S.) exporters of specialty crops face new challenges in ensuring microbiological food safety using reusable packaging. A novel antimicrobial formulation consisting of ammonium carboxylate salt of capric acid and L-arginine (GS-2) was recently developed as a spray coating chemical for food packaging materials. In this study, we evaluated the antimicrobial efficacy of GS-2 against microbial strains representing three foodborne bacterial pathogens (Escherichia coli O157:H7, Listeria monocytogenes, Salmonella enterica), one fungal spoilage organism (Aspergillus niger), and one surrogate viral pathogen (murine norovirus) on three reusable plastic materials (acrylonitrile butadiene styrene, high-density polyethylene, and polypropylene) and one cardboard packaging material, respectively. Different chemical concentrations, exposure times, and storage conditions were individually evaluated for the relative antimicrobial efficacies of GS-2 against these microorganisms. Our results showed that GS-2 was highly effective for inactivating bacterial pathogens on both plastic and cardboard surfaces. For instance, 3% GS-2 achieved a >5 log CFU/in2 reduction in E. coli O157:H7, L. monocytogenes, and S. enterica on tested plastic surfaces at an exposure time of 60 min. However, its efficacy against A. niger and murine norovirus was less optimal, resulting in a ≤1 log CFU/in2 reduction on all tested surfaces. Based on our study, GS-2 demonstrated a strong potential as an antibacterial coating reagent for reusable food packaging materials to minimize pathogen contamination and ensure the safety of the specialty crops.
Collapse
Affiliation(s)
- Catherine W. Y. Wong
- Institute for Food Safety and Health, Department of Food Science and Nutrition, Illinois Institute of Technology, 6502 S Archer Rd., Bedford Park, IL 60501, USA; (C.W.Y.W.)
| | - Thomas Burton
- Department of Microbiology & Immunology, The Peter Doherty Institute for Infection & Immunity, University of Melbourne, 792 Elizabeth Street, Melbourne, VIC 3000, Australia; (T.B.); (J.M.M.)
| | - Julio Carrera Montoya
- Department of Microbiology & Immunology, The Peter Doherty Institute for Infection & Immunity, University of Melbourne, 792 Elizabeth Street, Melbourne, VIC 3000, Australia; (T.B.); (J.M.M.)
| | - Nupoor Birje
- Institute for Food Safety and Health, Department of Food Science and Nutrition, Illinois Institute of Technology, 6502 S Archer Rd., Bedford Park, IL 60501, USA; (C.W.Y.W.)
| | - Xinyi Zhou
- Institute for Food Safety and Health, Department of Food Science and Nutrition, Illinois Institute of Technology, 6502 S Archer Rd., Bedford Park, IL 60501, USA; (C.W.Y.W.)
| | - Joelle K. Salazar
- Division of Food Processing Science and Technology, U.S. Food and Drug Administration, 6502 S Archer Rd., Bedford Park, IL 60501, USA;
| | - Jason M. Mackenzie
- Department of Microbiology & Immunology, The Peter Doherty Institute for Infection & Immunity, University of Melbourne, 792 Elizabeth Street, Melbourne, VIC 3000, Australia; (T.B.); (J.M.M.)
| | - Thomas F. Rau
- Wintermute Biomedical Ltd., Corvallis, MT 59828, USA;
| | - Max Teplitski
- International Food Produce Association, 1901 Pennsylvania Ave. NW Suite 1100, Washington, DC 20006, USA;
| | - Wei Zhang
- Institute for Food Safety and Health, Department of Food Science and Nutrition, Illinois Institute of Technology, 6502 S Archer Rd., Bedford Park, IL 60501, USA; (C.W.Y.W.)
| |
Collapse
|
14
|
Noble JE, Hsiao YW, Kepiro IE, De Santis E, Hoose A, Augagneur C, Lamarre B, Briones A, Hammond K, Bray DJ, Crain J, Ryadnov MG. A Nonlinear Peptide Topology for Synthetic Virions. ACS NANO 2024; 18:29956-29967. [PMID: 39402499 DOI: 10.1021/acsnano.4c10662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
a nonlinear de novo peptide topology for the assembly of synthetic virions is reported. The topology is a backbone cyclized amino-acid sequence in which polar l- and hydrophobic d-amino acid residues of the same-type alternate. This arrangement introduces pseudo C4 symmetries of side chains within the same cyclopeptide ring, allowing for the lateral propagation of cyclopeptides into networks with a [3/6, 4]-fold rotational symmetry closing into virus-like shells. A combination of computational and experimental approaches was used to establish that the topology forms morphologically uniform, nonaggregating and nontoxic nanoscale shells. These effectively encapsulate genetic cargo and promote its intracellular delivery and a target genetic response. The design introduces a nanotechnology inspired solution for engineering virus-like systems thereby expanding traditional molecular biology approaches used to create artificial biology to chemical space.
Collapse
Affiliation(s)
- James E Noble
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, U.K
| | - Ya-Wen Hsiao
- The Hartree Centre, STFC Daresbury Laboratory, Warrington WA4 4AD, U.K
| | - Ibolya E Kepiro
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, U.K
| | | | - Alex Hoose
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, U.K
| | | | | | - Andrea Briones
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, U.K
| | - Katharine Hammond
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, U.K
| | - David J Bray
- The Hartree Centre, STFC Daresbury Laboratory, Warrington WA4 4AD, U.K
| | - Jason Crain
- IBM Research Europe, Hartree Centre, Daresbury WA4 4AD, U.K
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, U.K
| | - Maxim G Ryadnov
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, U.K
- Department of Physics, King's College London, Strand Lane, London WC2R 2LS, U.K
| |
Collapse
|
15
|
Asad Y, Jangili KP, Arshad A, Elma M, Rasuleva K, Akinlalu A, Gao T, Tida UR, Xia W, Sun D. Characterizing biomolecular structure features through an innovative elliptical dichroism spectrometry for cancer detection. Heliyon 2024; 10:e38399. [PMID: 39416845 PMCID: PMC11481634 DOI: 10.1016/j.heliyon.2024.e38399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
This research introduces a novel method for evaluating the structural features of biomolecules, utilizing our innovative Elliptical Dichroism (ED) spectrometer specifically designed for stereochemical analysis. By integrating ED spectrometry with autocorrelation (AC) analysis, we investigate the conformational characteristics of biological molecules such as amino acids, proteins, and extracellular vesicles (EVs) induced by elliptically polarized UV absorption. Our streamlined approach offers a cost-effective and portable solution with minimal sample consumption and supports multiple working modes to efficiently characterize biomolecular structures. The insight from this new approach demonstrates potential applications in using biomolecular characterization for cancer detection.
Collapse
Affiliation(s)
- Yusuf Asad
- Biomedical Engineering Program, North Dakota State University, 1401 Centennial Blvd, Engineering Administration, Room 203, Fargo, ND, 58102, United States
| | - Keerthi Priya Jangili
- Biomedical Engineering Program, North Dakota State University, 1401 Centennial Blvd, Engineering Administration, Room 203, Fargo, ND, 58102, United States
| | - Amara Arshad
- Materials and Nanotechnology, North Dakota State University, Fargo, ND 58108, United States
| | - Maliha Elma
- Department of Electrical and Computer Engineering, North Dakota State University, 1411 Centennial Blvd., 101S Fargo, ND, 58102, United States
| | - Komila Rasuleva
- Biomedical Engineering Program, North Dakota State University, 1401 Centennial Blvd, Engineering Administration, Room 203, Fargo, ND, 58102, United States
| | - Alfred Akinlalu
- Department of Electrical & Computer Engineering, University of Denver, 2155 E. Wesley Ave., Denver, CO, 80210, United States
| | - Tommy Gao
- Department of Electrical & Computer Engineering, University of Denver, 2155 E. Wesley Ave., Denver, CO, 80210, United States
| | - Umamaheswara Rao Tida
- Department of Electrical and Computer Engineering, North Dakota State University, 1411 Centennial Blvd., 101S Fargo, ND, 58102, United States
| | - Wenjie Xia
- Department of Aerospace Engineering, Iowa State University, Ames, IA, 50011, United States
| | - Dali Sun
- Department of Electrical & Computer Engineering, University of Denver, 2155 E. Wesley Ave., Denver, CO, 80210, United States
- Knoebel Institute for Healthy Aging, University of Denver, United States
| |
Collapse
|
16
|
El Mohamad M, Han Q, Dyett B, Yu H, Edgecomb S, Pride MC, Chism CM, Roberts A, Jones D, Tanner EEL, Drummond CJ, Greaves TL, Zhai J. Cytotoxicity and cell membrane interactions of choline-based ionic liquids: Comparing amino acids, acetate, and geranate anions. CHEMOSPHERE 2024; 364:143252. [PMID: 39236918 DOI: 10.1016/j.chemosphere.2024.143252] [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: 05/22/2024] [Revised: 08/29/2024] [Accepted: 09/01/2024] [Indexed: 09/07/2024]
Abstract
Ionic liquids (ILs) have found diverse applications in research and industry. Biocompatible ILs, a subset considered less toxic than traditional ILs, have expanded their applications into biomedical fields. However, there is limited understanding of the toxicity profiles, safe concentrations, and underlying factors driving their toxicity. In this study, we investigated the cytotoxicity of 13 choline-based ILs using four different cell lines: Human dermal fibroblasts (HDF), epidermoid carcinoma cells (A431), cervical cancer cells (HeLa), and gastric cancer cells (AGS). Additionally, we explored the haemolytic activity of these ILs. Our findings showed that the cytotoxic and haemolytic activities of ILs can be attributed to the hydrophobicity of the anions and the pH of the IL solutions. Furthermore, utilising quartz crystal microbalance with dissipation (QCM-D), we delved into the interaction of selected ILs, including choline acetate [Cho][Ac] and choline geranate [Cho][Ge], with model cell membranes composed of 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC). The QCM-D data showed that ILs with higher toxicities exhibited more pronounced interactions with membranes. Increased variations in frequency and dissipation reflected substantial changes in membrane fluidity and mass following the addition of the more toxic ILs. Furthermore, total internal reflection fluorescence microscopy study revealed that [Cho][Ac] could cause lipid rearrangements and pore formation in the membrane, while [Cho][Ge] disrupted the bilayer packing. This study advances our understanding of the cellular toxicities associated with choline-based ILs and provides valuable insights into their mechanisms of action concerning IL-membrane interactions. These findings have significant implications for the safe and informed utilisation of biocompatible ILs in the realm of drug delivery and biotechnology.
Collapse
Affiliation(s)
- Mohamad El Mohamad
- School of Science, STEM College, RMIT University, 124 La Trobe Street, Melbourne, VIC, 3000, Australia
| | - Qi Han
- School of Science, STEM College, RMIT University, 124 La Trobe Street, Melbourne, VIC, 3000, Australia
| | - Brendan Dyett
- School of Science, STEM College, RMIT University, 124 La Trobe Street, Melbourne, VIC, 3000, Australia
| | - Haitao Yu
- School of Science, STEM College, RMIT University, 124 La Trobe Street, Melbourne, VIC, 3000, Australia
| | - Sara Edgecomb
- Department of Chemistry and Biochemistry, The University of Mississippi, University, MS, 38677, USA
| | - Mercedes C Pride
- Department of Chemistry and Biochemistry, The University of Mississippi, University, MS, 38677, USA
| | - Claylee M Chism
- Department of Chemistry and Biochemistry, The University of Mississippi, University, MS, 38677, USA
| | - Angela Roberts
- Department of Chemistry and Biochemistry, The University of Mississippi, University, MS, 38677, USA
| | - Deauntaye Jones
- Department of Chemistry and Biochemistry, The University of Mississippi, University, MS, 38677, USA
| | - Eden E L Tanner
- Department of Chemistry and Biochemistry, The University of Mississippi, University, MS, 38677, USA
| | - Calum J Drummond
- School of Science, STEM College, RMIT University, 124 La Trobe Street, Melbourne, VIC, 3000, Australia.
| | - Tamar L Greaves
- School of Science, STEM College, RMIT University, 124 La Trobe Street, Melbourne, VIC, 3000, Australia.
| | - Jiali Zhai
- School of Science, STEM College, RMIT University, 124 La Trobe Street, Melbourne, VIC, 3000, Australia.
| |
Collapse
|
17
|
Gilbert MA, Keefer-Jacques E, Jadhav T, Antfolk D, Ming Q, Valente N, Shaw GTW, Sottolano CJ, Matwijec G, Luca VC, Loomes KM, Rajagopalan R, Hayeck TJ, Spinner NB. Functional characterization of 2,832 JAG1 variants supports reclassification for Alagille syndrome and improves guidance for clinical variant interpretation. Am J Hum Genet 2024; 111:1656-1672. [PMID: 39043182 PMCID: PMC11339624 DOI: 10.1016/j.ajhg.2024.06.011] [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: 04/10/2024] [Revised: 06/15/2024] [Accepted: 06/24/2024] [Indexed: 07/25/2024] Open
Abstract
Pathogenic variants in the JAG1 gene are a primary cause of the multi-system disorder Alagille syndrome. Although variant detection rates are high for this disease, there is uncertainty associated with the classification of missense variants that leads to reduced diagnostic yield. Consequently, up to 85% of reported JAG1 missense variants have uncertain or conflicting classifications. We generated a library of 2,832 JAG1 nucleotide variants within exons 1-7, a region with a high number of reported missense variants, and designed a high-throughput assay to measure JAG1 membrane expression, a requirement for normal function. After calibration using a set of 175 known or predicted pathogenic and benign variants included within the variant library, 486 variants were characterized as functionally abnormal (n = 277 abnormal and n = 209 likely abnormal), of which 439 (90.3%) were missense. We identified divergent membrane expression occurring at specific residues, indicating that loss of the wild-type residue itself does not drive pathogenicity, a finding supported by structural modeling data and with broad implications for clinical variant classification both for Alagille syndrome and globally across other disease genes. Of 144 uncertain variants reported in patients undergoing clinical or research testing, 27 had functionally abnormal membrane expression, and inclusion of our data resulted in the reclassification of 26 to likely pathogenic. Functional evidence augments the classification of genomic variants, reducing uncertainty and improving diagnostics. Inclusion of this repository of functional evidence during JAG1 variant reclassification will significantly affect resolution of variant pathogenicity, making a critical impact on the molecular diagnosis of Alagille syndrome.
Collapse
Affiliation(s)
- Melissa A Gilbert
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, The Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Pediatric Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| | - Ernest Keefer-Jacques
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Tanaya Jadhav
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Daniel Antfolk
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Qianqian Ming
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Nicolette Valente
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Grace Tzun-Wen Shaw
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Christopher J Sottolano
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, The Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Grace Matwijec
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Vincent C Luca
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Kathleen M Loomes
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ramakrishnan Rajagopalan
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, The Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tristan J Hayeck
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, The Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nancy B Spinner
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, The Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
18
|
Park S, Kim J, Oh SS, Choi SQ. Arginine-Rich Cell-Penetrating Peptides Induce Lipid Rearrangements for Their Active Translocation across Laterally Heterogeneous Membranes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404563. [PMID: 38932459 PMCID: PMC11348069 DOI: 10.1002/advs.202404563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/01/2024] [Indexed: 06/28/2024]
Abstract
Arginine-rich cell-penetrating peptides (CPPs) have emerged as valuable tools for the intracellular delivery of bioactive molecules, but their membrane perturbation during cell penetration is not fully understood. Here, nona-arginine (R9)-mediated membrane reorganization that facilitates the translocation of peptides across laterally heterogeneous membranes is directly visualized. The electrostatic binding of cationic R9 to anionic phosphatidylserine (PS)-enriched domains on a freestanding lipid bilayer induces lateral lipid rearrangements; in particular, in real-time it is observed that R9 fluidizes PS-rich liquid-ordered (Lo) domains into liquid-disordered (Ld) domains, resulting in the membrane permeabilization. The experiments with giant unilamellar vesicles (GUVs) confirm the preferential translocation of R9 through Ld domains without pore formation, even when Lo domains are more negatively charged. Indeed, whenever R9 comes into contact with negatively charged Lo domains, it dissolves the Lo domains first, promoting translocation across phase-separated membranes. Collectively, the findings imply that arginine-rich CPPs modulate lateral membrane heterogeneity, including membrane fluidization, as one of the fundamental processes for their effective cell penetration across densely packed lipid bilayers.
Collapse
Affiliation(s)
- Sujin Park
- Department of Chemical and Biomolecular EngineeringKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| | - Jinmin Kim
- Department of Materials Science and EngineeringPohang University of Science and Technology (POSTECH)Pohang37673Republic of Korea
| | - Seung Soo Oh
- Department of Materials Science and EngineeringPohang University of Science and Technology (POSTECH)Pohang37673Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology (I‐CREATE)Yonsei UniversityIncheon21983Republic of Korea
| | - Siyoung Q. Choi
- Department of Chemical and Biomolecular EngineeringKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| |
Collapse
|
19
|
Gandolfi MG, Taddei P, Zamparini F, Ottolenghi L, Polimeni A, Prati C. Dentine surface modification and remineralization induced by bioactive toothpastes. Int J Dent Hyg 2024; 22:554-574. [PMID: 37424392 DOI: 10.1111/idh.12710] [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: 03/28/2022] [Revised: 04/05/2023] [Accepted: 06/14/2023] [Indexed: 07/11/2023]
Abstract
OBJECTIVE In this study, dentine surface was analysed through Environmental-scanning-electron-microscopy (ESEM) with energy-dispersive-X-ray-spectrometry (EDX) and Fourier-transform-infrared-spectroscopy (FTIR) with attenuated total-reflectance (ATR) to assess the morpho-chemical changes and variations in mineralization degree after demineralizing treatment, after five toothpastes application (HA & Citrate toothpaste, Zinc-HA toothpaste, Calcium Sodium Phosphosilicate toothpaste, Arginine & Calcium carbonate toothpaste, Colgate-Triple-Action, and Control toothpaste), after soaking in artificial saliva and after citric acid attack. METHODS Ca/P, Ca/N and P/N ratios were calculated from EDX atomic data to evaluate the mineralization degree of dentine surface. The IR calcium phosphate (CaP)/collagen and carbonate/collagen ratios has been evaluated to assess the remineralization changes in dentine; the carbonate/collagen IR ratio was calculated to identify the nucleation of B-type-carbonated apatite and calcium carbonate. RESULTS ESEM-EDX and ATR-FTIR showed residuals of toothpastes after the treatments in all cases, with a general increase in the mineralization degree after soaking in artificial saliva and a decrease after acid attack. Treatment with Arginine & Calcium carbonate toothpaste showed the highest Ca/P value after treatment (Ca/P 1.62) and acid attack (Ca/P 1.5) in confirmation, IR showed the highest amount of carbonate after treatment and soaking in artificial saliva. Arginine and calcium carbonate toothpaste and HA and citrate toothpaste remained to a higher extent on the dentine surface and revealed a higher remineralization activity. These formulations showed higher resistance to demineralization attack, as demonstrated by a higher ICaP/IAmide II intensity ratio than those obtained after EDTA treatment. CONCLUSIONS Toothpastes that remained to a higher extent on dentine surface (arginine and calcium carbonate toothpaste in particular) were more able to promote remineralization. The formed calcium phosphate (CaPs) phase was intimately bound to dentine rather than a simple deposit.
Collapse
Affiliation(s)
- Maria Giovanna Gandolfi
- Laboratory of Green Biomaterials and Oral Pathology, School of Dentistry, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Paola Taddei
- Biochemistry Unit, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Fausto Zamparini
- Laboratory of Green Biomaterials and Oral Pathology, School of Dentistry, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Livia Ottolenghi
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, Rome, Italy
| | - Antonella Polimeni
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, Rome, Italy
| | - Carlo Prati
- Endodontic Clinical Section, School of Dentistry, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
20
|
Bai Y, Zhang W, Zheng W, Meng XZ, Duan Y, Zhang C, Chen F, Wang KJ. A 14-amino acid cationic peptide Bolespleenin 334-347 from the marine fish mudskipper Boleophthalmus pectinirostris exhibiting potent antimicrobial activity and therapeutic potential. Biochem Pharmacol 2024; 226:116344. [PMID: 38852647 DOI: 10.1016/j.bcp.2024.116344] [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: 02/07/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
Antimicrobial peptides (AMPs) are an important component of innate immunity in both vertebrates and invertebrates, and some of the unique characteristics of AMPs are usually associated with their living environment. The marine fish, mudskipper Boleophthalmus pectinirostris, usually live amphibiously in intertidal environments that are quite different from other fish species, which would be an exceptional source of new AMPs. In the study, an AMP named Bolespleenin334-347 was identified, which was a truncated peptide derived from a new functional gene found in B. pectinirostris, that was up-regulated in response to bacterial challenge. Bolespleenin334-347 had only 14 amino acid residues, including five consecutive arginine residues. It was found that the peptide had broad-spectrum antibacterial activity, good thermal stability and sodium ion tolerance. Bolespleenin334-347 killed Acinetobacter baumannii and Staphylococcus aureus by disrupting the structural integrity of the bacterial membrane, leading to leakage of the cellular contents, and inducing accumulation of bacterial endogenous reactive oxygen species (ROS). In addition, Bolespleenin334-347 effectively inhibited biofilm formation of A. baumannii and S. aureus and long-term treatment did not lead to the development of resistance. Importantly, Bolespleenin334-347 maintained stable activity against clinically multi-drug resistant bacterial strains. In addition, it was noteworthy that Bolespleenin334-347 showed superior efficacy to LL-37 and vancomycin in a constructed mouse model of MRSA-induced superficial skin infections, as evidenced by a significant reduction in bacterial load and more favorable wound healing. This study provides an effective antimicrobial agent for topical skin infections with potential therapeutic efficacy for infections with drug-resistant bacteria, including MRSA.
Collapse
Affiliation(s)
- Yuqi Bai
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China
| | - Weibin Zhang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China
| | - Wenbin Zheng
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xin-Zhan Meng
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yingyi Duan
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China
| | - Chang Zhang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China
| | - Fangyi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China; State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Innovation Research Institute for Marine Biological Antimicrobial Peptide Industrial Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen, China.
| | - Ke-Jian Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China; State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Innovation Research Institute for Marine Biological Antimicrobial Peptide Industrial Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen, China.
| |
Collapse
|
21
|
Todorovic J, Swapna I, Suma A, Carnevale V, Zakon H. Dual mechanisms contribute to enhanced voltage dependence of an electric fish potassium channel. Biophys J 2024; 123:2097-2109. [PMID: 38429925 PMCID: PMC11309972 DOI: 10.1016/j.bpj.2024.02.028] [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: 11/28/2023] [Revised: 01/31/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024] Open
Abstract
The voltage dependence of different voltage-gated potassium channels, described by the voltage at which half of the channels are open (V1/2), varies over a range of 80 mV and is influenced by factors such as the number of positive gating charges and the identity of the hydrophobic amino acids in the channel's voltage sensor (S4). Here we explore by experimental manipulations and molecular dynamics simulation the contributions of two derived features of an electric fish potassium channel (Kv1.7a) that is among the most voltage-sensitive Shaker family potassium channels known. These are a patch of four contiguous negatively charged glutamates in the S3-S4 extracellular loop and a glutamate in the S3b helix. We find that these negative charges affect V1/2 by separate, complementary mechanisms. In the closed state, the S3-S4 linker negative patch reduces the membrane surface charge biasing the channel to enter the open state while, upon opening, the negative amino acid in the S3b helix faces the second (R2) gating charge of the voltage sensor electrostatically biasing the channel to remain in the open state. This work highlights two evolutionary novelties that illustrate the potential influence of negatively charged amino acids in extracellular loops and adjacent helices to voltage dependence.
Collapse
Affiliation(s)
- Jelena Todorovic
- Department of Neuroscience, The University of Texas, Austin, Texas
| | - Immani Swapna
- Department of Neuroscience, The University of Texas, Austin, Texas
| | - Antonio Suma
- Institute for Computational Molecular Science, College of Science and Technology & Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, Pennsylvania
| | - Vincenzo Carnevale
- Institute for Computational Molecular Science, College of Science and Technology & Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, Pennsylvania
| | - Harold Zakon
- Department of Neuroscience, The University of Texas, Austin, Texas; Department of Integrative Biology, The University of Texas, Austin, Texas.
| |
Collapse
|
22
|
Naing MD, Juliano SA, Angeles-Boza AM. Synergy between the clavanins as a weapon against multidrug-resistant Enterobacter cloacae. RSC Med Chem 2024; 15:2160-2164. [PMID: 38911167 PMCID: PMC11187565 DOI: 10.1039/d4md00070f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 05/14/2024] [Indexed: 06/25/2024] Open
Abstract
Finding new antibiotics that can act synergistically with each other offers many benefits such as lower dosages used for each drug, improved pathogen clearance, and ability to act against multi-drug resistant strains. In this study, six peptides isolated from the tunicate Styela clava were evaluated for their synergistic interaction using the checkerboard assay and the time kill kinetics assay. Using two different tests, we report synergy between clavanin D and clavaspirin in both tests and synergy between clavanin A and B only in the checkerboard test when used against the multidrug resistant E. cloacae 0136. This work demonstrates the possible cooperativity between homologous AMPs from a single organism and the advantage of using two susceptibility tests instead of one when testing synergistic combinations.
Collapse
Affiliation(s)
- Marvin D Naing
- Department of Chemistry, University of Connecticut Storrs 06269 USA
| | - Samuel A Juliano
- Department of Chemistry, University of Connecticut Storrs 06269 USA
| | - Alfredo M Angeles-Boza
- Department of Chemistry, University of Connecticut Storrs 06269 USA
- Institute of Materials Science, University of Connecticut Storrs 06269 USA
| |
Collapse
|
23
|
Wang Y, Zhang Y, Su R, Wang Y, Qi W. Antimicrobial therapy based on self-assembling peptides. J Mater Chem B 2024; 12:5061-5075. [PMID: 38726712 DOI: 10.1039/d4tb00260a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
The emergence of drug-resistant microorganisms has threatened global health, and microbial infections have severely limited the use of medical materials. For example, the attachment and colonization of pathogenic bacteria to medical implant materials can lead to wound infections, inflammation and complications, as well as implant failure, shortening their lifespan and even resulting in patient death. In the era of antibiotic resistance, antimicrobial drug discovery needs to prioritize unconventional therapies that act on new targets or adopt new mechanisms. In this regard, supramolecular antimicrobial peptides have emerged as attractive therapeutic platforms, both as bactericides for combination antibiotics and as delivery vehicles. By taking advantage of their programmable intermolecular and intramolecular interactions, peptides can be modified to form higher-order structures (including nanofibers and nanoparticles) with unique functionality. This paper begins with an analysis of the relationship between peptide self-assembly and antimicrobial activity, describes in detail the research and development of various self-assembled antimicrobial peptides in recent years, and finally explores different combinatorial strategies for self-assembling antimicrobial peptides.
Collapse
Affiliation(s)
- Yuqi Wang
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China.
| | - Yexi Zhang
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China.
| | - Rongxin Su
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China.
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300072, P. R. China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Yuefei Wang
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China.
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Wei Qi
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China.
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300072, P. R. China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, P. R. China
| |
Collapse
|
24
|
Alavizargar A, Gass M, Krahn MP, Heuer A. Elucidating the Membrane Binding Process of a Disordered Protein: Dynamic Interplay of Anionic Lipids and the Polybasic Region. ACS PHYSICAL CHEMISTRY AU 2024; 4:167-179. [PMID: 38560754 PMCID: PMC10979486 DOI: 10.1021/acsphyschemau.3c00051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 04/04/2024]
Abstract
Intrinsically disordered regions of proteins are responsible for many biological processes such as in the case of liver kinase B1 (LKB1)-a serine/threonine kinase relevant for cell proliferation and cell polarity. LKB1 becomes fully activated upon recruitment to the plasma membrane by binding of its disordered C-terminal polybasic motif consisting of eight lysines/arginines to phospholipids. Here, we present extensive molecular dynamics (MD) simulations of the polybasic motif interacting with a model membrane composed of 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) and 1-palmitoyl-2-oleyl phosphatidic acid (PA) and cell culture experiments. Protein-membrane binding effects are due to the electrostatic interactions between the polybasic amino acids and PAs. For significant binding, the first three lysines turn out to be dispensable, which was also recapitulated in cell culture using transfected GFP-LKB1 variants. LKB1-membrane binding results in nonmonotonous changes in the structure of the protein as well as the membrane, in particular, accumulation of PAs and reduced thickness at the protein-membrane contact area. The protein-lipid binding turns out to be highly dynamic due to an interplay of PA-PA repulsion and protein-PA attraction. The thermodynamics of this interplay is captured by a statistical fluctuation model, which allows the estimation of both energies. Quantification of the significance of each polar amino acid in the polybasic provides detailed insights into the molecular mechanism of protein-membrane binding of LKB1. These results can likely be transferred to other proteins, which interact by intrinsically disordered polybasic regions with anionic membranes.
Collapse
Affiliation(s)
- Azadeh Alavizargar
- Institute
of Physical Chemistry, University of Münster, Corrensstr. 28/30, 48149 Münster, Germany
| | - Maximilian Gass
- Medical
Cell Biology, Medical Clinic D, University
Hospital of Münster, Albert-Schweitzer Campus 1-A14, 48149 Münster, Germany
| | - Michael P. Krahn
- Medical
Cell Biology, Medical Clinic D, University
Hospital of Münster, Albert-Schweitzer Campus 1-A14, 48149 Münster, Germany
| | - Andreas Heuer
- Institute
of Physical Chemistry, University of Münster, Corrensstr. 28/30, 48149 Münster, Germany
| |
Collapse
|
25
|
Gámez F, Avilés-Moreno JR, Martens J, Berden G, Oomens J, Martínez-Haya B. Vibrational signatures of dynamic excess proton storage between primary amine and carboxylic acid groups. J Chem Phys 2024; 160:094311. [PMID: 38450729 DOI: 10.1063/5.0192331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/20/2024] [Indexed: 03/08/2024] Open
Abstract
Ammonium and carboxylic moieties play a central role in proton-mediated processes of molecular recognition, charge transfer or chemical change in (bio)materials. Whereas both chemical groups constitute acid-base pairs in organic salt-bridge structures, they may as well host excess protons in acidic environments. The binding of excess protons often precedes proton transfer reactions and it is therefore of fundamental interest, though challenging from a quantum chemical perspective. As a benchmark for this process, we investigate proton storage in the amphoteric compound 5-aminovaleric acid (AV), within an intramolecular proton bond shared by its primary amine and carboxylic acid terminal groups. Infrared ion spectroscopy is combined with ab initio Molecular Dynamics (AIMD) calculations to expose and rationalize the spectral signatures of protonated AV and its deuterated isotopologues. The dynamic character of the proton bond confers a fluxional structure to the molecular framework, leading to wide-ranging bands in the vibrational spectrum. These features are reproduced with remarkable accuracy by AIMD computations, which serves to lay out microscopic insights into the excess proton binding scenario.
Collapse
Affiliation(s)
- F Gámez
- Department of Physical Chemistry, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - J R Avilés-Moreno
- Department of Applied Physical Chemistry, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - J Martens
- FELIX Laboratory, Institute for Molecules and Materials, Radboud University, Toernooiveld 7, 6525ED Nijmegen, The Netherlands
| | - G Berden
- FELIX Laboratory, Institute for Molecules and Materials, Radboud University, Toernooiveld 7, 6525ED Nijmegen, The Netherlands
| | - J Oomens
- FELIX Laboratory, Institute for Molecules and Materials, Radboud University, Toernooiveld 7, 6525ED Nijmegen, The Netherlands
| | - B Martínez-Haya
- Center for Nanoscience and Sustainable Technologies (CNATS), Universidad Pablo de Olavide, 41013 Seville, Spain
| |
Collapse
|
26
|
Zhuang Y, Quirk S, Stover ER, Bureau HR, Allen CR, Hernandez R. Tertiary Plasticity Drives the Efficiency of Enterocin 7B Interactions with Lipid Membranes. J Phys Chem B 2024; 128:2100-2113. [PMID: 38412510 PMCID: PMC10926100 DOI: 10.1021/acs.jpcb.3c08199] [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: 02/29/2024]
Abstract
The ability of antimicrobial peptides to efficiently kill their bacterial targets depends on the efficiency of their binding to the microbial membrane. In the case of enterocins, there is a three-part interaction: initial binding, unpacking of helices on the membrane surface, and permeation of the lipid bilayer. Helical unpacking is driven by disruption of the peptide hydrophobic core when in contact with membranes. Enterocin 7B is a leaderless enterocin antimicrobial peptide produced from Enterococcus faecalis that functions alone, or with its cognate partner enterocin 7A, to efficiently kill a wide variety of Gram-stain positive bacteria. To better characterize the role that tertiary structural plasticity plays in the ability of enterocin 7B to interact with the membranes, a series of arginine single-site mutants were constructed that destabilize the hydrophobic core to varying degrees. A series of experimental measures of structure, stability, and function, including CD spectra, far UV CD melting profiles, minimal inhibitory concentrations analysis, and release kinetics of calcein, show that decreased stabilization of the hydrophobic core is correlated with increased efficiency of a peptide to permeate membranes and in killing bacteria. Finally, using the computational technique of adaptive steered molecular dynamics, we found that the atomistic/energetic landscape of peptide mechanical unfolding leads to free energy differences between the wild type and its mutants, whose trends correlate well with our experiment.
Collapse
Affiliation(s)
- Yi Zhuang
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Stephen Quirk
- Kimberly-Clark Corporation, Atlanta, Georgia 30076-2199, United States
| | - Erica R Stover
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Hailey R Bureau
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Caley R Allen
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Rigoberto Hernandez
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Materials Science & Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| |
Collapse
|
27
|
Clarke RJ. Electrostatic switch mechanisms of membrane protein trafficking and regulation. Biophys Rev 2023; 15:1967-1985. [PMID: 38192346 PMCID: PMC10771482 DOI: 10.1007/s12551-023-01166-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 11/17/2023] [Indexed: 01/10/2024] Open
Abstract
Lipid-protein interactions are normally classified as either specific or general. Specific interactions refer to lipid binding to specific binding sites within a membrane protein, thereby modulating the protein's thermal stability or kinetics. General interactions refer to indirect effects whereby lipids affect membrane proteins by modulating the membrane's physical properties, e.g., its fluidity, thickness, or dipole potential. It is not widely recognized that there is a third distinct type of lipid-protein interaction. Intrinsically disordered N- or C-termini of membrane proteins can interact directly but nonspecifically with the surrounding membrane. Many peripheral membrane proteins are held to the cytoplasmic surface of the plasma membrane via a cooperative combination of two forces: hydrophobic anchoring and electrostatic attraction. An acyl chain, e.g., myristoyl, added post-translationally to one of the protein's termini inserts itself into the lipid matrix and helps hold peripheral membrane proteins onto the membrane. Electrostatic attraction occurs between positively charged basic amino acid residues (lysine and arginine) on one of the protein's terminal tails and negatively charged phospholipid head groups, such as phosphatidylserine. Phosphorylation of either serine or tyrosine residues on the terminal tails via regulatory protein kinases allows for an electrostatic switch mechanism to control trafficking of the protein. Kinase action reduces the positive charge on the protein's tail, weakening the electrostatic attraction and releasing the protein from the membrane. A similar mechanism regulates many integral membrane proteins, but here only electrostatic interactions are involved, and the electrostatic switch modulates protein activity by altering the stabilities of different protein conformational states.
Collapse
Affiliation(s)
- Ronald J. Clarke
- School of Chemistry, University of Sydney, Sydney, NSW 2006 Australia
- The University of Sydney Nano Institute, Sydney, NSW 2006 Australia
| |
Collapse
|
28
|
Courtney KC, Mandal T, Mehta N, Wu L, Li Y, Das D, Cui Q, Chapman ER. Synaptotagmin-7 outperforms synaptotagmin-1 to promote the formation of large, stable fusion pores via robust membrane penetration. Nat Commun 2023; 14:7761. [PMID: 38012142 PMCID: PMC10681989 DOI: 10.1038/s41467-023-42497-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 10/11/2023] [Indexed: 11/29/2023] Open
Abstract
Synaptotagmin-1 and synaptotagmin-7 are two prominent calcium sensors that regulate exocytosis in neuronal and neuroendocrine cells. Upon binding calcium, both proteins partially penetrate lipid bilayers that bear anionic phospholipids, but the specific underlying mechanisms that enable them to trigger exocytosis remain controversial. Here, we examine the biophysical properties of these two synaptotagmin isoforms and compare their interactions with phospholipid membranes. We discover that synaptotagmin-1-membrane interactions are greatly influenced by membrane order; tight packing of phosphatidylserine inhibits binding due to impaired membrane penetration. In contrast, synaptotagmin-7 exhibits robust membrane binding and penetration activity regardless of phospholipid acyl chain structure. Thus, synaptotagmin-7 is a super-penetrator. We exploit these observations to specifically isolate and examine the role of membrane penetration in synaptotagmin function. Using nanodisc-black lipid membrane electrophysiology, we demonstrate that membrane penetration is a critical component that underlies how synaptotagmin proteins regulate reconstituted, exocytic fusion pores in response to calcium.
Collapse
Affiliation(s)
- Kevin C Courtney
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Taraknath Mandal
- Department of Chemistry, Boston University, Boston, MA, 02215, USA
- Department of Physics, Indian Institute of Technology - Kanpur, Kanpur, 208016, India
| | - Nikunj Mehta
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Lanxi Wu
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Yueqi Li
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
- Center for Bioanalytical Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Debasis Das
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Navy Nagar, Colaba, Mumbai, 400005, India
| | - Qiang Cui
- Department of Chemistry, Boston University, Boston, MA, 02215, USA
| | - Edwin R Chapman
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA.
| |
Collapse
|
29
|
Kim H, Kim HT, Jung SH, Han JW, Jo S, Kim IG, Kim RK, Kahm YJ, Choi TI, Kim CH, Lee JH. A Novel Anticancer Peptide Derived from Bryopsis plumosa Regulates Proliferation and Invasion in Non-Small Cell Lung Cancer Cells. Mar Drugs 2023; 21:607. [PMID: 38132928 PMCID: PMC10744475 DOI: 10.3390/md21120607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
The discovery of new highly effective anticancer drugs with few side effects is a challenge for drug development research. Natural or synthetic anticancer peptides (ACPs) represent a new generation of anticancer agents with high selectivity and specificity. The rapid emergence of chemoradiation-resistant lung cancer has necessitated the discovery of novel anticancer agents as alternatives to conventional therapeutics. In this study, we synthesized a peptide containing 22 amino acids and characterized it as a novel ACP (MP06) derived from green sea algae, Bryopsis plumosa. Using the ACP database, MP06 was predicted to possess an alpha-helical secondary structure and functionality. The anti-proliferative and apoptotic effects of the MP06, determined using the cytotoxicity assay and Annexin V/propidium iodide staining kit, were significantly higher in non-small-cell lung cancer (NSCLC) cells than in non-cancerous lung cells. We confirmed that MP06 suppressed cellular migration and invasion and inhibited the expression of N-cadherin and vimentin, the markers of epithelial-mesenchymal transition. Moreover, MP06 effectively reduced the metastasis of tumor xenografts in zebrafish embryos. In conclusion, we suggest considering MP06 as a novel candidate for the development of new anticancer drugs functioning via the ERK signaling pathway.
Collapse
Affiliation(s)
- Heabin Kim
- Department of Genetic Resources, National Marine Biodiversity Institute of Korea, Seocheon 33662, Republic of Korea; (H.K.); (S.-H.J.); (J.W.H.); (S.J.)
| | - Hyun-Taek Kim
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Republic of Korea;
| | - Seung-Hyun Jung
- Department of Genetic Resources, National Marine Biodiversity Institute of Korea, Seocheon 33662, Republic of Korea; (H.K.); (S.-H.J.); (J.W.H.); (S.J.)
| | - Jong Won Han
- Department of Genetic Resources, National Marine Biodiversity Institute of Korea, Seocheon 33662, Republic of Korea; (H.K.); (S.-H.J.); (J.W.H.); (S.J.)
| | - Seonmi Jo
- Department of Genetic Resources, National Marine Biodiversity Institute of Korea, Seocheon 33662, Republic of Korea; (H.K.); (S.-H.J.); (J.W.H.); (S.J.)
| | - In-Gyu Kim
- Department of Radiation Biology, Environmental Safety Assessment Research Division, Korea Atomic Energy Research Institute, Daejeon 34057, Republic of Korea; (I.-G.K.); (R.-K.K.); (Y.-J.K.)
| | - Rae-Kwon Kim
- Department of Radiation Biology, Environmental Safety Assessment Research Division, Korea Atomic Energy Research Institute, Daejeon 34057, Republic of Korea; (I.-G.K.); (R.-K.K.); (Y.-J.K.)
- Department of Radiation Science and Technology, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Yeon-Jee Kahm
- Department of Radiation Biology, Environmental Safety Assessment Research Division, Korea Atomic Energy Research Institute, Daejeon 34057, Republic of Korea; (I.-G.K.); (R.-K.K.); (Y.-J.K.)
- Department of Radiation Science and Technology, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Tae-Ik Choi
- Department of Biology, Chungnam National University, Yuseong-gu, Daejeon 34134, Republic of Korea; (T.-I.C.); (C.-H.K.)
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Yuseong-gu, Daejeon 34134, Republic of Korea; (T.-I.C.); (C.-H.K.)
| | - Jei Ha Lee
- Department of Genetic Resources, National Marine Biodiversity Institute of Korea, Seocheon 33662, Republic of Korea; (H.K.); (S.-H.J.); (J.W.H.); (S.J.)
| |
Collapse
|
30
|
Bui Thi Phuong H, Le Uyen C, Doan Ngan H, Luong Xuan H. Impact of chemical modifications on the antimicrobial and hemolytic activity of helical amphipathic peptide Lasioglossin LL-III. Amino Acids 2023; 55:1531-1544. [PMID: 37737904 DOI: 10.1007/s00726-023-03326-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 08/29/2023] [Indexed: 09/23/2023]
Abstract
Insect venom is abundant in potential antimicrobial peptides (AMPs), which can serve as novel alternatives to conventional antibiotics. Among them, Lasioglossin III LL-III) is a promising candidate with a broad spectrum against many fungi strains and both types of bacteria, whereas almost non-toxic to red blood cells. Many chemical approaches have been recently applied to improve its pharmacological properties and provide useful information regarding structure-activity relationships. Hence, this review focused on highlighting the lesson learned from each modification and supporting the future design of potent, selective, and metabolically stable AMPs.
Collapse
Affiliation(s)
| | - Chi Le Uyen
- Faculty of Pharmacy, Phenikaa University, Hanoi, 12116, Vietnam
| | - Hoa Doan Ngan
- Faculty of Medical Technology, Phenikaa University, Hanoi, 12116, Vietnam
| | - Huy Luong Xuan
- Faculty of Pharmacy, Phenikaa University, Hanoi, 12116, Vietnam.
- Phenikaa Institute for Advanced Study (PIAS), Phenikaa University, Hanoi, 12116, Vietnam.
| |
Collapse
|
31
|
Dowell JA, Mason C. Candidate pathway association and genome-wide association approaches reveal alternative genetic architectures of carotenoid content in cultivated sunflower ( Helianthus annuus). APPLICATIONS IN PLANT SCIENCES 2023; 11:e11558. [PMID: 38106540 PMCID: PMC10719882 DOI: 10.1002/aps3.11558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 05/10/2023] [Accepted: 05/19/2023] [Indexed: 12/19/2023]
Abstract
Premise The explosion of available genomic data poses significant opportunities and challenges for genome-wide association studies. Current approaches via linear mixed models (LMM) are straightforward but prevent flexible assumptions of an a priori genomic architecture, while Bayesian sparse LMMs (BSLMMs) allow this flexibility. Complex traits, such as specialized metabolites, are subject to various hierarchical effects, including gene regulation, enzyme efficiency, and the availability of reactants. Methods To identify alternative genetic architectures, we examined the genetic architecture underlying the carotenoid content of an association mapping panel of Helianthus annuus individuals using multiple BSLMM and LMM frameworks. Results The LMMs of genome-wide single-nucleotide polymorphisms (SNPs) identified a single transcription factor responsible for the observed variations in the carotenoid content; however, a BSLMM of the SNPs with the bottom 1% of effect sizes from the results of the LMM identified multiple biologically relevant quantitative trait loci (QTLs) for carotenoid content external to the known (annotated) carotenoid pathway. A candidate pathway analysis (CPA) suggested a β-carotene isomerase to be the enzyme with the highest impact on the observed carotenoid content within the carotenoid pathway. Discussion While traditional LMM approaches suggested a single unknown transcription factor associated with carotenoid content variation in sunflower petals, BSLMM proposed several QTLs with interpretable biological relevance to this trait. In addition, the CPA allowed for the dissection of the regulatory vs. biosynthetic genetic architectures underlying this metabolic trait.
Collapse
Affiliation(s)
- Jordan A. Dowell
- Department of Plant SciencesUniversity of CaliforniaDavisCalifornia95616USA
- Present address:
Department of Biological SciencesLouisiana State UniversityBaton RougeLouisiana70803USA
| | - Chase Mason
- Department of BiologyUniversity of Central FloridaOrlandoFlorida32816USA
| |
Collapse
|
32
|
Xing H, Loya-Perez V, Franzen J, Denton PW, Conda-Sheridan M, Rodrigues de Almeida N. Designing peptide amphiphiles as novel antibacterials and antibiotic adjuvants against gram-negative bacteria. Bioorg Med Chem 2023; 94:117481. [PMID: 37776750 DOI: 10.1016/j.bmc.2023.117481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 10/02/2023]
Abstract
Gram-negative strains are intrinsically resistant to most antibiotics due to the robust and impermeable characteristic of their outer membrane. Self-assembling cationic peptide amphiphiles (PAs) have the ability to disrupt bacteria membranes, constituting an excellent antibacterial alternative to small molecule drugs that can be used alone or as antibiotic adjuvants to overcome bacteria resistance. PA1 (C16KHKHK), self-assembled into micelles, which exhibited low antibacterial activity against all strains tested, and showed strong synergistic antibacterial activity in combination with Vancomycin with a Fractional Inhibitory Concentration index (FICi) of 0.15 against E. coli. The molecules, PA2 (C16KRKR) and PA3 (C16AAAKRKR), also self-assembled into micelles, displayed a broad-spectrum antibacterial activity against all strains tested, and low susceptibility to resistance development over 21 days. Finally, PA1, PA 2 and PA3 displayed low cytotoxicity against mammalian cells, and PA2 showed a potent antibacterial activity and low toxicity in preliminary in vivo models using G. mellonella. The results show that PAs are a great platform for the future development of effective antibiotics to slow down the antibiotic resistance and can act as antibiotic adjuvants with synergistic mechanism of action, which can be repurposed for use with existing antibiotics commonly used to treat gram-positive bacteria to treat infections caused by gram-negative bacteria.
Collapse
Affiliation(s)
- Huihua Xing
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Vanessa Loya-Perez
- Department of Chemistry, University of Nebraska Omaha, Omaha, NE 68182, United States
| | - Joshua Franzen
- Department of Biology, University of Nebraska Omaha, Omaha, NE 68182, United States
| | - Paul W Denton
- Department of Biology, University of Nebraska Omaha, Omaha, NE 68182, United States
| | - Martin Conda-Sheridan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | | |
Collapse
|
33
|
Wu Y, Hu J, Du Y, Lu G, Li Y, Feng Y, Chen L, Tu Y, Xiang M, Gui Y, Shu T, Yu L. Mechanistic Insights into the Halophilic Xylosidase Xylo-1 and Its Role in Xylose Production. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:15375-15387. [PMID: 37773011 DOI: 10.1021/acs.jafc.3c05045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
The Xylo-1 xylosidase, which belongs to the GH43 family, exhibits a high salt tolerance. The present study demonstrated that the catalytic activity of Xylo-1 increased by 195% in the presence of 5 M NaCl. Additionally, the half-life of Xylo-1 increased 25.9-fold in the presence of 1 M NaCl. Through comprehensive analysis including circular dichroism, fluorescence spectroscopy, and molecular dynamics simulations, we elucidated that the presence of Na+ ions increased the contact frequency between the surface acidic amino acids and the surrounding water molecules. This resulted in the stabilization of the surrounding hydration layer of Xylo-1. Additionally, Na+ ions also stabilized the substrate-binding conformation and the fluctuation of water molecules within the active site, which enhanced the catalytic activity of Xylo-1 by increasing the nucleophilic attack by the water molecules. Ultimately, the optimal reaction conditions for the production of xylose by synergistic catalysis with Xylo-1 and xylanase were determined. The results demonstrated that the conversion yield of the method was high for various sources of xylan, indicating the method could have potential industrial applications. This study explored the structure-activity relationship of catalysis in Xylo-1 under high-salt conditions, provides novel insights into the mechanism of halophilic enzymes, and serves as a reference for the industrial application of Xylo-1.
Collapse
Affiliation(s)
- Ya Wu
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics, Ministry of Education, 1037 Luoyu Road, Wuhan 430074, China
| | - Jiayue Hu
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China
| | - Yikai Du
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China
| | - Gen Lu
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics, Ministry of Education, 1037 Luoyu Road, Wuhan 430074, China
| | - Yingnan Li
- Ministry of Education Key Laboratory of Industrial Biotechnology, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Yujia Feng
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China
| | - Liting Chen
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China
| | - Yuhao Tu
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China
| | - Mengxiong Xiang
- Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, 28 Nanli Road, Wuhan 430068, China
| | - Yifan Gui
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics, Ministry of Education, 1037 Luoyu Road, Wuhan 430074, China
| | - Tong Shu
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics, Ministry of Education, 1037 Luoyu Road, Wuhan 430074, China
| | - Longjiang Yu
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics, Ministry of Education, 1037 Luoyu Road, Wuhan 430074, China
| |
Collapse
|
34
|
Sweany RR, Cary JW, Jaynes JM, Rajasekaran K. Broad-Spectrum Antimicrobial Activity of Synthetic Peptides GV185 and GV187. PLANT DISEASE 2023; 107:3211-3221. [PMID: 36947838 DOI: 10.1094/pdis-11-22-2572-re] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Optimizing synthetic antimicrobial peptides for safe and enhanced activity against fungal and bacterial pathogens is useful for genetic engineering of plants for resistance to plant pathogens and their associated mycotoxins. Nine synthetic peptides modeled after lytic peptides tachyplesin 1, D4E1 from cecropin A, and protegrin 1 were added to germinated spores of fungal species Aspergillus flavus, Rhizopus stolonifer, Fusarium oxysporum f. sp. vasinfectum, F. verticillioides, F. graminearum, Claviceps purpurea, Verticillium dahliae, and Thielaviopsis basicola and bacterial cultures of Pseudomonas syringae pv. tabaci and Xanthomonas campestris pv. campestris at different doses and inhibitory dose response curves, and were modeled to assess antimicrobial activity. Peptides GV185 and GV187, modified from tachyplesin 1, had superior abilities to inhibit fungal and bacterial growth (50% inhibitory concentrations [IC50] ranging from 0.1 to 8.7 µM). R. stolonifer (IC50 = 8.1 µM), A. flavus (IC50 = 3.1 µM), and F. graminearum (IC50 = 2.2 µM) were less inhibited by GV185 and GV187 than all the remaining fungi (IC50 = 1.4 µM) and bacteria (IC50 = 0.1 µM). Of the remaining peptides, GV193, GV195, and GV196 (IC50 range of 0.9 to 6.6 µM) inhibited fungal growth of A. flavus, F. verticillioides, and F. graminearum less than GV185 and GV187 (IC50 range of 0.8 to 3.9 µM), followed by GV197 (IC50 range of 0.8 to 9.1 µM), whereas GV190 and GV192 inhibited poorly (IC50 range of 28.2 to 36.6 µM and 15.5 to 19.4 µM, respectively) and GV198 stimulated growth. GV185 and GV187 had slightly weaker hydrophobic and cationic residues than other tachyplesin 1 modified peptides but still had unexpectedly high lytic activity. Germinated fungal spores of R. stolonifer and F. graminearum exposed to these two peptides and D4E1 and AGM182 appeared wrinkled, with perforations near potential cytoplasmic leakage, which provided evidence of plasma membrane and cell wall lysis. We conclude that peptides GV185 and GV187 are promising candidates for genetic engineering of crops for resistance to plant-pathogenic bacteria and fungi, including A. flavus and aflatoxin contamination.
Collapse
Affiliation(s)
- Rebecca R Sweany
- Food and Feed Safety Research Unit, Southern Regional Research Center, United States Department of Agriculture-Agricultural Research Service, New Orleans, LA 70124
| | - Jeffrey W Cary
- Food and Feed Safety Research Unit, Southern Regional Research Center, United States Department of Agriculture-Agricultural Research Service, New Orleans, LA 70124
| | - Jesse M Jaynes
- Genvor, LLC, Dallas, TX 75240
- College of Agriculture, Environment and Nutrition Sciences, and College of Arts and Sciences, Tuskegee University, Tuskegee, AL 36088
| | - Kanniah Rajasekaran
- Food and Feed Safety Research Unit, Southern Regional Research Center, United States Department of Agriculture-Agricultural Research Service, New Orleans, LA 70124
| |
Collapse
|
35
|
Ghosh S, Chatterjee S, Satpati P. Effect of Spacer Length Modification of the Cationic Side Chain on the Energetics of Antimicrobial Peptide Binding to Membrane-Mimetic Bilayers. J Chem Inf Model 2023; 63:5823-5833. [PMID: 37684221 DOI: 10.1021/acs.jcim.3c01080] [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: 09/10/2023]
Abstract
Understanding the mechanism of action of the antimicrobial peptide (AMP) in terms of its structure and energetics is the key to designing new potent and selective AMPs. Recently, we reported a membranolytic 14-residue-long lysine-rich cationic antimicrobial peptide (LL-14: NH3+-LKWLKKLLKWLKKL-CONH2) against Pseudomonas aeruginosa, Klebsiella pneumoniae, and Staphylococcus aureus, which is limited by cytotoxicity and expected to undergo facile protease degradation. Aliphatic side-chain-length modification of the cationic amino-acid residues (Lys and Arg) is a popular strategy for designing protease-resistant AMPs. However, the effect of the peptide side-chain length modifications on the membrane binding affinity and its relation to the atomic structure remain an unsolved problem. We report computer simulations that quantitatively calculated the difference in peptide binding affinity to membrane-mimetic-bilayer models (bacterial: 1-palmitoyl-2-oleoyl-phosphatidylethanolamine (POPE)/1-palmitoyl-2-oleoyl-phosphatidylglycerol (POPG) bilayer and mammalian: 1-palmitoyl-2-oleoyl-phosphatidylcholine (POPC) bilayer) upon decreasing or increasing the spacer length of the cationic lysine residues of LL-14 (as well as their arginine analogues). We show that the peptide/bilayer interaction energetics varies drastically in response to spacer length modification. The strength of peptide discrimination depends strongly on the nature of the bilayer (bacterial or mammalian mimetic model). An increase in the lysine spacer length by one carbon (i.e., homolysine analogue of LL-14) is weakly/strongly disfavored by the bacterial/mammalian-membrane-mimetic bilayer. Recently, we have demonstrated an excellent correlation between the antimicrobial activity of the membranolytic cationic peptides and their binding affinity to membrane-mimetic-bilayer models. Thus, the homolysine analogue of LL-14 is a promising noncytotoxic AMP with conserved activity. On the other hand, homoarginine analogue (arginine spacer length increment by a single carbon) was preferred by both the bacteria and the mammalian mimetic bilayers and displayed the strongest affinity for the former among the peptides studied in this work. Thus, the promising most potent homoarginine analogue is likely to be cytotoxic. Shortening the Lys/Arg side chain to a three-carbon spacer (Dab/Agb) improves the binding affinity to bacterial and mammalian-membrane-mimetic bilayers. Arginine and arginine-derivative peptides exhibited stronger binding affinity to the bilayers relative to the lysine analogue. The results provide a plausible explanation to the previous experimental observations, viz., superior antimicrobial activity of the arginine peptides relative to Lys peptides and the improvement of antimicrobial activity upon substitution of Lys with Dab in the cationic peptides. The simulations revealed that the small change in the peptide hydrophobicity by Lys/Arg spacer length modification could drastically alter the energetics of peptide/bilayer binding by fine-tuning the electrostatic interactions. The energetics underlying the peptide selectivity by simple membrane-mimetic bilayer models may be beneficial for designing new selective and protease-resistant AMPs.
Collapse
Affiliation(s)
- Suvankar Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Sunanda Chatterjee
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Priyadarshi Satpati
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| |
Collapse
|
36
|
Yao A, Ma Y, Sun R, Zou W, Chen X, Zhou M, Ma C, Chen T, Shaw C, Wang L. A Designed Analog of an Antimicrobial Peptide, Crabrolin, Exhibits Enhanced Anti-Proliferative and In Vivo Antimicrobial Activity. Int J Mol Sci 2023; 24:14472. [PMID: 37833918 PMCID: PMC10572522 DOI: 10.3390/ijms241914472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/07/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
Antimicrobial peptides have gradually attracted interest as promising alternatives to conventional agents to control the worldwide health threats posed by antibiotic resistance and cancer. Crabrolin is a tridecapeptide extracted from the venom of the European hornet (Vespa crabro). Its antibacterial and anticancer potentials have been underrated compared to other peptides discovered from natural resources. Herein, a series of analogs were designed based on the template sequence of crabrolin to study its structure-activity relationship and enhance the drug's potential by changing the number, type, and distribution of charged residues. The cationicity-enhanced derivatives were shown to have improved antibacterial and anticancer activities with a lower toxicity. Notably, the double-arginine-modified product, crabrolin-TR, possessed a potent capacity against Pseudomonas aeruginosa (minimum inhibitory concentration (MIC) = 4 μM), which was around thirty times stronger than the parent peptide (MIC = 128 μM). Furthermore, crabrolin-TR showed an in vivo treatment efficacy in a Klebsiella-pneumoniae-infected waxworm model and was non-toxic under its maximum MBC value (MIC = 8 μM), indicating its therapeutic potency and better selectivity. Overall, we rationally designed functional peptides by progressively increasing the number and distribution of charged residues, demonstrating new insights for developing therapeutic molecules from natural resources with enhanced properties, and proposed crabrolin-TR as an appealing antibacterial and anticancer agent candidate for development.
Collapse
Affiliation(s)
- Aifang Yao
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (Y.M.); (R.S.); (X.C.); (M.Z.); (C.M.); (T.C.); (C.S.); (L.W.)
| | - Yingxue Ma
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (Y.M.); (R.S.); (X.C.); (M.Z.); (C.M.); (T.C.); (C.S.); (L.W.)
| | - Ruize Sun
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (Y.M.); (R.S.); (X.C.); (M.Z.); (C.M.); (T.C.); (C.S.); (L.W.)
| | - Wanchen Zou
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (Y.M.); (R.S.); (X.C.); (M.Z.); (C.M.); (T.C.); (C.S.); (L.W.)
| | - Xiaoling Chen
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (Y.M.); (R.S.); (X.C.); (M.Z.); (C.M.); (T.C.); (C.S.); (L.W.)
| | - Mei Zhou
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (Y.M.); (R.S.); (X.C.); (M.Z.); (C.M.); (T.C.); (C.S.); (L.W.)
| | - Chengbang Ma
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (Y.M.); (R.S.); (X.C.); (M.Z.); (C.M.); (T.C.); (C.S.); (L.W.)
| | - Tianbao Chen
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (Y.M.); (R.S.); (X.C.); (M.Z.); (C.M.); (T.C.); (C.S.); (L.W.)
| | - Chris Shaw
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (Y.M.); (R.S.); (X.C.); (M.Z.); (C.M.); (T.C.); (C.S.); (L.W.)
| | - Lei Wang
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (Y.M.); (R.S.); (X.C.); (M.Z.); (C.M.); (T.C.); (C.S.); (L.W.)
| |
Collapse
|
37
|
de Sousa FFO, Pinazo A, Hafidi Z, García MT, Bautista E, Moran MDC, Pérez L. Arginine Gemini-Based Surfactants for Antimicrobial and Antibiofilm Applications: Molecular Interactions, Skin-Related Anti-Enzymatic Activity and Cytotoxicity. Molecules 2023; 28:6570. [PMID: 37764346 PMCID: PMC10536132 DOI: 10.3390/molecules28186570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The antimicrobial and antibiofilm properties of arginine-based surfactants have been evaluated. These two biological properties depend on both the alkyl chain length and the spacer chain nature. These gemini surfactants exhibit good activity against a wide range of bacteria, including some problematic resistant microorganisms such us methicillin-resistant Staphylococcus aureus (MRSA) and Pseudomonas aeruginosa. Moreover, surfactants with a C10 alkyl chain and C3 spacer inhibit the (MRSA) and Pseudomonas aeruginosa biofilm formation at concentrations as low as 8 µg/mL and are able to eradicate established biofilms of these two bacteria at 32 µg/mL. The inhibitory activities of the surfactants over key enzymes enrolled in the skin repairing processes (collagenase, elastase and hyaluronidase) were evaluated. They exhibited moderate anti-collagenase activity while the activity of hyaluronidase was boosted by the presence of these surfactants. These biological properties render these gemini arginine-based surfactants as perfect promising candidates for pharmaceutical and biological properties.
Collapse
Affiliation(s)
- Francisco Fábio Oliveira de Sousa
- Laboratory of Quality Control, Bromatology & Microbiology, Department of Biological & Health Sciences, School of Pharmacy, Federal University of Amapá, Rodovia Juscelino Kubitscheck, km 02, Macapá 68903-419, Brazil
| | - Aurora Pinazo
- Department of Surfactants and Nanobiotechnology, Instituto de Química Avanzada de Cataluña, Centro Superior de Investigaciones Científicas IQAC-CSIC, 08035 Barcelona, Spain; (A.P.); (Z.H.); (M.T.G.); (E.B.)
| | - Zakaria Hafidi
- Department of Surfactants and Nanobiotechnology, Instituto de Química Avanzada de Cataluña, Centro Superior de Investigaciones Científicas IQAC-CSIC, 08035 Barcelona, Spain; (A.P.); (Z.H.); (M.T.G.); (E.B.)
| | - María Teresa García
- Department of Surfactants and Nanobiotechnology, Instituto de Química Avanzada de Cataluña, Centro Superior de Investigaciones Científicas IQAC-CSIC, 08035 Barcelona, Spain; (A.P.); (Z.H.); (M.T.G.); (E.B.)
| | - Elena Bautista
- Department of Surfactants and Nanobiotechnology, Instituto de Química Avanzada de Cataluña, Centro Superior de Investigaciones Científicas IQAC-CSIC, 08035 Barcelona, Spain; (A.P.); (Z.H.); (M.T.G.); (E.B.)
| | - Maria del Carmen Moran
- Secció de Fisiologia, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, Avda. Joan XXIII 27-31, 08028 Barcelona, Spain;
- Institut de Nanociència i Nanotecnologia—IN2UB, Universitat de Barcelona, Avda. Diagonal 645, 08028 Barcelona, Spain
| | - Lourdes Pérez
- Department of Surfactants and Nanobiotechnology, Instituto de Química Avanzada de Cataluña, Centro Superior de Investigaciones Científicas IQAC-CSIC, 08035 Barcelona, Spain; (A.P.); (Z.H.); (M.T.G.); (E.B.)
| |
Collapse
|
38
|
Machado SK, Peil H, Kraushaar T, Claar P, Mischnik M, Lind H, Herzog E, Bacher M, Nolte MW, Bielohuby M, Pestel S, Ponnuswamy P. Modulation of Extravascular Binding of Recombinant Factor IX Impacts the Duration of Efficacy in Mouse Models. Thromb Haemost 2023; 123:751-762. [PMID: 37164314 PMCID: PMC10365885 DOI: 10.1055/a-2090-9739] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 01/11/2023] [Indexed: 05/12/2023]
Abstract
BACKGROUND There is an emerging concept that in addition to circulating coagulation factor IX (FIX), extravascular FIX contributes to hemostasis. OBJECTIVE Our objective was to evaluate the efficacy of extravascular FIX using animal models of tail clip bleeding and ferric chloride-induced thrombosis. METHODS Mutant rFIX proteins with described enhanced (rFIXK5R) or reduced (rFIXK5A) binding to extracellular matrix were generated and characterized using in vitro aPTT, one-stage clotting, and modified FX assays. Using hemophilia B mice, pharmacokinetic (PK) parameters and in vivo efficacy of these proteins were compared against rFIX wild-type protein (rFIXWT) in a tail clip bleeding and FeCl3-induced thrombosis model. Respective tissue disposition of FIX was evaluated using immunofluorescence. RESULTS In vitro characterization demonstrated comparable clotting activity of rFIX proteins. The PK profile showed that rFIXK5A displayed the highest plasma exposure compared to rFIXWT and rFIXK5R. Immunofluorescence evaluation of liver tissue showed that rFIXK5R was detectable up to 24 hours, whereas rFIXWT and rFIXK5A were detectable only up to 15 minutes. In the tail clip bleeding model, rFIXK5R displayed significant hemostatic protection against bleeding incidence for up to 72 hours postintravenous administration of 50 IU/kg, whereas the efficacy of rFIXK5A was already reduced at 24 hours. Similarly, in the mesenteric artery thrombus model, rFIXK5R and rFIXWT demonstrated prolonged efficacy compared to rFIXK5A. CONCLUSION Using two different in vivo models of hemostasis and thrombosis, we demonstrate that mutated rFIX protein with enhanced binding (rFIXK5R) to extravascular space confers prolonged hemostatic efficacy in vivo despite lower plasma exposure, whereas rFIXK5A rapidly lost its efficacy despite higher plasma exposure.
Collapse
Affiliation(s)
| | | | | | | | | | - Holger Lind
- CSL Behring Innovation GmbH, Marburg, Germany
| | - Eva Herzog
- CSL Behring LLC, King of Prussia, Pennsylvania, United States
| | - Michael Bacher
- Institute of Immunology—Philipps University Marburg, Marburg, Germany
| | | | | | | | | |
Collapse
|
39
|
Pašalić L, Jakas A, Pem B, Bakarić D. Adsorption/Desorption of Cationic-Hydrophobic Peptides on Zwitterionic Lipid Bilayer Is Associated with the Possibility of Proton Transfer. Antibiotics (Basel) 2023; 12:1216. [PMID: 37508312 PMCID: PMC10376034 DOI: 10.3390/antibiotics12071216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Cell-penetrating peptides (CPPs) are short peptides built up from dominantly cationic and hydrophobic amino acid residues with a distinguished ability to pass through the cell membrane. Due to the possibility of linking and delivering the appropriate cargo at the desired location, CPPs are considered an economic and less invasive alternative to antibiotics. Besides knowing that their membrane passage mechanism is a complex function of CPP chemical composition, the ionic strength of the solution, and the membrane composition, all other details on how they penetrate cell membranes are rather vague. The aim of this study is to elucidate the ad(de)sorption of arginine-/lysine- and phenylalanine-rich peptides on a lipid membrane composed of 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) lipids. DSC and temperature-dependent UV-Vis measurements confirmed the impact of the adsorbed peptides on thermotropic properties of DPPC, but in an inconclusive way. On the other hand, FTIR spectra acquired at 30 °C and 50 °C (when DPPC lipids are found in the gel and fluid phase, respectively) unambiguously confirmed the proton transfer between particular titratable functional groups of R5F2/K5F2 that highly depend on their immediate surroundings (DPPC or a phosphate buffer). Molecular dynamic simulations showed that both peptides may adsorb onto the bilayer, but K5F2 desorbs more easily and favors the solvent, while R5F2 remains attached. The results obtained in this work highlight the importance of proton transfer in the design of CPPs with their desired cargo, as its charge and composition dictates the possibility of entering the cell.
Collapse
Affiliation(s)
- Lea Pašalić
- Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Bijenička Cesta 54, 10000 Zagreb, Croatia
| | - Andreja Jakas
- Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Bijenička Cesta 54, 10000 Zagreb, Croatia
| | - Barbara Pem
- Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Bijenička Cesta 54, 10000 Zagreb, Croatia
| | - Danijela Bakarić
- Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Bijenička Cesta 54, 10000 Zagreb, Croatia
| |
Collapse
|
40
|
Jakubec M, Rylandsholm FG, Rainsford P, Silk M, Bril'kov M, Kristoffersen T, Juskewitz E, Ericson JU, Svendsen JSM. Goldilocks Dilemma: LPS Works Both as the Initial Target and a Barrier for the Antimicrobial Action of Cationic AMPs on E. coli. Biomolecules 2023; 13:1155. [PMID: 37509189 PMCID: PMC10377513 DOI: 10.3390/biom13071155] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Antimicrobial peptides (AMPs) are generally membrane-active compounds that physically disrupt bacterial membranes. Despite extensive research, the precise mode of action of AMPs is still a topic of great debate. This work demonstrates that the initial interaction between the Gram-negative E. coli and AMPs is driven by lipopolysaccharides (LPS) that act as kinetic barriers for the binding of AMPs to the bacterial membrane. A combination of SPR and NMR experiments provide evidence suggesting that cationic AMPs first bind to the negatively charged LPS before reaching a binding place in the lipid bilayer. In the event that the initial LPS-binding is too strong (corresponding to a low dissociation rate), the cationic AMPs cannot effectively get from the LPS to the membrane, and their antimicrobial potency will thus be diminished. On the other hand, the AMPs must also be able to effectively interact with the membrane to exert its activity. The ability of the studied cyclic hexapeptides to bind LPS and to translocate into a lipid membrane is related to the nature of the cationic charge (arginine vs. lysine) and to the distribution of hydrophobicity along the molecule (alternating vs. clumped tryptophan).
Collapse
Affiliation(s)
- Martin Jakubec
- Department of Chemistry, Faculty of Science and Technology, UiT the Arctic University of Norway, 9019 Tromsø, Norway
| | - Fredrik G Rylandsholm
- Department of Chemistry, Faculty of Science and Technology, UiT the Arctic University of Norway, 9019 Tromsø, Norway
| | - Philip Rainsford
- Department of Chemistry, Faculty of Science and Technology, UiT the Arctic University of Norway, 9019 Tromsø, Norway
| | - Mitchell Silk
- Department of Chemistry, Faculty of Science and Technology, UiT the Arctic University of Norway, 9019 Tromsø, Norway
| | - Maxim Bril'kov
- Department of Pharmacy, Faculty of Health Sciences, UiT the Arctic University of Norway, 9019 Tromsø, Norway
| | - Tone Kristoffersen
- Department of Chemistry, Faculty of Science and Technology, UiT the Arctic University of Norway, 9019 Tromsø, Norway
| | - Eric Juskewitz
- Department of Medical Biology, Faculty of Health Sciences, UiT the Arctic University of Norway, 9019 Tromsø, Norway
| | - Johanna U Ericson
- Department of Medical Biology, Faculty of Health Sciences, UiT the Arctic University of Norway, 9019 Tromsø, Norway
| | - John Sigurd M Svendsen
- Department of Chemistry, Faculty of Science and Technology, UiT the Arctic University of Norway, 9019 Tromsø, Norway
| |
Collapse
|
41
|
Motamedi H, Alvandi A, Fathollahi M, Ari MM, Moradi S, Moradi J, Abiri R. In silico designing and immunoinformatics analysis of a novel peptide vaccine against metallo-beta-lactamase (VIM and IMP) variants. PLoS One 2023; 18:e0275237. [PMID: 37471423 PMCID: PMC10358925 DOI: 10.1371/journal.pone.0275237] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 09/12/2022] [Indexed: 07/22/2023] Open
Abstract
The rapid spread of acquired metallo-beta-lactamases (MBLs) among gram negative pathogens is becoming a global concern. Improper use of broad-spectrum antibiotics can trigger the colonization and spread of resistant strains which lead to increased mortality and significant economic loss. In the present study, diverse immunoinformatic approaches are applied to design a potential epitope-based vaccine against VIM and IMP MBLs. The amino acid sequences of VIM and IMP variants were retrieved from the GenBank database. ABCpred and BCPred online Web servers were used to analyze linear B cell epitopes, while IEDB was used to determine the dominant T cell epitopes. Sequence validation, allergenicity, toxicity and physiochemical analysis were performed using web servers. Seven sequences were identified for linear B cell dominant epitopes and 4 sequences were considered as dominant CD4+ T cell epitopes, and the predicted epitopes were joined by KK and GPGPG linkers. Stabilized multi-epitope protein structure was obtained using molecular dynamics simulation. Molecular docking showed that the designed vaccine exhibited sustainable and strong binding interactions with Toll-like receptor 4 (TLR4). Finally, codon adaptation and in silico cloning studies were performed to design an effective vaccine production strategy. Immune simulation significantly provided high levels of immunoglobulins, T helper cells, T-cytotoxic cells and INF-γ. Even though the introduced vaccine candidate demonstrates a very potent immunogenic potential, but wet-lab validation is required to further assessment of the effectiveness of this proposed vaccine candidate.
Collapse
Affiliation(s)
- Hamid Motamedi
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Student Research Committee, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amirhoushang Alvandi
- Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Matin Fathollahi
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Student Research Committee, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Marzie Mahdizade Ari
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Sajad Moradi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Jale Moradi
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ramin Abiri
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
42
|
Boaro A, Ageitos L, Torres MDT, Blasco EB, Oztekin S, de la Fuente-Nunez C. Structure-function-guided design of synthetic peptides with anti-infective activity derived from wasp venom. CELL REPORTS. PHYSICAL SCIENCE 2023; 4:101459. [PMID: 38239869 PMCID: PMC10795512 DOI: 10.1016/j.xcrp.2023.101459] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2024]
Abstract
Antimicrobial peptides (AMPs) derived from natural toxins and venoms offer a promising alternative source of antibiotics. Here, through structure-function-guided design, we convert two natural AMPs derived from the venom of the solitary eumenine wasp Eumenes micado into α-helical AMPs with reduced toxicity that kill Gram-negative bacteria in vitro and in a preclinical mouse model. To identify the sequence determinants conferring antimicrobial activity, an alanine scan screen and strategic single lysine substitutions are made to the amino acid sequence of these natural peptides. These efforts yield a total of 34 synthetic derivatives, including alanine substituted and lysine-substituted sequences with stabilized α-helical structures and increased net positive charge. The resulting lead synthetic peptides kill the Gram-negative pathogens Escherichia coli and Pseudomonas aeruginosa (PAO1 and PA14) by rapidly permeabilizing both their outer and cytoplasmic membranes, exhibit anti-infective efficacy in a mouse model by reducing bacterial loads by up to three orders of magnitude, and do not readily select for bacterial resistance.
Collapse
Affiliation(s)
- Andreia Boaro
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Present address: Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, São Paulo 09210-580, Brazil
- These authors contributed equally
| | - Lucía Ageitos
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Present address: CICA - Centro Interdisciplinar de Química e Bioloxía, Departamento de Química, Facultade de Ciencias, Universidade da Coruña, 15008 A Coruña, Spain
- These authors contributed equally
| | - Marcelo Der Torossian Torres
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Esther Broset Blasco
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sebahat Oztekin
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Present address: Faculty of Engineering, Department of Food Engineering, Bayburt University, Bayburt 69000, Turkey
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lead contact
| |
Collapse
|
43
|
Povilaitis SC, Webb LJ. Leaflet-Dependent Effect of Anionic Lipids on Membrane Insertion by Cationic Cell-Penetrating Peptides. J Phys Chem Lett 2023; 14:5841-5849. [PMID: 37339513 PMCID: PMC10478718 DOI: 10.1021/acs.jpclett.3c00725] [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] [Indexed: 06/22/2023]
Abstract
Cationic membrane-permeating peptides can cross membranes unassisted by transmembrane protein machinery, and there is consensus that anionic lipids facilitate this process. Although membranes are asymmetric in lipid composition, investigations of the impact of anionic lipids on peptide-membrane insertion in model vesicles primarily use symmetric anionic lipid distributions between bilayer leaflets. Here, we investigate the leaflet-specific influence of three anionic lipid headgroups [phosphatidic acid (PA), phosphatidylserine (PS), and phosphatidylglycerol (PG)] on insertion into model membranes by three cationic membrane-permeating peptides (NAF-144-67, R6W3, and WWWK). We report that outer leaflet anionic lipids enhanced peptide-membrane insertion for all peptides while inner leaflet anionic lipids did not have a significant effect except in the case of NAF-144-67 incubated with PA-containing vesicles. The insertion enhancement was headgroup-dependent for arginine-containing peptides but not WWWK. These results provide significant new insight into the potential role of membrane asymmetry in insertion of peptides into model membranes.
Collapse
Affiliation(s)
- Sydney C Povilaitis
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Lauren J Webb
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
44
|
Legrand D, Herbaut M, Durin Z, Brysbaert G, Bardor M, Lensink MF, Foulquier F. New insights into the pathogenicity of TMEM165 variants using structural modeling based on AlphaFold 2 predictions. Comput Struct Biotechnol J 2023; 21:3424-3436. [PMID: 37416081 PMCID: PMC10319644 DOI: 10.1016/j.csbj.2023.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/15/2023] [Accepted: 06/15/2023] [Indexed: 07/08/2023] Open
Abstract
TMEM165 is a Golgi protein playing a crucial role in Mn2+ transport, and whose mutations in patients are known to cause Congenital Disorders of Glycosylation. Some of those mutations affect the highly-conserved consensus motifs E-φ-G-D-[KR]-[TS] characterizing the CaCA2/UPF0016 family, presumably important for the transport of Mn2+ which is essential for the function of many Golgi glycosylation enzymes. Others, like the G>R304 mutation, are far away from these motifs in the sequence. Until recently, the classical membrane protein topology prediction methods were unable to provide a clear picture of the organization of TMEM165 inside the cell membrane, or to explain in a convincing manner the impact of patient and experimentally-generated mutations on the transporter function of TMEM165. In this study, AlphaFold 2 was used to build a TMEM165 model that was then refined by molecular dynamics simulation with membrane lipids and water. This model provides a realistic picture of the 3D protein scaffold formed from a two-fold repeat of three transmembrane helices/domains where the consensus motifs face each other to form a putative acidic cation-binding site at the cytosolic side of the protein. It sheds new light on the impact of mutations on the transporter function of TMEM165, found in patients and studied experimentally in vitro, formerly and within this study. More particularly and very interestingly, this model explains the impact of the G>R304 mutation on TMEM165's function. These findings provide great confidence in the predicted TMEM165 model whose structural features are discussed in the study and compared to other structural and functional TMEM165 homologs from the CaCA2/UPF0016 family and the LysE superfamily.
Collapse
Affiliation(s)
- Dominique Legrand
- Univ. Lille, CNRS, UMR 8576 – UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Mélissandre Herbaut
- Univ. Lille, CNRS, UMR 8576 – UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Zoé Durin
- Univ. Lille, CNRS, UMR 8576 – UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Guillaume Brysbaert
- Univ. Lille, CNRS, UMR 8576 – UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Muriel Bardor
- Univ. Lille, CNRS, UMR 8576 – UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
- Université de Rouen Normandie, Laboratoire GlycoMEV UR 4358, SFR Normandie Végétal FED 4277, Innovation Chimie Carnot, F-76000 Rouen, France
| | - Marc F. Lensink
- Univ. Lille, CNRS, UMR 8576 – UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - François Foulquier
- Univ. Lille, CNRS, UMR 8576 – UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| |
Collapse
|
45
|
Kumar Mahato A, Pal S, Dey K, Reja A, Paul S, Shelke A, Ajithkumar TG, Das D, Banerjee R. Covalent Organic Framework Cladding on Peptide-Amphiphile-Based Biomimetic Catalysts. J Am Chem Soc 2023. [PMID: 37267597 DOI: 10.1021/jacs.3c03562] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Peptide-based biomimetic catalysts are promising materials for efficient catalytic activity in various biochemical transformations. However, their lack of operational stability and fragile nature in non-aqueous media limit their practical applications. In this study, we have developed a cladding technique to stabilize biomimetic catalysts within porous covalent organic framework (COF) scaffolds. This methodology allows for the homogeneous distribution of peptide nanotubes inside the COF (TpAzo and TpDPP) backbone, creating strong noncovalent interactions that prevent leaching. We synthesized two different peptide-amphiphiles, C10FFVK and C10FFVR, with lysine (K) and arginine (R) at the C-termini, respectively, which formed nanotubular morphologies. The C10FFVK peptide-amphiphile nanotubes exhibit enzyme-like behavior and efficiently catalyze C-C bond cleavage in a buffer medium (pH 7.5). We produced nanotubular structures of TpAzo-C10FFVK and TpDPP-C10FFVK through COF cladding by using interfacial crystallization (IC). The peptide nanotubes encased in the COF catalyze C-C bond cleavage in a buffer medium as well as in different organic solvents (such as acetonitrile, acetone, and dichloromethane). The TpAzo-C10FFVK catalyst, being heterogeneous, is easily recoverable, enabling the reaction to be performed for multiple cycles. Additionally, the synthesis of TpAzo-C10FFVK thin films facilitates catalysis in flow. As control, we synthesized another peptide-amphiphile, C10FFVR, which also forms tubular assemblies. By depositing TpAzo COF crystallites on C10FFVR nanotubes through IC, we produced TpAzo-C10FFVR nanotubular structures that expectedly did not show catalysis, suggesting the critical role of the lysines in the TpAzo-C10FFVK.
Collapse
Affiliation(s)
- Ashok Kumar Mahato
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
- Centre for Advanced Functional Materials, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
| | - Sumit Pal
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
- Centre for Advanced Functional Materials, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
| | - Kaushik Dey
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
- Centre for Advanced Functional Materials, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
| | - Antara Reja
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
- Centre for Advanced Functional Materials, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
| | - Satyadip Paul
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
- Centre for Advanced Functional Materials, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
| | - Ankita Shelke
- Central NMR Facility and Physical/Materials Chemistry Division, CSIR-National Chemical Laboratory, Pune 411008, India
| | - Thalasseril G Ajithkumar
- Central NMR Facility and Physical/Materials Chemistry Division, CSIR-National Chemical Laboratory, Pune 411008, India
| | - Dibyendu Das
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
- Centre for Advanced Functional Materials, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
| | - Rahul Banerjee
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
- Centre for Advanced Functional Materials, Indian Institute of Science Education and Research, Kolkata, Mohanpur 741246, India
| |
Collapse
|
46
|
Stillger L, Viau L, Kamm L, Holtmann D, Müller D. Optimization of antimicrobial peptides for the application against biocorrosive bacteria. Appl Microbiol Biotechnol 2023:10.1007/s00253-023-12562-9. [PMID: 37154907 DOI: 10.1007/s00253-023-12562-9] [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: 01/12/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/10/2023]
Abstract
Microbiologically influenced corrosion is a common problem in the industrial field due to the deterioration of metals in the presence of various microorganisms, in particular sulfate-reducing bacteria (SRB) and sulfur-oxidizing bacteria (SOB). A common method to reduce microbiologically influenced corrosion is the application of biocides. The limited number of suitable biocides and the resulting development of resistance, high dosage, and high application rate hinder an effective application. An environmentally friendly alternative could be the application of antimicrobial peptides (AMP), which have already been established in the field of medical devices for a while. Here, the successful treatment of different AMPs against 3 SRB and 1 SOB was demonstrated. The peptide L5K5W was favored due to its broad activity, high stability, and simple structure resulting in low synthesis costs. An alanine scan showed that substitution of leucine with tryptophan increased the activity of this peptide twofold compared to the original peptide against D. vulgaris, the main representative of SRB. Additional optimization of this modified peptide through changes in amino acid composition and lipidations significantly increased the effectiveness, finally resulting in a minimum inhibitory concentration (MIC) of 15.63 μg/mL against Desulfovibrio vulgaris. Even against the marine SRB Desulfovibrio indonesiensis with a required salt concentration of min. 2%, an activity of the peptides can be observed (MIC: 31.25 μg/mL). The peptides also remained stable and active for 7 days in the supernatant of the bacterial culture. KEY POINTS: • Antimicrobial peptides provide an alternative to combat biocorrosive bacteria. • Optimization of the peptide sequence leads to a significant increase in activity. • The investigated peptides exhibit high stability, both in the medium and in the bacterial supernatant.
Collapse
Affiliation(s)
- L Stillger
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstrasse 14, 35390, Giessen, Germany
| | - L Viau
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstrasse 14, 35390, Giessen, Germany
| | - L Kamm
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstrasse 14, 35390, Giessen, Germany
| | - D Holtmann
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstrasse 14, 35390, Giessen, Germany
| | - D Müller
- Institute of Pharmaceutical Technology and Biopharmacy, Philipps-University Marburg, Biegenstraße 10, 35307, Marburg, Germany.
| |
Collapse
|
47
|
Mitra S, Coopershlyak M, Li Y, Chandersekhar B, Koenig R, Chen MT, Evans B, Heinrich F, Deslouches B, Tristram-Nagle S. Novel Helical Trp- and Arg-Rich Antimicrobial Peptides Locate Near Membrane Surfaces and Rigidify Lipid Model Membranes. ADVANCED NANOBIOMED RESEARCH 2023; 3:2300013. [PMID: 37476397 PMCID: PMC10358585 DOI: 10.1002/anbr.202300013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023] Open
Abstract
Antibiotics are losing effectiveness as bacteria become resistant to conventional drugs. To find new alternatives, antimicrobial peptides (AMPs) are rationally designed with different lengths, charges, hydrophobicities (H ), and hydrophobic moments (μH ), containing only three types of amino acids: arginine, tryptophan, and valine. Six AMPs with low minimum inhibitory concentrations (MICs) and <25% toxicity to mammalian cells are selected for biophysical studies. Their secondary structures are determined using circular dichroism (CD), which finds that the % α -helicity of AMPs depends on composition of the lipid model membranes (LMMs): gram-negative (G ( - ) ) inner membrane (IM) >gram-positive (G ( + ) ) > Euk33 (eukaryotic with 33 mol% cholesterol). The two most effective peptides, E2-35 (16 amino acid [AA] residues) and E2-05 (22 AAs), are predominantly helical in G ( - ) IM and G ( + ) LMMs. AMP/membrane interactions such as membrane elasticity, chain order parameter, and location of the peptides in the membrane are investigated by low-angle and wide-angle X-ray diffuse scattering (XDS). It is found that headgroup location correlates with efficacy and toxicity. The membrane bending modulus K C displays nonmonotonic changes due to increasing concentrations of E2-35 and E2-05 in G ( - ) and G ( + ) LMMs, suggesting a bacterial killing mechanism where domain formation causes ion and water leakage.
Collapse
Affiliation(s)
- Saheli Mitra
- Biological Physics Group Physics Department Carnegie Mellon University Pittsburgh, PA 15213, USA
| | - Mark Coopershlyak
- Biological Physics Group Physics Department Carnegie Mellon University Pittsburgh, PA 15213, USA
| | - Yunshu Li
- Biological Physics Group Physics Department Carnegie Mellon University Pittsburgh, PA 15213, USA
| | - Bhairavi Chandersekhar
- Biological Physics Group Physics Department Carnegie Mellon University Pittsburgh, PA 15213, USA
| | - Rachel Koenig
- Biological Physics Group Physics Department Carnegie Mellon University Pittsburgh, PA 15213, USA
| | - Mei-Tung Chen
- Biological Physics Group Physics Department Carnegie Mellon University Pittsburgh, PA 15213, USA
| | - Brandt Evans
- Biological Physics Group Physics Department Carnegie Mellon University Pittsburgh, PA 15213, USA
| | - Frank Heinrich
- Biological Physics Group Physics Department Carnegie Mellon University Pittsburgh, PA 15213, USA
- Center for Neutron Research National Institute of Standards and Technology Gaithersburg, MD 20878, USA
| | - Berthony Deslouches
- Department of Environmental and Occupational Health University of Pittsburgh Pittsburgh, PA 15261, USA
| | - Stephanie Tristram-Nagle
- Biological Physics Group Physics Department Carnegie Mellon University Pittsburgh, PA 15213, USA
| |
Collapse
|
48
|
Avti PK, Singh J, Dahiya D, Khanduja KL. Dual functionality of pyrimidine and flavone in targeting genomic variants of EGFR and ER receptors to influence the differential survival rates in breast cancer patients. Integr Biol (Camb) 2023; 15:zyad014. [PMID: 38084900 DOI: 10.1093/intbio/zyad014] [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: 05/17/2023] [Revised: 10/01/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023]
Abstract
Breast cancer ranks as one of the most prevalent forms of cancer and stands as the primary global cause of mortality among women. Overexpression of EGFR and ER receptors or their genomic alterations leads to malignant transformation, disease aggression and is linked to poor patient survival outcomes. The clinical breast cancer patient's genomic expression, survival analysis, and computational drug-targeting approaches were used to identify best-hit phytochemicals for therapeutic purposes. Breast cancer patients have genomic alterations in EGFR (4%, n = 5699) and ER (9%, n = 8461), with the highest proportion being missense mutations. No statistically significant difference was observed in the patient survival rates between the altered and unaltered ER groups, unlike EGFR, with the lowest survival rates in the altered group. Computational screening of natural compound libraries (7711) against each EGFR (3POZ) and ER (3ERT) receptor shortlists the best-hit 3 compounds with minimum docking score (ΔG = -7.9 to -10.8), MMGBSA (-40.16 to -51.91 kcal/mol), strong intermolecular H-bonding, drug-like properties with least kd, and ki. MD simulation studies display stable RMSD, RMSF, and good residual correlation of best-hit common compounds (PubChem ID: 5281672 and 5280863) targeting both EGFR and ER receptors. In vitro, studies revealed that these common drugs exhibited a high anti-proliferative effect on MCF-7 and MDA-MB-231 breast cancer cells, with effective IC50 values (15-40 μM) and lower free energy, kd, and ki (5281672 > 5280863 > 5330286) much affecting HEK-293 non-cancerous cells, indicating the safety profile. The experimental and computational correlation studies suggest that the highly expressed EGFR and ER receptors in breast cancer patients having poor survival rates can be effectively targeted with best-hit common potent drugs with a multi-target therapeutic approach. Insight Box: The findings of this study provide valuable insights into the genomic/proteomic data, breast cancer patient's survival analysis, and EGFR and ER receptor variants structural analysis. The genetic alterations analysis of EGFR and ER/ESR1 in breast cancer patients reveals the high frequency of mutation types, which affect patient's survival rate and targeted therapies. The common best-hit compounds affect the cell survival patterns with effective IC50, drug-like properties having lower equilibrium and dissociation constants demonstrating the anti-proliferative effects. This work integrates altered receptor structural analysis, molecular interaction-based simulations, and ADMET properties to illuminate the identified best hits phytochemicals potential efficacy targeting both EGFR and ER receptors, demonstrating a multi-target therapeutic approach.
Collapse
Affiliation(s)
- Pramod K Avti
- Department of Biophysics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Jitender Singh
- Department of Biophysics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Divya Dahiya
- Department of General Surgery, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Krishan L Khanduja
- Department of Biophysics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
49
|
Noble JE, Vila-Gómez P, Rey S, Dondi C, Briones A, Aggarwal P, Hoose A, Baran M, Ryadnov MG. Folding-Mediated DNA Delivery by α-Helical Amphipathic Peptides. ACS Biomater Sci Eng 2023; 9:2584-2595. [PMID: 37014978 DOI: 10.1021/acsbiomaterials.3c00221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2023]
Abstract
The renaissance gene therapy experiences these days requires specialist biomaterials and a systemic understanding of major factors influencing their ability to deliver genetic material. Peptide transfection systems represent a major class of such biomaterials. Several peptidic reagents have been commercialized to date. However, a comparative assessment of peptide sequences alone without auxiliary support or excipients against a common determinant for their ability to complex and deliver DNA has been lacking. This study cross-compares commercial and experimental transfection reagents from the same family of helical amphiphiles. Factors defining the efficacy of DNA delivery including cell uptake and gene expression are assessed along with cytotoxicity and DNA complexation. The results show that despite differences in sequence composition, length, and origin, peptide reagents of the same structural family exhibit similar characteristics and limitations with common variability trends. The cross-comparison revealed that functional DNA delivery is independent of the peptide sequence used but is mediated by the ability of the reagents to co-fold with DNA. Peptide folding proved to be the common determinant for DNA complexation and delivery by peptidic transfection reagents.
Collapse
Affiliation(s)
- James E Noble
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, U.K
| | - Paula Vila-Gómez
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, U.K
- Department of Brain Sciences, Imperial College London, London W12 0TR, U.K
| | - Stephanie Rey
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, U.K
| | - Camilla Dondi
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, U.K
| | - Andrea Briones
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, U.K
| | - Purnank Aggarwal
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, U.K
| | - Alex Hoose
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, U.K
| | - Maryana Baran
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, U.K
| | - Maxim G Ryadnov
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, U.K
- Department of Physics, King's College London, London WC2R 2LS, U.K
| |
Collapse
|
50
|
Interaction of guanidinium and ammonium cations with phosphatidylcholine and phosphatidylserine lipid bilayers - Calorimetric, spectroscopic and molecular dynamics simulations study. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2023; 1865:184122. [PMID: 36739930 DOI: 10.1016/j.bbamem.2023.184122] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/10/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023]
Abstract
The ability of arginine-rich peptides to cross the lipid bilayer and enter cytoplasm, unlike their lysine-based analogues, is intensively studied in the context of cell-penetrating peptides. Although the experiments have not yet reconstructed their internalization mechanism, the computational studies have shown that the type or charge of lipid polar groups is one of the crucial factors in their translocation. In order to gain more detailed insight into the interaction of guanidinium (Gdm+) and ammonium (NH4+) cations, as important building blocks in arginine and lysine amino acids, with lipid bilayers, we conducted the experimental and computational study that tackles this phenomenon. The adsorption of Gdm+ and NH4+ on lipid bilayers prepared from a zwitterionic (DPPC) and an anionic (DPPS) lipid was examined by thermoanalytic and spectroscopic techniques. Using temperature-dependent UV-Vis spectroscopy and DSC calorimetry we determined the impact of Gdm+ and NH4+ on the thermotropic properties of lipid bilayers. FTIR data, along with molecular dynamics simulations, unraveled the molecular-level details on the nature of their interactions, showing the proton transfer between NH4+ and DPPS, but not between Gdm+ and DPPS. The findings originated from this work imply that Gdm+ and NH4+ form qualitatively different interactions with lipids of different charge which is reflected in the physico-chemical interactions that arginine-and lysine-based peptides establish at a complex and chemically heterogeneous environment such as the biological membrane.
Collapse
|