1
|
Nair SS, Kleffmann T, Smith B, Morris V, Göbl C, Pletzer D, Fellner M. Comparative lipidomics profiles of planktonic and biofilms of methicillin-resistant and -susceptible Staphylococcus aureus. Anal Biochem 2025; 698:115746. [PMID: 39672221 DOI: 10.1016/j.ab.2024.115746] [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: 10/30/2024] [Revised: 11/27/2024] [Accepted: 12/06/2024] [Indexed: 12/15/2024]
Abstract
Staphylococcus aureus is a significant human pathogen causing acute life-threatening, and chronic infections often linked to biofilms. This study conducted a comparative lipidomic analysis of a methicillin-resistant (MRSA) and a methicillin-susceptible (MSSA) S. aureus strain in both planktonic and biofilm cultures using liquid chromatography-mass spectrometry (LC-MS) and nuclear magnetic resonance (NMR) spectroscopy. The developed protocol successfully differentiates between the strains in various living states (planktonic and biofilm) and growth media (Tryptic Soy Broth and Brain Heart Infusion) using S. aureus USA300 LAC (MRSA) and S. aureus Newman (MSSA). LC-MS and NMR lipidomics profiles revealed global differences and particular ones among the following classes of bacterial lipids: phosphatidylglycerols, diacylglycerols, monoglycosyldiacylglycerols, diglycosyldiacylglycerols, and cardiolipins. Lipid content was higher in the biofilm states for most of these classes. Growth media differences were significant, while differences between MRSA and MSSA were less pronounced but still detectable. Additionally, we provide data on hundreds of unknown compounds that differ based on living state, strain background, or growth media. This study offer insights into the dynamic nature of S. aureus lipid composition and the used methods are adaptable to other organisms.
Collapse
Affiliation(s)
- Shilpa Saseendran Nair
- Biochemistry Department, University of Otago, Dunedin, New Zealand; Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Torsten Kleffmann
- Division of Health Sciences, Research Infrastructure Centre, University of Otago, Dunedin, New Zealand
| | - Briana Smith
- Mātai Hāora - Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Vanessa Morris
- School of Biological Sciences, University of Canterbury, Christchurch, New Zealand; Biomolecular Interaction Centre, University of Canterbury, Christchurch, New Zealand
| | - Christoph Göbl
- Mātai Hāora - Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand; Biomolecular Interaction Centre, University of Canterbury, Christchurch, New Zealand
| | - Daniel Pletzer
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Matthias Fellner
- Biochemistry Department, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
2
|
Bollati M, Fasola E, Pieraccini S, Freddi F, Cocomazzi P, Oliva F, Klußmann M, Maspero A, Piarulli U, Ferrara S, Pellegrino S, Bertoni G, Gazzola S. Impairing protein-protein interactions in an essential tRNA modification complex: An innovative antimicrobial strategy against Pseudomonas aeruginosa. J Pept Sci 2025; 31:e3658. [PMID: 39434676 DOI: 10.1002/psc.3658] [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: 05/29/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/23/2024]
Abstract
Protein-protein interactions (PPIs) have been recognized as a promising target for the development of new drugs, as proved by the growing number of PPI modulators reaching clinical trials. In this context, peptides represent a valid alternative to small molecules, owing to their unique ability to mimic the target protein structure and interact with wider surface areas. Among the possible fields of interest, bacterial PPIs represent an attractive target to face the urgent necessity to fight antibiotic resistance. Growing attention has been paid to the YgjD/YeaZ/YjeE complex responsible for the essential t6A37 tRNA modification in bacteria. We previously identified an α-helix on the surface of Pseudomonas aeruginosa YeaZ, crucial for the YeaZ-YeaZ homodimer formation and the conserved YeaZ-YgjD interactions. Herein, we present our studies for impairing the PPIs involved in the formation of the YeaZ dimers through synthetic peptide derivatives of this helical moiety, both in vitro with purified components and on P. aeruginosa cells. Our results proved the possibility of targeting those PPIs which are usually essential for protein functioning and thus are refractory to mutational changes and antibiotic resistance development.
Collapse
Affiliation(s)
- Michela Bollati
- Institute of Biophysics, National Research Council, Milan, Italy
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Elettra Fasola
- Department of Science and High Technology, Università degli Studi dell'Insubria, Como, Italy
| | | | - Francesca Freddi
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Paolo Cocomazzi
- Institute of Biophysics, National Research Council, Milan, Italy
| | - Francesco Oliva
- Department of Chemistry, Università degli Studi di Milano, Milan, Italy
| | - Merlin Klußmann
- Department of Chemistry, Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Angelo Maspero
- Department of Science and High Technology, Università degli Studi dell'Insubria, Como, Italy
| | - Umberto Piarulli
- Department of Science and High Technology, Università degli Studi dell'Insubria, Como, Italy
| | - Silvia Ferrara
- Institute of Biophysics, National Research Council, Milan, Italy
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Sara Pellegrino
- Pharmaceutical Science Department, University of Milan, Milan, Italy
| | - Giovanni Bertoni
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Silvia Gazzola
- Department of Science and High Technology, Università degli Studi dell'Insubria, Como, Italy
| |
Collapse
|
3
|
Tu MM, Carfrae LA, Rachwalski K, French S, Catacutan D, Gordzevich R, MacNair CR, Speagle ME, Werah F, Stokes JM, Brown ED. Exploiting the fitness cost of metallo-β-lactamase expression can overcome antibiotic resistance in bacterial pathogens. Nat Microbiol 2025:10.1038/s41564-024-01883-8. [PMID: 39747690 DOI: 10.1038/s41564-024-01883-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 11/13/2024] [Indexed: 01/04/2025]
Abstract
Carbapenems are last-resort antibiotics for treating bacterial infections. The widespread acquisition of metallo-β-lactamases, such as VIM-2, contributes to the emergence of carbapenem-resistant pathogens, and currently, no metallo-β-lactamase inhibitors are available in the clinic. Here we show that bacteria expressing VIM-2 have impaired growth in zinc-deprived environments, including human serum and murine infection models. Using transcriptomic, genomic and chemical probes, we identified molecular pathways critical for VIM-2 expression under zinc limitation. In particular, disruption of envelope stress response pathways reduced the growth of VIM-2-expressing bacteria in vitro and in vivo. Furthermore, we showed that VIM-2 expression disrupts the integrity of the outer membrane, rendering VIM-2-expressing bacteria more susceptible to azithromycin. Using a systemic murine infection model, we showed azithromycin's therapeutic potential against VIM-2-expressing pathogens. In all, our findings provide a framework to exploit the fitness trade-offs of resistance, potentially accelerating the discovery of additional treatments for infections caused by multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Megan M Tu
- Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada
| | - Lindsey A Carfrae
- Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada
| | - Kenneth Rachwalski
- Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada
| | - Shawn French
- Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada
| | - Denise Catacutan
- Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada
| | - Rodion Gordzevich
- Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada
| | - Craig R MacNair
- Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada
| | - Melissa E Speagle
- Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada
| | - Firas Werah
- Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan M Stokes
- Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada
| | - Eric D Brown
- Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada.
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
4
|
Davoodi A, Akhbari K, Alirezvani M. H2O2-sensitive release of curcumin and zinc in normal and infected simulated cell tissues from a curcumin-zinc coordination complex with prolonged antibacterial activity. INORG CHEM COMMUN 2025; 171:113599. [DOI: 10.1016/j.inoche.2024.113599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
5
|
Pranathi AN, Devendra N, Bollikanda RK, Bangalore PK, Esaulkova IL, Mikhalsky MG, Niukalova MA, Zarubaev VV, Sridhar B, Kantevari S. 6-aryloxy-2-aminopyrimidine-benzonitrile hybrids as anti-infective agents: Synthesis, bioevaluation, and molecular docking. Arch Pharm (Weinheim) 2025; 358:e2400580. [PMID: 39721989 DOI: 10.1002/ardp.202400580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/20/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024]
Abstract
This report explores the potential of novel 6-aryloxy-2-aminopyrimidine-benzonitrile scaffolds as promising anti-infective agents in the face of the increasing threat of infectious diseases. Starting from 2-amino-4,6-dichloropyrimidine, a series of 24 compounds inspired from the antiviral drugs dapivirine, etravirine, and rilpivirine were designed and synthesized via a two-step reaction sequence in good yields. Biological testing of synthetic analogs revealed potent inhibition against both viral and tuberculosis targets. Notably, compounds 5p (2,4-dimethyl substitution; IC50 = 44 ± 4.9 µM; selectivity index [SI] = 20) and 5 s (3-thiophenphenyl; IC50 = 6 ± 1 µM; SI = 120) showed significant antiviral activity against pandemic influenza virus A/Puerto Rico/8/34 (H1N1) with positive toxicity profiles and also exhibited good IC50 values (5p, IC50 = 10 ± 2 µM; SI = 9 and 5 s, IC50 = 16 ± 2 µM; SI = 60) against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (Wuhan strain) compared with favipiravir. In addition, analogs 5a, 5r, 5t, and 5u showed good antitubercular activity against Mycobacterium tuberculosis H37Rv strain and compounds 3, 5f, 5n, and 5q showed moderate antibacterial activity against gram+ve and gram-ve bacterial strains, suggesting that this scaffold has a broad spectrum of therapeutic effects.
Collapse
Affiliation(s)
- Abburi Naga Pranathi
- Fluoro & Agro Chemicals Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Nagineni Devendra
- Fluoro & Agro Chemicals Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rakesh K Bollikanda
- Fluoro & Agro Chemicals Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Pavan K Bangalore
- Fluoro & Agro Chemicals Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Iana L Esaulkova
- Pasteur Institute of Epidemiology and Microbiology, St. Petersburg, Russia
| | | | - Maria A Niukalova
- Pasteur Institute of Epidemiology and Microbiology, St. Petersburg, Russia
| | | | - Balasubramanian Sridhar
- Centre for X-ray Crystallography, Analytical Chemistry Division, CSIR- Indian Institute of Chemical Technology, Hyderabad, Telangana, India
| | - Srinivas Kantevari
- Fluoro & Agro Chemicals Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
6
|
Jamal GA, Jahangirian E, Hamblin MR, Mirzaei H, Tarrahimofrad H, Alikowsarzadeh N. Proteases, a powerful biochemical tool in the service of medicine, clinical and pharmaceutical. Prep Biochem Biotechnol 2025; 55:1-25. [PMID: 38909284 DOI: 10.1080/10826068.2024.2364234] [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: 06/24/2024]
Abstract
Proteases, enzymes that hydrolyze peptide bonds, have various applications in medicine, clinical applications, and pharmaceutical development. They are used in cancer treatment, wound debridement, contact lens cleaning, prion degradation, biofilm removal, and fibrinolytic agents. Proteases are also crucial in cardiovascular disease treatment, emphasizing the need for safe, affordable, and effective fibrinolytic drugs. Proteolytic enzymes and protease biosensors are increasingly used in diagnostic and therapeutic applications. Advanced technologies, such as nanomaterials-based sensors, are being developed to enhance the sensitivity, specificity, and versatility of protease biosensors. These biosensors are becoming effective tools for disease detection due to their precision and rapidity. They can detect extracellular and intracellular proteases, as well as fluorescence-based methods for real-time and label-free detection of virus-related proteases. The active utilization of proteolytic enzymatic biosensors is expected to expand significantly in biomedical research, in-vitro model systems, and drug development. We focused on journal articles and books published in English between 1982 and 2024 for this study.
Collapse
Affiliation(s)
- Ghadir A Jamal
- Faculty of Allied Health Sciences, Kuwait University, Kuwait City, Kuwait
| | - Ehsan Jahangirian
- Department of Molecular, Zist Tashkhis Farda Company (tBioDx), Tehran, Iran
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Faculty of Health Science, Laser Research Center, University of Johannesburg, Doornfontein, South Africa
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Neda Alikowsarzadeh
- Molecular and Life Science Department, Han University of Applied Science, Arnhem, Nederland
| |
Collapse
|
7
|
Huang HY, Xue RY, Xiao SX, Huang LT, Liao XW, Wang JT, Duan XM, Yu RJ, Xiong YS. AIE-based ruthenium complexes as photosensitizers for specifically photo-inactivate gram-positive bacteria. J Inorg Biochem 2025; 262:112755. [PMID: 39388808 DOI: 10.1016/j.jinorgbio.2024.112755] [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: 07/15/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/12/2024]
Abstract
The emergence of multidrug-resistant bacterial have caused severe burden for public health. Particularly, Staphylococcus aureus as one of ESKAPE pathogens have induced various infectious diseases and resulted in increasing deaths. Developing new antibacterial agents is still urgent and challenging. Fortunately, in this study, based on aggregation-induced emission (AIE) ruthenium complexes were designed and synthesized, which realized the high efficiency of reactive oxygen species generation and remarkably killed S. aureus unlike conventional antibiotics action. Significantly, owing to good singlet oxygen production ability, Ru1 at only 4 μg/mL of concentration displayed good antibacterial photodynamic therapy effect upon white light irradiation and could deplete essential coenzyme NADH to disrupt intracellular redox balance. Also, the electrostatic interaction between Ru1 and bacteria enhanced the possibility of antibacterial. Under light irradiation, Ru1 could efficiently inhibit the biofilm growth and avoid the development of drug-resistant. Furthermore, Ru1 possessed excellent biocompatibility and displayed remarkable therapy effect in treating mice-wound infections in vivo. These findings indicated that AIE-based ruthenium complexes as new antibacterial agent had great potential in photodynamic therapy of bacteria and addressing the drug-resistance crisis.
Collapse
Affiliation(s)
- Hai-Yan Huang
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Run-Yu Xue
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Su-Xin Xiao
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Li-Ting Huang
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Xiang-Wen Liao
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Jin-Tao Wang
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Xue-Min Duan
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Ru-Jian Yu
- School of life science, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Yan-Shi Xiong
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China.
| |
Collapse
|
8
|
Elsharabasy SA, Sayed MT, Abdel-Aziem A. Novel coumarin linked pyrazoles, thiazoles, and thiadiazoles: synthetic strategies and in vitro antimicrobial investigation. Future Med Chem 2024:1-11. [PMID: 39723730 DOI: 10.1080/17568919.2024.2444867] [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: 10/09/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
AIM Emerging resistance among pathogens necessitates the development of novel antimicrobial agents. As a result, we aimed to synthesize new coumarins and study their antimicrobial activity with the hope of obtaining effective drugs. METHOD A series of coumarins were synthesized, characterized, and assessed for antimicrobial activity using broth microdilution and agar diffusion methods against Gram-positive (Bacillus pumilis, Streptococcus faecalis), Gram-negative (Escherichia coli, Enterobacter cloacae) bacteria, and fungi (Saccharomyces cerevisiae, Candida albicans). RESULTS Pyrazoles 15 and 16 revealed promising activities against all bacterial strains with MIC values ranging from 1.95 to 15.6 µg/ml. Notably, pyrazole 15 with CF3 in 3-position of pyrazole ring demonstrated higher ability to inhibit Streptococcus faecalis strain with MIC value equal to penicillin G (3.91 µg/ml). It also exhibited the best bactericidal potency against Escherichia coli with MBC value of 15.6 µg/ml while, pyrazole 16 recorded the same MBC value against Enterobacter cloacae. Pyrazole 15 demonstrated the strongest antifungal activity against both fungal strains with MIC and MFC values of 15.6, 7.81, 62.5, and 31.3 µg/ml against Saccharomyces cerevisiae and Candida albicans, respectively. CONCLUSION These findings underscore the potential of coumarins, particularly compounds 15 and 16, as effective antimicrobial agents and provide critical insights into the design of bioactive molecules.
Collapse
Affiliation(s)
- Salwa A Elsharabasy
- Chemistry Department, Faculty of science(Girls), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Mariam T Sayed
- Chemistry Department, Faculty of science(Girls), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Anhar Abdel-Aziem
- Chemistry Department, Faculty of science(Girls), Al-Azhar University, Nasr City, Cairo, Egypt
| |
Collapse
|
9
|
Poulsen BE, Warrier T, Barkho S, Bagnall J, Romano KP, White T, Yu X, Kawate T, Nguyen PH, Raines K, Ferrara K, Golas AL, FitzGerald M, Boeszoermenyi A, Kaushik V, Serrano-Wu M, Shoresh N, Hung DT. Discovery of a Pseudomonas aeruginosa-specific small molecule targeting outer membrane protein OprH-LPS interaction by a multiplexed screen. Cell Chem Biol 2024:S2451-9456(24)00490-2. [PMID: 39732052 DOI: 10.1016/j.chembiol.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 08/19/2024] [Accepted: 12/03/2024] [Indexed: 12/30/2024]
Abstract
The surge of antimicrobial resistance threatens efficacy of current antibiotics, particularly against Pseudomonas aeruginosa, a highly resistant gram-negative pathogen. The asymmetric outer membrane (OM) of P. aeruginosa combined with its array of efflux pumps provide a barrier to xenobiotic accumulation, thus making antibiotic discovery challenging. We adapted PROSPECT, a target-based, whole-cell screening strategy, to discover small molecule probes that kill P. aeruginosa mutants depleted for essential proteins localized at the OM. We identified BRD1401, a small molecule that has specific activity against a P. aeruginosa mutant depleted for the essential lipoprotein, OprL. Genetic and chemical biological studies identified that BRD1401 acts by targeting the OM β-barrel protein OprH to disrupt its interaction with LPS and increase membrane fluidity. Studies with BRD1401 also revealed an interaction between OprL and OprH, directly linking the OM with peptidoglycan. Thus, a whole-cell, multiplexed screen can identify species-specific chemical probes to reveal pathogen biology.
Collapse
Affiliation(s)
- Bradley E Poulsen
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Thulasi Warrier
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sulyman Barkho
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Keith P Romano
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Tiantian White
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Xiao Yu
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Tomohiko Kawate
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Phuong H Nguyen
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kyra Raines
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kristina Ferrara
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - A Lorelei Golas
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | | | - Virendar Kaushik
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; 3 Point Bio LLC, Cambridge, MA 02142, USA
| | | | - Noam Shoresh
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Deborah T Hung
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
10
|
Zhao H, Sun J, Cheng Y, Nie S, Li W. Advances in peptide/polymer antimicrobial assemblies. J Mater Chem B 2024. [PMID: 39714335 DOI: 10.1039/d4tb02144d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Antimicrobial peptides (AMPs) have been extensively exploited as promising drugs to cope with antibiotic-resistant bacteria in clinical treatment. Peptide/polymer assembly provides a particularly important contribution to this topic and has emerged as a new paradigm for the development of nano-antimicrobial systems with previously unattainable outcomes. In this review article, we systematically summarize the recent advances in antimicrobial peptide/polymer assemblies. We describe a brief background and several classified systems based on peptide/polymer assemblies. We discuss the molecular design and the general rules behind the assembled nanostructures and bioactivities. The key role of polymers in improving the antimicrobial activity, stability, cytotoxicity, and bioavailability of peptides is emphasized based on the reported systems. The resulting peptide/polymer assemblies with stimuli-responsiveness, value-added properties and potential applications are demonstrated. The outlook of the antimicrobial peptide/polymer assemblies is also presented from the viewpoint of bio-applications.
Collapse
Affiliation(s)
- He Zhao
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Qianjin Avenue 2699, Changchun 130012, China.
| | - Jiayi Sun
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Qianjin Avenue 2699, Changchun 130012, China.
| | - Yi Cheng
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Qianjin Avenue 2699, Changchun 130012, China.
| | - Shuaishuai Nie
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Qianjin Avenue 2699, Changchun 130012, China.
| | - Wen Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Qianjin Avenue 2699, Changchun 130012, China.
| |
Collapse
|
11
|
Leng J, Liu X, Xu Y, Zhu SE, Zhang Y, Tan Z, Yang X, Jin JE, Shi Y, Fan H, Yang Y, Yao H, Zhang Y, Chong H, Wang C. Evaluation of the alkyl chain length and photocatalytic antibacterial performance of cation g-C3N4. J Mater Chem B 2024; 13:264-273. [PMID: 39535027 DOI: 10.1039/d4tb01118j] [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: 11/16/2024]
Abstract
Several cation graphite carbon nitrides (g-C3N4-(CH2)n-ImI+) were synthesized by chemically attaching imidazolium appended alkane chains with different lengths (n = 2, 4, 8, 12 and 16) to g-C3N4. The introduction of a cation segment potentially improved the interaction between the carbon material and Gram negative (MDR-A. baumannii) and Gram positive (S. aureus) bacteria as characterized by ζ potential measurement. Short alkane chain (carbon numbers of 2, 4 and 8) carbon materials displayed relatively stronger bacterial interactions compared to long alkane chain bearing ones (n = 12 and 16). In addition, short chain carbon materials (g-C3N4-(CH2)4-ImI+) displayed relatively higher photocatalytic reactive oxygen species (1O2, ˙O2- and ˙OH) production efficiency. Bacterial interaction and ROS production efficiency synergistically contribute to photocatalytic antibacterial performance. The current data revealed that g-C3N4 with short flexible cations attached exhibited bacterial interaction and ROS production. Among these synthesized materials, g-C3N4-(CH2)4-ImI+ exhibited the most pronounced photocatalytic antibacterial efficiency (>99%).
Collapse
Affiliation(s)
- Junling Leng
- Department of Emergency, Affiliated Hospital of Yangzhou University, Yangzhou 225000, Jiangsu, China
| | - Xuanwei Liu
- Department of Chemical and Chemical Engineering, Yangzhou University, No. 180, Si-Wang-Ting Rd., Yangzhou, Jiangsu, 225009, China.
| | - Yin Xu
- Department of Chemical and Chemical Engineering, Yangzhou University, No. 180, Si-Wang-Ting Rd., Yangzhou, Jiangsu, 225009, China.
| | - Shi-En Zhu
- Department of Chemical and Chemical Engineering, Yangzhou University, No. 180, Si-Wang-Ting Rd., Yangzhou, Jiangsu, 225009, China.
| | - Yuefei Zhang
- Department of Emergency, Affiliated Hospital of Yangzhou University, Yangzhou 225000, Jiangsu, China
| | - Zhongbing Tan
- Department of Chemical and Chemical Engineering, Yangzhou University, No. 180, Si-Wang-Ting Rd., Yangzhou, Jiangsu, 225009, China.
| | - Xiaofei Yang
- Department of Chemical and Chemical Engineering, Yangzhou University, No. 180, Si-Wang-Ting Rd., Yangzhou, Jiangsu, 225009, China.
| | - Jia-En Jin
- Department of Chemical and Chemical Engineering, Yangzhou University, No. 180, Si-Wang-Ting Rd., Yangzhou, Jiangsu, 225009, China.
| | - Yufeng Shi
- Department of Chemical and Chemical Engineering, Yangzhou University, No. 180, Si-Wang-Ting Rd., Yangzhou, Jiangsu, 225009, China.
| | - Hongying Fan
- Testing Center of Yangzhou University, Yangzhou, 225009, China
| | - Yi Yang
- Center Laboratory, Affiliated Hospital of Yangzhou University, Yangzhou 225000, Jiangsu, China
| | - Hang Yao
- Department of Chemical and Chemical Engineering, Yangzhou University, No. 180, Si-Wang-Ting Rd., Yangzhou, Jiangsu, 225009, China.
| | - Yu Zhang
- School of Nursing, Yangzhou University, Yangzhou, China.
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, No. 88 South University Rd., Yangzhou, 225009, China
| | - Hui Chong
- Department of Chemical and Chemical Engineering, Yangzhou University, No. 180, Si-Wang-Ting Rd., Yangzhou, Jiangsu, 225009, China.
- Institute of Innovation Materials and Energy, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Chengyin Wang
- Department of Chemical and Chemical Engineering, Yangzhou University, No. 180, Si-Wang-Ting Rd., Yangzhou, Jiangsu, 225009, China.
| |
Collapse
|
12
|
Wang T, Teng R, Wu M, Ge Z, Liu Y, Yang B, Li C, Fan Z, Du J. A Polypeptosome Spray To Heal Antibiotic-Resistant Bacteria-Infected Wound by Photocatalysis-Induced Metabolism-Interference. ACS NANO 2024. [PMID: 39688563 DOI: 10.1021/acsnano.4c13965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
With the booming antimicrobial drug resistance worldwide, traditional antibacterial agents (e.g., antibiotics) are usually powerless against superbug. Targeting antibacterial pathways different from traditional antibiotics could be an effective approach to treating wounds with a resistant bacterial infection. In this work, an antibacterial polymersome was developed to physically induce bacterial membrane damage and interfere with bacterial metabolism. First, we synthesized an antibacterial poly(ε-caprolactone)-block-poly(glutamic acid)-block-poly(Lys-stat-Phe) copolymer, which was then self-assembled into polypeptosome with the amplification of surface positive charges to disrupt bacterial membranes. In addition, the polypeptosome was further decorated with photocatalytic bismuth sulfide (Bi2S3) nanoparticles as a photocatalyst to interfere with reduced nicotinamide adenine dinucleotide (NADH) conversion. Specifically, near-infrared light generated free electrons from Bi2S3 nanoparticles could effectively interfere with NADH homeostasis to induce antibiotic-resistant bacteria death, as verified by transcriptome sequence analysis. Moreover, effective healing of antibiotic-resistant bacteria-infected wounds of mice was achieved with a spray of polypeptosome dispersion. Overall, we provided a fresh strategy to integrate bacterial membrane damage and metabolism interference functions within antibacterial polymersomes for healing antibiotic-resistant bacteria-infected wound.
Collapse
Affiliation(s)
- Tao Wang
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Runxin Teng
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Mengjie Wu
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Zhenghong Ge
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Yaping Liu
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Biao Yang
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Chang Li
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
- Institute for Advanced Study, Tongji University, Shanghai 200092, China
| | - Zhen Fan
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Jianzhong Du
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
- School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
13
|
Tran E, Cheung C, Li L, Carter GP, Gable RW, West NP, Kaur A, Gee YS, Cook GM, Baell JB, Jörg M. Phenotypic-Based Discovery and Exploration of a Resorufin Scaffold with Activity against Mycobacterium tuberculosis. ChemMedChem 2024; 19:e202400482. [PMID: 39248310 PMCID: PMC11648835 DOI: 10.1002/cmdc.202400482] [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: 06/25/2024] [Revised: 08/16/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Tuberculosis remains a leading cause of death by infectious disease. The long treatment regimen and the spread of drug-resistant strains of the causative agent Mycobacterium tuberculosis (Mtb) necessitates the development of new treatment options. In a phenotypic screen, nitrofuran-resorufin conjugate 1 was identified as a potent sub-micromolar inhibitor of whole cell Mtb. Complete loss of activity was observed for this compound in Mtb mutants affected in enzyme cofactor F420 biosynthesis (fbiC), suggesting that 1 undergoes prodrug activation in a manner similar to anti-tuberculosis prodrug pretomanid. Exploration of the structure-activity relationship led to the discovery of novel resorufin analogues that do not rely on the deazaflavin-dependent nitroreductase (Ddn) bioactivation pathway for their antimycobacterial activity. These analogues are of interest as they work through an alternative, currently unknown mechanism that may expand our chemical arsenal towards the treatment of this devastating disease.
Collapse
Affiliation(s)
- Eric Tran
- Medicinal Chemistry, Monash Institute of Pharmaceutical SciencesMonash UniversityParkville, Victoria3052Australia
| | - Chen‐Yi Cheung
- Department of Microbiology and Immunology, Otago School of Medical SciencesUniversity of OtagoDunedin9054New Zealand
| | - Lucy Li
- Department of Microbiology & ImmunologyThe University of Melbourne at The Peter Doherty Institute for Infection & ImmunityMelbourne, Victoria3000Australia
| | - Glen P. Carter
- Department of Microbiology & ImmunologyThe University of Melbourne at The Peter Doherty Institute for Infection & ImmunityMelbourne, Victoria3000Australia
| | - Robert W. Gable
- School of ChemistryUniversity of MelbourneParkville, Victoria3010Australia
| | - Nicholas P. West
- School of Chemistry and Molecular BiosciencesThe University of QueenslandBrisbane, Queensland4072Australia
| | - Amandeep Kaur
- Medicinal Chemistry, Monash Institute of Pharmaceutical SciencesMonash UniversityParkville, Victoria3052Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein ScienceMonash UniversityMelbourne, Victoria3052Australia
| | - Yi Sing Gee
- Medicinal Chemistry, Monash Institute of Pharmaceutical SciencesMonash UniversityParkville, Victoria3052Australia
| | - Gregory M. Cook
- Department of Microbiology and Immunology, Otago School of Medical SciencesUniversity of OtagoDunedin9054New Zealand
| | - Jonathan B. Baell
- Medicinal Chemistry, Monash Institute of Pharmaceutical SciencesMonash UniversityParkville, Victoria3052Australia
| | - Manuela Jörg
- Medicinal Chemistry, Monash Institute of Pharmaceutical SciencesMonash UniversityParkville, Victoria3052Australia
- Chemistry-School of Natural & Environmental SciencesNewcastle University Centre for Cancer, Newcastle UniversityBedson BuildingNewcastle Upon TyneNE1 7RUUK
| |
Collapse
|
14
|
Zhang R, Wang L, Meng L, Shang W, Ren Y, Qi Q, Liu J, Cui B, Meng Z, Jiang X, Ding L, Gou Y, He Y, Zhang Q, Ren C. A slime-inspired phycocyanin/κ-carrageenan-based hydrogel bandage with ultra-stretchability, self-healing, antioxidative, and antibacterial activity for wound healing. Int J Biol Macromol 2024; 289:138786. [PMID: 39675612 DOI: 10.1016/j.ijbiomac.2024.138786] [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: 09/28/2024] [Revised: 11/23/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
Hydrogels have attracted extensive attention as wound dressing owing to their excellent multifunctionality, flexibility, and biocompatibility. Due to the frequent movement and stretching of skin as well as complex surface of wound, traditional wound dressings have difficulty to adapt to motion and irregular wounds. Furthermore, excessive reactive oxygen species (ROS) and bacterial infection can induce delayed wound healing. To this end, we developed a set of versatile phycocyanin-based dual network hydrogels (PPC hydrogels) with polyvinyl alcohol (PVA) and κ-carrageenan (CRG) as substrate via forming of borate ester bonds, hydrogen bonds, and electrostatic interaction. The PPC hydrogels not only possessed adaptivity, ultra-stretchability (7036.12 %), efficient self-healing and injectability, but also possessed antioxidative and antibacterial capacities conferred by C-phycocyanin (PC) and rhein. Moreover, the hydrogels also exhibited excellent hemostatic ability and high biocompatibility. More remarkably, the PPC-I hydrogel could accelerate wound healing by effect of anti-inflammation (downregulating TNF-α and IL-6) and promoting collagen deposition and angiogenesis (upregulating CD31), which may be utilized as hydrogel bandages and applied to motion and irregular wounds, thereby promising the application prospect of the hydrogels as wound dressing.
Collapse
Affiliation(s)
- Renlong Zhang
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China
| | - Linlin Wang
- Department of Food Engineering, Shandong Business Institute, Yantai 264670, Shandong, China
| | - Lingjie Meng
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China
| | - Wenshuo Shang
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China
| | - Yuhang Ren
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China
| | - Qianfen Qi
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China
| | - Jiaxin Liu
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China
| | - Benke Cui
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China
| | - Zhihao Meng
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China
| | - Xue Jiang
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China
| | - Luyao Ding
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China
| | - Yanzhe Gou
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China
| | - Yanhao He
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China.
| | - Qiuyan Zhang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, Shandong, China.
| | - Chunguang Ren
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China.
| |
Collapse
|
15
|
Wu Y, Wei G, Cao X, Wang R, Gou X. Stimuli-responsive dual-drug loaded microspheres with differential drug release for antibacterial and wound repair promotion. Colloids Surf B Biointerfaces 2024; 248:114455. [PMID: 39700570 DOI: 10.1016/j.colsurfb.2024.114455] [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: 10/31/2024] [Revised: 12/02/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
The healing of infected wounds is a complex and dynamic process requiring tailored treatment strategies that address both antimicrobial and reparative needs. Despite the development of numerous drugs, few approaches have been devised to optimize the timing of drug release for targeting distinct phases of infection control and tissue repair, limiting the overall treatment efficacy. Here, a stimuli-responsive microsphere encapsulating dual drugs was developed to facilitate differential drug release during distinct phases of antibacterial and repair promotion, thereby synergistically enhancing wound healing. Specifically, zeolite imidazolate backbone in poly (lactic-co-glycolic acid) (PLGA) microsphere was employed for the encapsulation of ciprofloxacin (CIP), responding to acidic environment of bacteria and releasing antibiotic for antibacterial therapy. Meanwhile, curcumin (CUR) encapsulated in PLGA exhibited a gradual release profile, contributing to synergistic antibacterial effects. During the tissue repair phase, near-infrared light stimulation of Fe3O4 embedded in PLGA generated heat, elevating the temperature to the glass transition point of PLGA, which significantly enhanced the release of CUR thereby promoting tissue repair. In vitro experiments demonstrated that the release of CIP and CUR achieved significant antibacterial effects in the early stages of treatment. Additionally, CUR could effectively enhance fibroblast migration and proliferation. In vivo studies using a mouse abscess model revealed that the microspheres exhibited remarkable antibacterial and wound-healing capabilities, effectively enhancing the re-epithelialization of wound tissue and reducing the infiltration of inflammatory cells. This study provides novel strategies for constructing drug delivery systems that match dynamic stages of wound healing, offering improved therapeutic outcomes for infected wounds.
Collapse
Affiliation(s)
- Yating Wu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, PR China
| | - Guihua Wei
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, PR China
| | - Xin Cao
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, PR China
| | - Ran Wang
- China National Research Institute of Food and Fermentation Industries Corporation Limited, Building 6, Yard 24, Jiuxianqiao Middle Road, Chaoyang District, Beijing 100015, PR China; Beijing Agricultural and Food Synthetic Biological Innovation Center, Zhongguancun Pinggu Agricultural Science and Technology Park, Yukou Town, Pinggu District, Beijing 101299, PR China
| | - Xue Gou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, PR China.
| |
Collapse
|
16
|
Hickl J, Argyropoulou A, Al-Ahmad A, Hellwig E, Skaltsounis AL, Wittmer A, Vach K, Karygianni L. Unleashing nature's defense: potent antimicrobial power of plant extracts against oral pathogens and Streptococcus mutans biofilms. FRONTIERS IN ORAL HEALTH 2024; 5:1469174. [PMID: 39726767 PMCID: PMC11669686 DOI: 10.3389/froh.2024.1469174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Objectives The increasing demand for alternatives to antibiotics against resistant bacteria has led to research on natural products. The aim of this study was to analyze the antimicrobial and antibiofilm activity of 16 Mediterranean herb extracts. Materials and methods The extracts were analyzed using High Performance Thin Layer Chromatography. The minimum inhibitory concentration and minimum bactericidal concentration of the extracts from Achillea taygetea, Cistus creticus ssp. creticus, Cistus monspeliensis, Lavandula stoechas, Mentha aquatica, Mentha longifolia, Origanum vulgare, Phlomis cretica, Rosmarinus officinalis, Salvia sclarea, Satureja parnassica, Satureja thymbra, Sideritis euboea, Sideritis syriaca, Stachys spinosa, and Thymus longicaulis were determined against eight oral bacteria and fungus Candida albicans. Microtiter plate test was conducted to evaluate the antibiofilm activity against Streptococcus mutans. Results Overall, all tested extracts efficiently suppressed the growth of obligate anaerobic bacteria. When applied at concentrations ≥0.15 mg/ml, the extracts exhibited moderate to high antibiofilm activity comparable to that of chlorhexidine (CHX) against S. mutans. Interestingly, R. officinalis (MIC: 0.01-0.06 mg/ml) and O. vulgare (MIC: 0.04-1.25 mg/ml) demonstrated the highest antibacterial activity against oral bacteria. Additionally, R. officinalis and L. stoechas significantly inhibited S. mutans biofilm formation at 0.15 mg/ml. Conclusions The tested plant extracts can be considered as alternative natural antimicrobial and antibiofilm agents. Clinical relevance Mediterranean herb extracts show promise as natural alternatives to combat oral bacteria and biofilm formation, offering potential new therapies for infectious oral diseases in the context of antibiotic resistance.
Collapse
Affiliation(s)
- Joachim Hickl
- Department of Operative Dentistry and Periodontology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Aikaterini Argyropoulou
- Department of Pharmacognosy and Natural Products Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Ali Al-Ahmad
- Department of Operative Dentistry and Periodontology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Elmar Hellwig
- Department of Operative Dentistry and Periodontology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alexios Leandros Skaltsounis
- Department of Pharmacognosy and Natural Products Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Annette Wittmer
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kirstin Vach
- Institute for Medical Biometry and Statistics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Lamprini Karygianni
- Clinic of Conservative and Preventive Dentistry, Center of Dental Medicine University of Zurich, Zurich, Switzerland
| |
Collapse
|
17
|
Li L, Li H, Wang J, Xie Y, Gao M, Yang Z, Li C. An Asymmetric Bacterial Cellulose Membrane Incorporating CuPt Nanozymes and Curcumin for Accelerating Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:67166-67177. [PMID: 39586586 DOI: 10.1021/acsami.4c10720] [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: 11/27/2024]
Abstract
Damaged skin compromises its ability to effectively prevent the invasion of harmful bacteria into the tissue, leading to bacterial infection of the wound and hindering the healing process. To address this challenge, we have developed a multifunctional asymmetric wound dressing (CuPt-Cur-ABC) that effectively addresses the lack of bactericidal activity and the release of active ingredients in conventional bacterial cellulose (BC), which can be employed to create a barrier of defense between the wound and its surrounding environment. Compared with BC, asymmetric bacterial cellulose (ABC) used starch as a pore-causing agent, forming holes of different sizes at the top and bottom, which enhanced the ability of ABC to load and moderate-release drugs. First, as-synthesized CuPt nanozymes with an octopod nanoframe structure had multiple enzymatic activities including peroxidase-like, catalase-like, and glutathione peroxidase-like activities. Then, CuPt and curcumin (Cur) were loaded into ABC under ultrasound. Under 808 nm laser irradiation, the nanocomposites possessed good photothermal properties. So the photothermal therapy combined with chemodynamic therapy and inherent antibacterial performance of Cur achieved 99.3% and 99.6% in vitro bactericidal efficacy against Staphylococcus aureus and Escherichia coli, respectively. Moderate release of Cur can clear the excess reactive oxygen species and promote the polarization of macrophages toward the M2 type. In vivo experiments additionally confirmed that the constructed wound dressing achieved multiple functions, including effective antibacterial activity, reversing the inflammatory microenvironment, and promoting wound healing.
Collapse
Affiliation(s)
- Lei Li
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, Shandong 266237, PR China
| | - Haoze Li
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, Shandong 266237, PR China
| | - Junrong Wang
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, Shandong 266237, PR China
| | - Yulin Xie
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, Shandong 266237, PR China
| | - Minghong Gao
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, Shandong 266237, PR China
| | - Zhongjun Yang
- Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, Shandong 266035, PR China
| | - Chunxia Li
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, Shandong 266237, PR China
| |
Collapse
|
18
|
Lin YW, Cheung SYA. Model-Informed Drug Development (MIDD) for Antimicrobials. Int J Antimicrob Agents 2024; 64:107392. [PMID: 39549766 DOI: 10.1016/j.ijantimicag.2024.107392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Affiliation(s)
- Yu-Wei Lin
- Infection Program, Biomedicine Discovery Institute and Department of Microbiology, 19 Innovation Walk, Monash University, Clayton, VIC, 3800, Australia; Malaya Translational and Clinical Pharmacometrics Group, Faculty of Pharmacy, University of Malaya, Malaysia; Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, University of Malaya, Malaysia; Certara, Radnor, USA.
| | | |
Collapse
|
19
|
Wang J, Zhang M, Li C, Liu M, Qi Y, Xie X, Zhou C, Ma L. A novel cathelicidin TS-CATH derived from Thamnophis sirtalis combats drug-resistant gram-negative bacteria in vitro and in vivo. Comput Struct Biotechnol J 2024; 23:2388-2406. [PMID: 38882682 PMCID: PMC11176561 DOI: 10.1016/j.csbj.2024.05.020] [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: 12/28/2023] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 06/18/2024] Open
Abstract
Antimicrobial peptides are promising therapeutic agents for treating drug-resistant bacterial disease due to their broad-spectrum antimicrobial activity and decreased susceptibility to evolutionary resistance. In this study, three novel cathelicidin antimicrobial peptides were identified from Thamnophis sirtalis, Balaenoptera musculus, and Lipotes vexillifer by protein database mining and sequence alignment and were subsequently named TS-CATH, BM-CATH, and LV-CATH, respectively. All three peptides exhibited satisfactory antibacterial activity and broad antibacterial spectra against clinically isolated E. coli, P. aeruginosa, K. pneumoniae, and A. baumannii in vitro. Among them, TS-CATH displayed the best antimicrobial/bactericidal activity, with a rapid elimination efficiency against the tested drug-resistant gram-negative bacteria within 20 min, and exhibited the lowest cytotoxicity toward mammalian cells. Furthermore, TS-CATH effectively enhanced the survival rate of mice with ceftazidime-resistant E. coli bacteremia and promoted wound healing in meropenem-resistant P. aeruginosa infection. These results were achieved through the eradication of bacterial growth in target organs and wounds, further inhibiting the systemic dissemination of bacteria and the inflammatory response. TS-CATH exhibited direct antimicrobial activity by damaging the inner and outer membranes, resulting in leakage of the bacterial contents at super-MICs. Moreover, TS-CATH disrupted the bacterial respiratory chain, which inhibited ATP synthesis and induced ROS formation, significantly contributing to its antibacterial efficacy at sub-MICs. Overall, TS-CATH has potential for use as an antibacterial agent.
Collapse
Affiliation(s)
- Jian Wang
- Institution of all authors: College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Meina Zhang
- Institution of all authors: College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Chao Li
- Institution of all authors: College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Mengyuan Liu
- Institution of all authors: College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yixin Qi
- Institution of all authors: College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Xiaolin Xie
- Institution of all authors: College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Changlin Zhou
- Institution of all authors: College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Lingman Ma
- Institution of all authors: College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| |
Collapse
|
20
|
Yu T, Hou J, Hafeez F, Ge P, Zou A, Fu Y, Zhang J, Xianyu Y. Fungus-mediated biosynthesis of gold nanoparticles with synergistic antifungal activity against multidrug-resistant Candida albicans. NANO TODAY 2024; 59:102486. [DOI: 10.1016/j.nantod.2024.102486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
21
|
Pals MJ, Lindberg A, Velema WA. Chemical strategies for antisense antibiotics. Chem Soc Rev 2024; 53:11303-11320. [PMID: 39436264 PMCID: PMC11495246 DOI: 10.1039/d4cs00238e] [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: 03/13/2024] [Indexed: 10/23/2024]
Abstract
Antibacterial resistance is a severe threat to modern medicine and human health. To stay ahead of constantly-evolving bacteria we need to expand our arsenal of effective antibiotics. As such, antisense therapy is an attractive approach. The programmability allows to in principle target any RNA sequence within bacteria, enabling tremendous selectivity. In this Tutorial Review we provide guidelines for devising effective antibacterial antisense agents and offer a concise perspective for future research. We will review the chemical architectures of antibacterial antisense agents with a special focus on the delivery and target selection for successful antisense design. This Tutorial Review will strive to serve as an essential guide for antibacterial antisense technology development.
Collapse
Affiliation(s)
- Mathijs J Pals
- Institute for Molecules and Materials, Radboud University Nijmegen, the Netherlands. Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands.
| | - Alexander Lindberg
- Institute for Molecules and Materials, Radboud University Nijmegen, the Netherlands. Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands.
| | - Willem A Velema
- Institute for Molecules and Materials, Radboud University Nijmegen, the Netherlands. Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands.
| |
Collapse
|
22
|
Zhai Y, Pei S, Qin X, Zhang L, Liu X, Cai S, Zhang W. Ultralow Dose Iron-Copper Bimetallic Single-Atom Nanozymes for Efficient Photothermal-Chemodynamic Antibacterial and Wound Healing. Adv Healthc Mater 2024:e2403920. [PMID: 39562175 DOI: 10.1002/adhm.202403920] [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: 10/09/2024] [Revised: 11/10/2024] [Indexed: 11/21/2024]
Abstract
The combination of photothermal and chemodynamic therapy (PTT-CDT) using single-atom nanozymes (SAzymes) shows great promise in combating pathogenic and drug-resistant bacteria. However, the photothermal conversion efficiency and catalytic activity of SAzymes with solely metal sites remain inadequate, often requiring high doses for effectiveness. Herein, a bimetallic single-atomic nanozymes with Fe and Cu active sites (FeCu BSNs) designed is reported for efficient treatment of bacterial infections through hyperthermia-amplified nanozyme catalysis strategy. The FeCu BSNs demonstrate remarkable peroxidase (POD) activity with a specific activity (SA) of 752.25 U mg-1, which is 2.3 folds larger than that of Fe SAzymes (323.45 U mg-1). Additionally, their photothermal effect achieve a photothermal conversion efficiency up to 56.26%, which is two times higher that of Fe SAzymes (29.69%) and Cu SAzymes (25.55%). These enhancements can be attributed to the hybridization of Fe and Cu sites. The FeCu BSNs-mediated PTT-CDT combination therapy demonstrates potent antibacterial effects in both in vitro and in vivo models, attributed to high levels of reactive oxygen species (ROS) generation and hyperthermia. This study effectively validates the application of ultralow-dose bimetallic single-atom nanozymes in PTT-CDT for wound healing, offering a promising approach for enhanced wound recovery.
Collapse
Affiliation(s)
- Yujie Zhai
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, China
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, School of Resources, Environment and Materials, Guangxi University, Nanning, Guangxi, 530004, China
| | - Shenglin Pei
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, China
| | - Xuhui Qin
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, China
| | - Lu Zhang
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, China
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, China
| | - Xijun Liu
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, School of Resources, Environment and Materials, Guangxi University, Nanning, Guangxi, 530004, China
| | - Shuangqi Cai
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Weiqing Zhang
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, China
- University Engineering Research Center of Oncolytic & Nanosystem Development, Nanning, Guangxi, 530021, China
| |
Collapse
|
23
|
Kumar SD, Lee JK, Radhakrishnan NK, Bang JK, Kim B, Chaudhary SC, Chelladurai A, Ganbaatar B, Kim EY, Lee CW, Yang S, Kim Y, Shin SY. Antibacterial, Antibiofilm, and Anti-inflammatory Effects of a Novel Thrombin-Derived Peptide in Sepsis Models: Insights into Underlying Mechanisms. J Med Chem 2024; 67:19791-19812. [PMID: 39475485 DOI: 10.1021/acs.jmedchem.4c02157] [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: 11/15/2024]
Abstract
We developed two short helical antimicrobial peptides, HVF18-a3 and its d-enantiomer, HVF18-a3-d, derived from the thrombin C-terminal peptide HVF18. These peptides exhibit potent antimicrobial activity against various bacteria by compromising both the outer and inner membranes, with low hemolytic activity. They are stable in the presence of physiological salts and human serum, exhibiting a low potential for developing drug resistance and excellent antibiofilm activity against Gram-negative bacteria. HVF18-a3-d also neutralized lipopolysaccharide (LPS) through direct binding interactions and suppressed the production of inflammatory cytokines through the inflammatory signaling pathway mediated by Toll-like receptor 4 in RAW264.7 cells stimulated with LPS. Both pre- and post-treatment with HVF18-a3-d significantly protected mice against fatal septic shock induced by carbapenem resistant Acinetobacter baumannii. These findings suggest HVF18-a3 and HVF18-a3-d are promising candidates for developing antibiotics against Gram-negative sepsis.
Collapse
Affiliation(s)
- S Dinesh Kumar
- Department of Cellular & Molecular Medicine, School of Medicine, Chosun University, Gwangju 61452, Republic of Korea
| | - Jin Kyeong Lee
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Seoul 05029, Republic of Korea
| | | | - Jeong Kyu Bang
- Division of Magnetic Resonance, Korea Basic Science Institute (KBSI), Ochang, Chung Buk 28119, Republic of Korea
- Dandicure Inc, Ochang, Chung Buk 28119, Republic of Korea
| | - Byeongkwon Kim
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Seoul 05029, Republic of Korea
| | - Shubhash Chandra Chaudhary
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Seoul 05029, Republic of Korea
| | - Ajish Chelladurai
- Department of Cellular & Molecular Medicine, School of Medicine, Chosun University, Gwangju 61452, Republic of Korea
| | - Byambasuren Ganbaatar
- Department of Chemistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Eun Young Kim
- Department of Cellular & Molecular Medicine, School of Medicine, Chosun University, Gwangju 61452, Republic of Korea
| | - Chul Won Lee
- Department of Chemistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Sungtae Yang
- Department of Microbiology, School of Medicine, Chosun University, Gwangju 61452, Republic of Korea
- Institute of Well-Aging Medicare & CSU G-LAMP Project Group, Chosun University, Gwangju 61452, Republic of Korea
| | - Yangmee Kim
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Seoul 05029, Republic of Korea
| | - Song Yub Shin
- Department of Cellular & Molecular Medicine, School of Medicine, Chosun University, Gwangju 61452, Republic of Korea
| |
Collapse
|
24
|
Zhang M, Yuan F, Jia H, Xu Y, Yan L, Zhang T, Xu X, Liu Y, Wang X, Li D. Rapidly in situ forming antibiotic-free injectable hydrogel wound dressing for eradicating drug-resistant bacterial infections in human skin organoids. Int J Biol Macromol 2024; 282:137542. [PMID: 39537051 DOI: 10.1016/j.ijbiomac.2024.137542] [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: 09/06/2024] [Revised: 11/04/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
The rising prevalence of global antibiotic resistance evokes the urgent requirement to explore the alternative antimicrobial candidates. It is of great significance to overcome these serious threats of multidrug-resistant bacterial infections and difficult-to-heal cutaneous wounds to human health. Herein, we proposed a rapidly in situ forming innovative antibiotic-free hydrogel dressing with excellent biocompatibility, easy injectability, strong tissue adhesion and superior antibacterial activity against drug-resistant bacteria. An octa-armed poly(ethylene glycol) amine (Octa-PEG-NH2) was quickly crosslinked with a green industrial microbicide tetrakis (hydroxymethyl) phosphonium chloride (THPC) to form an antibacterial hydrogel (OPTH) by simple mixing without any other initiators or crosslinkers. A significant broad-spectrum antibacterial efficacy was demonstrated against Gram-positive Staphylococcus aureus (S. aureus), Gram-negative Escherichia coli (E. coli) and methicillin-resistant Staphylococcus aureus (MRSA). Significantly, benefiting from its flexible injectability and reliable tissue adhesion, the excellent antibacterial performances were further evidenced by employing human-induced pluripotent stem cell (iPSC)-derived skin organoids in a 3D culture system and rat animal wound models in vivo with MRSA infection, thus allowing for reepithelization promotion and wound healing. Collectively, the findings not only propose a facile gelatinization strategy for readily accessible antibiotic-free hydrogel dressings for effective MRSA therapy but also hold great clinical translation potential in obliterating multi-pathogenic bacteria and accelerating wound healing.
Collapse
Affiliation(s)
- Mingming Zhang
- The Ninth Medical Center of Chinese PLA General Hospital, Beijing 100101, China; Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100091, China
| | - Fuzhen Yuan
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, China
| | - Huaping Jia
- The Ninth Medical Center of Chinese PLA General Hospital, Beijing 100101, China
| | - Yongjie Xu
- The Ninth Medical Center of Chinese PLA General Hospital, Beijing 100101, China
| | - Ling Yan
- The Ninth Medical Center of Chinese PLA General Hospital, Beijing 100101, China
| | - Ting Zhang
- The Ninth Medical Center of Chinese PLA General Hospital, Beijing 100101, China
| | - Xiong Xu
- The Ninth Medical Center of Chinese PLA General Hospital, Beijing 100101, China; Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China; Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100091, China
| | - Yixuan Liu
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China; Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100091, China
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Dawei Li
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100091, China.
| |
Collapse
|
25
|
Chen Z, Zhe M, Wu W, Yu P, Xiao Y, Liu H, Liu M, Xiang Z, Xing F. Injectable Photocrosslinked Hydrogel Dressing Encapsulating Quercetin-Loaded Zeolitic Imidazolate Framework-8 for Skin Wound Healing. Pharmaceutics 2024; 16:1429. [PMID: 39598552 PMCID: PMC11597769 DOI: 10.3390/pharmaceutics16111429] [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: 10/24/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Wound management is a critical component of clinical practice. Promoting timely healing of wounds is essential for patient recovery. Traditional treatments have limited efficacy due to prolonged healing times, excessive inflammatory responses, and susceptibility to infection. Methods: In this research, we created an injectable hydrogel wound dressing formulated from gelatin methacryloyl (GelMA) that encapsulates quercetin-loaded zeolitic imidazolate framework-8 (Qu@ZIF-8) nanoparticles. Next, its ability to promote skin wound healing was validated through in vitro experiments and animal studies. Results: Research conducted both in vitro and in vivo indicated that this hydrogel dressing effectively mitigates inflammation, inhibits bacterial growth, and promotes angiogenesis and collagen synthesis, thus facilitating a safe and efficient healing process for wounds. Conclusions: This cutting-edge scaffold system provides a novel strategy for wound repair and demonstrates significant potential for clinical applications.
Collapse
Affiliation(s)
- Zhao Chen
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.C.); (H.L.); (M.L.)
| | - Man Zhe
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Wenting Wu
- Department of Pediatric Surgery, Division of Orthopedic Surgery, Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Peiyun Yu
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany;
| | - Yuzhen Xiao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China;
| | - Hao Liu
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.C.); (H.L.); (M.L.)
| | - Ming Liu
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.C.); (H.L.); (M.L.)
| | - Zhou Xiang
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.C.); (H.L.); (M.L.)
| | - Fei Xing
- Department of Pediatric Surgery, Division of Orthopedic Surgery, Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610041, China;
| |
Collapse
|
26
|
Weber B, Ritchie NE, Hilker S, Chan DCK, Peukert C, Deisinger JP, Ives R, Årdal C, Burrows LL, Brönstrup M, Magolan J, Raivio TL, Brown ED. High-Throughput Discovery of Synthetic Siderophores for Trojan Horse Antibiotics. ACS Infect Dis 2024; 10:3821-3841. [PMID: 39438291 PMCID: PMC11556397 DOI: 10.1021/acsinfecdis.4c00359] [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: 05/01/2024] [Revised: 10/09/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024]
Abstract
To cause infection, bacterial pathogens must overcome host immune factors and barriers to nutrient acquisition. Reproducing these aspects of host physiology in vitro has shown great promise for antibacterial drug discovery. When used as a bacterial growth medium, human serum replicates several aspects of the host environment, including innate immunity and iron limitation. We previously reported that a high-throughput chemical screen using serum as the growth medium enabled the discovery of novel growth inhibitors overlooked by conventional screens. Here, we report that a subset of compounds from this high-throughput serum screen display an unexpected growth enhancing phenotype and are enriched for synthetic siderophores. We selected 35 compounds of diverse chemical structure and quantified their ability to enhance bacterial growth in human serum. We show that many of these compounds chelate iron, suggesting they were acting as siderophores and providing iron to the bacteria. For two different pharmacophores represented among these synthetic siderophores, conjugation to the β-lactam antibiotic ampicillin imparted iron-dependent enhancement in antibacterial activity. Conjugation of the most potent growth-enhancing synthetic siderophore with the monobactam aztreonam produced MLEB-22043, a broad-spectrum antibiotic with significantly improved activity against Klebsiella pneumoniae, Escherichia coli, Acinetobacter baumannii, and Pseudomonas aeruginosa. This synthetic siderophore-monobactam conjugate uses multiple TonB-dependent transporters for uptake into P. aeruginosa. Like aztreonam, MLEB-22043 demonstrated activity against metallo-β-lactamase expressing bacteria, and, when combined with the β-lactamase inhibitor avibactam, was active against clinical strains coexpressing the NDM-1 metallo-β-lactamase and serine β-lactamases. Our work shows that human serum is an effective bacterial growth medium for the high-throughput discovery of synthetic siderophores, enabling the development of novel Trojan Horse antibiotics.
Collapse
Affiliation(s)
- Brent
S. Weber
- Department
of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Michael
G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S
4L8, Canada
- Department
of Biological Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Nikki E. Ritchie
- Department
of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Michael
G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S
4L8, Canada
| | - Simon Hilker
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Derek C. K. Chan
- Department
of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Michael
G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S
4L8, Canada
| | - Carsten Peukert
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Julia P. Deisinger
- Department
of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Michael
G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S
4L8, Canada
| | - Rowan Ives
- Department
of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Michael
G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S
4L8, Canada
| | - Christine Årdal
- Antimicrobial
Resistance Centre, Norwegian Institute of
Public Health, 0213 Oslo, Norway
| | - Lori L. Burrows
- Department
of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Michael
G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S
4L8, Canada
| | - Mark Brönstrup
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research Inhoffenstraße 7, 38124 Braunschweig, Germany
- German
Center for Infection Research (DZIF), Site
Hannover-Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany
- Institute
for Organic Chemistry (IOC), Leibniz Universität
Hannover, Schneiderberg
1B, 30167 Hannover, Germany
| | - Jakob Magolan
- Department
of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Michael
G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S
4L8, Canada
| | - Tracy L. Raivio
- Department
of Biological Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Eric D. Brown
- Department
of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Michael
G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S
4L8, Canada
| |
Collapse
|
27
|
NANAWARE RAJESHB, CHABUKSWAR ANURUDDHAR, ADSULE PRAJAKTAV, JAGDALE SWATIC, RAUT KUNALG. DEVELOPMENT OF 1H-INDAZOLE DERIVATIVES AS ANTI-INFLAMMATORY AGENTS USING COMPUTATIONAL METHODS. INTERNATIONAL JOURNAL OF APPLIED PHARMACEUTICS 2024:299-304. [DOI: 10.22159/ijap.2024v16i6.51634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Objective: Due to the rising prevalence of disorders linked to inflammation, there is a greater emphasis on the discovery and development of anti-inflammatory drugs, with a focus on producing new structural compounds.
Methods: In this research, molecular docking and Molecular Dynamics (MD) simulation study were carried out to evaluate the 1H-indazole analogs as potent anti-inflammatory agents.
Results: The compounds containing difluorophenyl, para-toulene and 4-methoxyphenyl group shows significant binding results (9.11, 8.80 and 8.46 kcal/mol respectively) when docked with Cyclooxygenase-2 (COX-2) enzyme 3NT1. The results of the MD simulation indicated that test compound BDF was relatively stable in the COX-2 enzymes active sites. The compound BDF-3NT1 demonstrated substantial affinities for binding with all of its aimed targets following a dynamic Molecular Mechanics with Generalized Born Surface Area (MM-GBSA) analysis.
Conclusion: In accordance to this study, newly developed 1H-indazole compounds have the potential for treating inflammation.
Collapse
|
28
|
Sun R, Zhao D, Yu X, Zhang F, You R, Luo X, Li L. Discovery of a family of menaquinone-targeting cyclic lipodepsipeptides for multidrug-resistant Gram-positive pathogens. Commun Biol 2024; 7:1453. [PMID: 39506023 PMCID: PMC11541763 DOI: 10.1038/s42003-024-07159-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Menaquinone (MK) in bacterial membrane is an attractive target for the development of novel therapeutic agents. Mining the untapped chemical diversity encoded by Gram-negative bacteria presents an opportunity to identify additional MK-binding antibiotics (MBAs). By MK-binding motif searching of bioinformatically predicted linear non-ribosomal peptides from 14,298 sequenced genomes of 45 underexplored Gram-negative bacterial genera, here we identify a novel MBA structural family, including silvmeb and pseudomeb, using structure prediction-guided chemical synthesis. Both MBAs show rapid bacteriolysis by MK-dependent membrane depolarization to achieve their potent activities against a panel of Gram-positive pathogens. Furthermore, both MBAs are proven to be effective against methicillin-resistant Staphylococcus aureus in a murine peritonitis-sepsis model. Our findings suggest that MBAs are a kind of structurally diverse and still underexplored antibacterial lipodepsipeptide class. The interrogation of underexplored bacterial taxa using synthetic bioinformatic natural product methods is an appealing strategy for discovering novel biomedically relevant agents to confront the crisis of antimicrobial resistance.
Collapse
Affiliation(s)
- Runze Sun
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Shanghai, 200240, China
| | - Di Zhao
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Shanghai, 200240, China
| | - Xuchang Yu
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Shanghai, 200240, China
- State Key Laboratory of Bioreactor Engineering and School of Biotechnology, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China
| | - Fei Zhang
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Shanghai, 200240, China
- College of Life Science, Tarim University, E1487 Tarim Avenue, Alar, 43300, China
| | - Ruixiang You
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Shanghai, 200240, China
| | - Xiaoxia Luo
- College of Life Science, Tarim University, E1487 Tarim Avenue, Alar, 43300, China
| | - Lei Li
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Shanghai, 200240, China.
| |
Collapse
|
29
|
Tooke AK, Hodges RE, Pyrah JF, Bayles KW, Renshaw SA, Foster SJ. Tetracycline and Oxacillin Act Synergistically on Biofilms and Display Increased Efficacy In Vivo Against Staphylococcus aureus. Curr Microbiol 2024; 81:447. [PMID: 39505760 PMCID: PMC11541413 DOI: 10.1007/s00284-024-03959-4] [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: 07/04/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024]
Abstract
Oxacillin (bactericidal) and tetracycline (bacteriostatic) are clinically relevant antibiotics that are routinely prescribed to treat Staphylococcus aureus infections but not conventionally used in combination. There is an urgent need for treatment regimens that can act upon biofilms during infection, associated with chronic infections on indwelling devices, as well as acute planktonic (systemic) infection. Here we show that in an in vitro model oxacillin and tetracycline act synergistically against S. aureus UAMS-1 biofilms, reducing the concentration of both antibiotics necessary to eradicate an established biofilm. Using an in vivo zebrafish larval infection model with S. aureus NewHG, they display improved bacterial clearance compared to each drug alone and can counteract a loss of host phagocytes, an important innate defence against S. aureus. In these cases, the bacteriostatic nature of tetracycline enhances rather than dampens the bactericidal action of oxacillin, although an exact mechanism remains to be elucidated. We suggest a dual therapy could be of clinical use against biofilm-forming S. aureus and has a potential use in patients with a compromised immune system.
Collapse
Affiliation(s)
- Amy K Tooke
- School of Biosciences, University of Sheffield, Sheffield, S10 2TH, UK
- Microbes in Health and Disease Theme, Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Rebecca E Hodges
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Josie F Pyrah
- School of Biosciences, University of Sheffield, Sheffield, S10 2TH, UK
| | - Kenneth W Bayles
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Stephen A Renshaw
- The Bateson Centre, University of Sheffield, Western Bank, Sheffield, S10 2TH, UK
- School of Medicine and Population Health, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Simon J Foster
- School of Biosciences, University of Sheffield, Sheffield, S10 2TH, UK.
- Florey Institute, University of Sheffield, Sheffield, S10 2TH, UK.
| |
Collapse
|
30
|
Deng W, Xue RY, Xiao SX, Wang JT, Liao XW, Yu RJ, Xiong YS. Discovery of quaternized pyridine-thiazole-ruthenium complexes as potent anti-Staphylococcus aureus agents. Eur J Med Chem 2024; 277:116712. [PMID: 39106657 DOI: 10.1016/j.ejmech.2024.116712] [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: 05/21/2024] [Revised: 07/03/2024] [Accepted: 07/23/2024] [Indexed: 08/09/2024]
Abstract
Quaternization of ruthenium complexes may be a promising strategy for the development of new antibiotics. In response to the increasing bacterial resistance, we integrated the quaternary amine structure into the design of ruthenium complexes and evaluated their antibacterial activity. All the ruthenium complexes showed good antibacterial activity against the tested Staphylococcus aureus (S. aureus). Ru-8 was the most effective antibacterial agent that displayed excellent antibacterial activity against S. aureus (MIC = 0.78-1.56 μg/mL). In vitro experiments showed that all nine ruthenium complexes had low hemolytic toxicity to rabbit erythrocytes. Notably, Ru-8 was found to disrupt bacterial cell membranes, alter their permeability, and induce ROS production in bacteria, all the above leading to the death of bacteria without inducing drug resistance. To further explore the antibacterial activity of Ru-8in vivo, we established a mouse skin wound infection model and a G. mellonella larvae infection model. Ru-8 exhibited significant antibacterial efficacy against S. aureus in vivo and low toxicity to mouse tissues. The Ru-8 showed low toxicity to Raw264.7 cells (mouse monocyte macrophage leukemia cells). This study indicates that the ruthenium complex ruthenium quaternary was a promising strategy for the development of new antibacterial agents.
Collapse
Affiliation(s)
- Wei Deng
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - Run-Yu Xue
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - Su-Xin Xiao
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - Jin-Tao Wang
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - Xiang-Wen Liao
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - Ru-Jian Yu
- School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, 330013, China.
| | - Yan-Shi Xiong
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China.
| |
Collapse
|
31
|
Badgujar P, Malik AK, Mehata AK, Setia A, Verma N, Randhave N, Shukla VN, Kande V, Singh P, Tiwari P, Mahto SK, Muthu MS. Polyvinyl alcohol-chitosan based oleanolic acid nanofibers against bacterial infection: In vitro studies and in vivo evaluation by optical and laser Doppler imaging modalities. Int J Biol Macromol 2024; 279:135532. [PMID: 39265903 DOI: 10.1016/j.ijbiomac.2024.135532] [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: 05/11/2024] [Revised: 07/26/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
The present work focuses on the fabrication of polyvinyl alcohol-chitosan-loaded oleanolic acid-nanofibers (PVA-CS-OLA-NFs) for bacterial infection. The prepared PVA-CS-OLA-NFs were characterized for contact angle, SEM, AFM, XRD, FTIR, and TGA. The solid-state characterization and in vitro performance evaluation of nanofibers reveal consistent interconnection and diameters ranging from 102 ± 9.5 to 386 ± 11.6 nm. The nanofibers have a flat surface topography and exhibit efficient drug entrapment. Moreover, the in vitro release profile of PVA-CS-OLA-NFs was found to be 51.82 ± 1.49 % at 24 h. Furthermore, the hemocompatibility study showed that the developed PVA-CS-OLA-NFs are non-hemolytic to human blood. The PVA-CS-OLA-NFs demonstrate remarkable antibacterial capabilities, as evidenced by their MBC and MIC values, which range from 128 and 32 μg/mL, against the strains of S. aureus. The in-vivo fluorescence optical imaging showed the sustained PVA-CS-OLA-NFs release at the wound site infected with S. aureus for a longer duration of time. Moreover, the PVA-CS-OLA-NFs showed superior wound healing performance against S. aureus infected wounds compared to the marketed formulation. Further, the laser Doppler imaging system improved oxygen saturation, blood supply, and wound healing by providing real-time blood flow and oxygen saturation information.
Collapse
Affiliation(s)
- Paresh Badgujar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Ankit Kumar Malik
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Abhishesh Kumar Mehata
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Aseem Setia
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Nidhi Verma
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Nandini Randhave
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Vishwa Nath Shukla
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Vilas Kande
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Priya Singh
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Punit Tiwari
- Department of Microbiology, Institute of Medical Sciences, BHU, Varanasi 221005, Uttar Pradesh, India
| | - Sanjeev Kumar Mahto
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Madaswamy S Muthu
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India.
| |
Collapse
|
32
|
Cai D, Ye Y, Song J, Liu S, Zeng X, Zhu B, Tao Y, Cheng J, Yang Y, Zhang Y, Zou Q, Guo Y, Sun H, Zeng H. A chitosan-modified tea tree oil self-nanoemulsion improves antibacterial effects in vivo and in vitro and promotes wound healing by influencing metabolic processes against multidrug-resistant bacterial infections. Int J Biol Macromol 2024; 281:136404. [PMID: 39389511 DOI: 10.1016/j.ijbiomac.2024.136404] [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: 04/18/2024] [Revised: 09/18/2024] [Accepted: 10/06/2024] [Indexed: 10/12/2024]
Abstract
Infectious diseases, especially multidrug-resistant bacterial infections, have caused crises and majorly disrupted public health and economic stability worldwide. Many natural essential oils, especially tea tree oil, have potential to treat multidrug-resistant bacteria, such as H. pylori and P. aeruginosa. However, there are some problems need to be solved, such as poor stability upon light or oxygen exposure. Therefore, it is urgent to develop the ideal formation to tackle these difficulties. Herein, we reported the novel chitosan-modified self-nanoemulsion (TNE) encapsulating natural essential tea tree oil with strong antibacterial and stability characterize. In this study, we found that this self-nanoemulsion (size: 212 nm, PDI: 0.124, zeta potential: -20.5 mV, 6 % tea tree oil) not only had physical properties, good stability and tissue safety, but also had better antibacterial synergism (2-8 times) than that of water suspension against various multidrug-resistant bacterial (such as H. pylori, MRSA and P. aeruginosa). Additionally, TNE showed high antibacterial effectiveness in vivo, reduced inflammation, promoted ulcer healing after H. pylori infection and accelerated wound healing after P. aeruginosa infection. Importantly, this novel self-nanoemulsion can induce 274 protein down-regulated and 251 protein up-regulated, and disrupt H. pylori metabolic processes (glyoxylate, dicarboxylic acid, glutamate and tryptophan metabolism) and reduced its viability, leading to significant synergistic antibacterial effects. TNE is a potential treatment for skin wounds or ulcers caused by multidrug-resistant bacterial infections.
Collapse
Affiliation(s)
- Dingyi Cai
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China; Department of Stomatology, The 79th Group Army Hospital of PLA, Liaoyang, Liaoning 111000, China
| | - Yan Ye
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Jianye Song
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Shulin Liu
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Xiaoqiang Zeng
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Baohang Zhu
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Yikun Tao
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Jingjing Cheng
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Yun Yang
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Yi Zhang
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Quanming Zou
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Ying Guo
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China; Department of Clinical Laboratory, Affiliated Banan Hospital of Chongqing Medical University, Chongqing 401320, China.
| | - Hongwu Sun
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China.
| | - Hao Zeng
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
33
|
Cai W, Song Y, Xie Q, Wang S, Yin D, Wang S, Wang S, Zhang R, Lee M, Duan J, Zhang X. Dual osmotic controlled release platform for antibiotics to overcome antimicrobial-resistant infections and promote wound healing. J Control Release 2024; 375:627-642. [PMID: 39284525 DOI: 10.1016/j.jconrel.2024.09.022] [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: 06/18/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 09/26/2024]
Abstract
Methicillin-Resistant Staphylococcus aureus forming into biofilms can trigger chronic inflammation and disrupt skin wound healing processes. Prolonged and excessive use of antibiotics can expedite the development of resistance, primarily because of their limited ability to penetrate microbial membranes and biofilms, especially antibiotics with intracellular drug targets. Herein, we devise a strategy in which virus-inspired nanoparticles control the release of antibiotics through rapid penetration into both bacterial cells and biofilms, thereby combating antimicrobial-resistant infections and promoting skin wound healing. Lipid-based nanoparticles based on stearamine and cholesterol were designed to mimic viral highly ordered nanostructures. To mimic the arginine-rich fragments in viral protein transduction domains, the primary amines on the surface of the lipid-based nanoparticles were exchanged by guanidine segments. Levofloxacin, an antibiotic that inhibits DNA replication, was chosen as the model drug to be incorporated into nanoparticles. Hyaluronic acid was coated on the surface of nanoparticles acting as a capping agent to achieve bacterial-specific degradation and guanidine explosion in the bacterial microenvironment. Our virus-inspired nanoparticles displayed long-acting antibacterial effects and powerful biofilm elimination to overcome antimicrobial-resistant infections and promote skin wound healing. This work demonstrates the ability of virus-inspired nanoparticles to achieve a dual penetration of microbial cell membranes and biofilm structures to address antimicrobial-resistant infections and trigger skin wound healing.
Collapse
Affiliation(s)
- Wanni Cai
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510000, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China
| | - Yan Song
- Department of Pharmacy, Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Qing Xie
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China
| | - Shiyu Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China
| | - Donghong Yin
- Department of Pharmacy, Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Shuyun Wang
- Department of Pharmacy, Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Song Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China
| | - Rui Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China
| | - Min Lee
- Division of Oral and Systemic Health Sciences, University of California at Los Angeles, Los Angeles, CA 90095, USA.
| | - Jinju Duan
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Department of Pharmacy, Second Hospital of Shanxi Medical University, Taiyuan 030001, China.
| | - Xiao Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Department of Pharmacy, Second Hospital of Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
34
|
Zong L, Teng R, Zhang H, Liu W, Feng Y, Lu Z, Zhou Y, Fan Z, Li M, Pu X. Ultrasound-Responsive HBD Peptide Hydrogel with Antibiofilm Capability for Fast Diabetic Wound Healing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406022. [PMID: 39248340 PMCID: PMC11558141 DOI: 10.1002/advs.202406022] [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: 06/01/2024] [Revised: 07/26/2024] [Indexed: 09/10/2024]
Abstract
Despite advancements in therapeutic agents for diabetic chronic wounds, challenges such as suboptimal bioavailability, intricate disease milieus, and inadequate delivery efficacy have impeded treatment outcomes. Here, ultrasound-responsive hydrogel incorporated with heparin-binding domain (HBD) peptide nanoparticles is developed to promote diabetic wound healing. HBD peptide, derived from von Willebrand Factor with angiogenic activity, are first engineered to self-assemble into nanoparticles with enhanced biostability and bioavailability. Ultrasound responsive cargo release and hydrogel collapses are first verified through breakage of crosslinking. In addition, desired antioxidant and antibacterial activity of such hydrogel is observed. Moreover, the degradation of hydrogel under ultrasound stimulation into smaller fragments facilitated the deeper wound penetration of ≈400 µm depth. Complete wound closure is observed from diabetic mice with chronic wounds after being treated with the proposed hydrogel. In detail, in vivo studies revealed that hydrogels loaded with HBD peptide nanoparticles increased the levels of angiogenesis-related growth factors (VEGF-A, CD31, and α-SMA) to effectively accelerate wound repair. Overall, this study demonstrates that ultrasound-responsive HBD peptide hydrogel provides a synergistic therapeutic strategy for external biofilm elimination and internal effective delivery for diabetic wounds with biofilm infection.
Collapse
Affiliation(s)
- Lanlan Zong
- State Key Laboratory of Antiviral DrugsHenan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River BasinSchool of PharmacyHenan UniversityN. Jinming Ave.Kaifeng475004P. R. China
| | - Runxin Teng
- Department of Polymeric MaterialsSchool of Materials Science and EngineeringTongji University4800 CaoanRoadShanghai201804P. R. China
| | - Huiqi Zhang
- State Key Laboratory of Antiviral DrugsHenan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River BasinSchool of PharmacyHenan UniversityN. Jinming Ave.Kaifeng475004P. R. China
| | - Wenshang Liu
- State Key Laboratory of Antiviral DrugsHenan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River BasinSchool of PharmacyHenan UniversityN. Jinming Ave.Kaifeng475004P. R. China
- Department of DermatologyShanghai Children's Medical CenterShanghai Jiaotong University School of MedicineShanghai200127P. R. China
| | - Yu Feng
- State Key Laboratory of Antiviral DrugsHenan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River BasinSchool of PharmacyHenan UniversityN. Jinming Ave.Kaifeng475004P. R. China
| | - Zhengmao Lu
- Department of Gastrointestinal SurgeryThe First Affiliated Hospital of Naval Medical UniversityShanghai200433P. R. China
| | - Yuxiao Zhou
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain‐Like IntelligenceShanghai Fourth People's Hospital, School of MedicineTongji UniversityShanghai200434P. R. China
| | - Zhen Fan
- Department of Polymeric MaterialsSchool of Materials Science and EngineeringTongji University4800 CaoanRoadShanghai201804P. R. China
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain‐Like IntelligenceShanghai Fourth People's Hospital, School of MedicineTongji UniversityShanghai200434P. R. China
| | - Meng Li
- Department of DermatologyShanghai Children's Medical CenterShanghai Jiaotong University School of MedicineShanghai200127P. R. China
| | - Xiaohui Pu
- State Key Laboratory of Antiviral DrugsHenan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River BasinSchool of PharmacyHenan UniversityN. Jinming Ave.Kaifeng475004P. R. China
| |
Collapse
|
35
|
Wang H, Sun S, Zhao Y, Wang P, Zhou Y, Sun H, Yang J, Cheng K, Li S, Lin H. Carbon Dots with Integrated Photothermal Antibacterial and Heat-Enhanced Antioxidant Properties for Diabetic Wound Healing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403160. [PMID: 39051538 DOI: 10.1002/smll.202403160] [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: 04/19/2024] [Revised: 06/26/2024] [Indexed: 07/27/2024]
Abstract
Diabetic wounds pose a persistent challenge due to their slow healing nature, primarily caused by bacterial infection and excessive reactive oxygen species (ROS)-induced inflammation. In this study, carbon dots with synergistic antibacterial and antioxidant properties, referred to as AA-CDs, are developed specifically for diabetic wound healing using a straightforward solvothermal method. By utilizing cost-effective precursors like citric acid and ascorbic acid, AA-CDs are engineered to possess tailored functions of photothermal sterilization and ROS scavenging. The resulting AA-CDs demonstrats broad-spectrum antibacterial activity, particularly against multidrug-resistant strains, along with efficient ROS scavenging both in solution and within cells. Additionally, AA-CDs exhibits a protective effect against oxidative stress-induced damage. Notably, with a high photothermal conversion efficiency (41.18%), AA-CDs displays heat-enhanced antioxidant performance, providing not only augmented ROS scavenging but also additional protection against oxidative stress, yielding a true "1 + 1 > 2" effect. To facilitate their use in vivo, AA-CDs are incorporated into a thermally responsive hydrogel, which exhibits evident anti-inflammatory properties by modulating inflammatory factors and significantly promots the healing of diabetic wounds. This study underscores the value of integrated platforms for diabetic wound healing and highlights the potential of versatile CDs as promising therapeutic agents in biomedical applications.
Collapse
Affiliation(s)
- Henggang Wang
- International Joint Research Center for Photo-responsive Molecules and Materials School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Shan Sun
- International Joint Research Center for Photo-responsive Molecules and Materials School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Ye Zhao
- International Joint Research Center for Photo-responsive Molecules and Materials School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Peng Wang
- Department of radiology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Yonghua Zhou
- Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu, 214122, China
| | - Haoyi Sun
- International Joint Research Center for Photo-responsive Molecules and Materials School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Jin Yang
- International Joint Research Center for Photo-responsive Molecules and Materials School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Ke Cheng
- International Joint Research Center for Photo-responsive Molecules and Materials School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Si Li
- International Joint Research Center for Photo-responsive Molecules and Materials School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Hengwei Lin
- International Joint Research Center for Photo-responsive Molecules and Materials School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
36
|
Sun J, Wang X, Gao Y, Li S, Hu Z, Huang Y, Fan B, Wang X, Liu M, Qiao C, Zhang W, Wang Y, Ji X. H 2S scavenger as a broad-spectrum strategy to deplete bacteria-derived H 2S for antibacterial sensitization. Nat Commun 2024; 15:9422. [PMID: 39482291 PMCID: PMC11527999 DOI: 10.1038/s41467-024-53764-7] [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: 06/12/2024] [Accepted: 10/18/2024] [Indexed: 11/03/2024] Open
Abstract
Bacteria-derived H2S plays multifunctional protective roles against antibiotics insult, and the H2S biogenesis pathway is emerging as a viable target for the antibacterial adjuvant design. However, the development of a pan-inhibitor against H2S-synthesizing enzymes is challenging and underdeveloped. Herein, we propose an alternative strategy to downregulate the H2S levels in H2S-producing bacteria, which depletes the bacteria-derived H2S chemically by H2S scavengers without acting on the synthesizing enzymes. After the screening of chemically diversified scaffolds and a structural optimization campaign, a potent and specific H2S scavenger is successfully identified, which displays efficient H2S depletion in several H2S-producing bacteria, potentiates both bactericidal agents and photodynamic therapy, enhances the bacterial clearance of macrophages and polymorphonuclear neutrophils, disrupts the formation of bacterial biofilm and increases the sensitivity of bacterial persister cells to antibiotics. Most importantly, such an H2S scavenger exhibits sensitizing effects with gentamicin in Pseudomonas aeruginosa -infected pneumonia and skin wound female mouse models. In aggregate, our results not only provide an effective strategy to deplete bacteria-derived H2S and establish the H2S biogenesis pathway as a viable target for persisters and drug-resistant bacteria, but also deliver a promising antibacterial adjuvant for potential clinical translation.
Collapse
Affiliation(s)
- Jiekai Sun
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Xu Wang
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Ye Gao
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Shuangyu Li
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Ziwei Hu
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Yan Huang
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Baoqiang Fan
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong, China
| | - Xia Wang
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Miao Liu
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Chunhua Qiao
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Wei Zhang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong, China.
| | - Yipeng Wang
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China.
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, China.
| | - Xingyue Ji
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
37
|
Yi J, Pei C, Zhang T, Qin Q, Gu X, Li Y, Ruan D, Wan J, Qiao L. Nanoscale Multipatterning Zn,Co-ZIF@FeOOH for Eradication of Multidrug-Resistant Bacteria and Antibacterial Treatment of Wounds. ACS APPLIED MATERIALS & INTERFACES 2024; 16:58217-58225. [PMID: 39435754 DOI: 10.1021/acsami.4c10935] [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: 10/23/2024]
Abstract
The rising incidence of infections caused by multidrug-resistant bacteria highlights the urgent need for innovative bacterial eradication strategies. Metal ions, such as Zn2+ and Co2+, have bactericidal effects by disrupting bacterial cell membranes and interfering with essential cellular processes. This has led to increased attention toward metal-organic frameworks (MOFs) as potential nonantibiotic bactericidal agents. However, the uniform and enhanced localized release of bactericidal metal ions remains a challenge. Herein, we introduce a nanoscale multipatterned Zn,Co-ZIF@FeOOH, featuring a multipod-like morphology with spiky corners, and dual-bactericidal metal ions. Compared to pure Zn,Co-ZIF, the multipod-like morphology of Zn,Co-ZIF@FeOOH exhibits enhanced adhesion toward bacterial surfaces via topological and multiple interactions of electrostatic interaction, significantly increasing the local release of Zn2+ and Co2+. Additionally, the spiky corners of the spindle-shaped FeOOH nanorods physically penetrate bacterial membranes, causing damage and further enhancing adhesion to bacteria. Nine Gram-negative and one Gram-positive bacteria were selected for in vitro test. Notably, the nanoscale multipatterned Zn,Co-ZIF@FeOOH exhibited high bactericidal efficacy against various multidrug-resistant bacteria, including extended-spectrum β-lactamase-producing (ESBL+) bacteria and carbapenem-resistant bacteria, performing well in both acidic and neutral environments. The wound healing activity of Zn,Co-ZIF@FeOOH was further demonstrated using female Balb/c mouse models infected with bacteria, where the materials show robust antibacterial efficacy and commendable biocompatibility. This study showcases the assembly of metal oxide/MOF composites for nanoscale multipatterning, aims at synergistic bacterial eradication and offers insights into developing nanomaterial-based strategies against multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Jia Yi
- Minhang Hospital, and Department of Chemistry, Fudan University, Shanghai 200000, China
| | - Congcong Pei
- School of Chemistry, Zhengzhou University, Zhengzhou 450001, China
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Tangming Zhang
- Minhang Hospital, and Department of Chemistry, Fudan University, Shanghai 200000, China
| | - Qin Qin
- Changhai Hospital, The Naval Military Medical University, Shanghai 200433, China
| | - Xiaoxia Gu
- Minhang Hospital, and Department of Chemistry, Fudan University, Shanghai 200000, China
| | - Yekan Li
- Minhang Hospital, and Department of Chemistry, Fudan University, Shanghai 200000, China
| | - Danping Ruan
- Minhang Hospital, and Department of Chemistry, Fudan University, Shanghai 200000, China
| | - Jingjing Wan
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Liang Qiao
- Minhang Hospital, and Department of Chemistry, Fudan University, Shanghai 200000, China
| |
Collapse
|
38
|
Li M, Wang C, Yu Q, Chen H, Ma Y, Wei L, Wu MX, Yao M, Lu M. A wearable and stretchable dual-wavelength LED device for home care of chronic infected wounds. Nat Commun 2024; 15:9380. [PMID: 39477919 PMCID: PMC11525593 DOI: 10.1038/s41467-024-53579-6] [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: 06/18/2024] [Accepted: 10/16/2024] [Indexed: 11/02/2024] Open
Abstract
Phototherapy can offer a safe and non-invasive solution against infections, while promoting wound healing. Conventional phototherapeutic devices are bulky and limited to hospital use. To overcome these challenges, we developed a wearable, flexible red and blue LED (r&bLED) patch controlled by a mobile-connected system, enabling safe self-application at home. The patch exhibits excellent skin compatibility, flexibility, and comfort, with high safety under system supervision. Additionally, we synthesized a sprayable fibrin gel (F-gel) containing blue light-sensitive thymoquinone and red light-synergistic NADH. Combined with bLED, thymoquinone eradicated microbes and biofilms within minutes, regardless of antibiotic resistance. Furthermore, NADH and rLED synergistically improved macrophage and endothelial cell mitochondrial function, promoting wound healing, reducing inflammation, and enhancing angiogenesis, as validated in infected diabetic wounds in mice and minipigs. This innovative technology holds great promise for revolutionizing at-home phototherapy for chronic infected wounds.
Collapse
Affiliation(s)
- Ming Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chenxi Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qiang Yu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Haoyi Chen
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yingying Ma
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Li Wei
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Mei X Wu
- Wellman Center for Photomedicine, Massachusetts General Hospital Department of Dermatology, Harvard Medical School, 50 Blossom Street, Boston, MA, 02114, USA.
| | - Min Yao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Min Lu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
39
|
Yan D, Kong H, Qu Y, Li R, Ampomah-Wireko M, Liu J, Qin S, Wang Z, Li W, Zhang E. Development of Phloroglucinol-Linked Tris-Quaternary Ammonium Salt Antimicrobial Peptide Mimics with Low Cytotoxicity and Broad-Spectrum Antibacterial Activity. J Med Chem 2024; 67:18576-18592. [PMID: 39376107 DOI: 10.1021/acs.jmedchem.4c01935] [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: 10/09/2024]
Abstract
Encouraged by the significantly different toxicities and antibacterial activities of diverse linkers, such as alkyl and aromatic nuclei linkers, and the unique structure of phloroglucinol, we synthesized a series of tris-quaternary ammonium salt (tris-QAS) antibacterial peptide mimics based on the marketed drug phloroglucinol. Among them, 2f displayed excellent activity against Staphylococcus aureus (MIC = 0.5 μg/mL) and high selectivity (SI > 2560). Surprisingly, the cytotoxicity of 2f (CC50 = 152.7 μg/mL) was dramatically better than those of alkyl QAS I and hydroquinone QAS II. Additionally, 2f possessed rapid bactericidal capability, was not prone to inducing bacterial resistance, and also exhibited excellent activity against S. aureus biofilms and persistent bacteria. Mechanistic research and transcriptome analysis revealed that 2f can interfere with the normal metabolism of bacterial cells, and it can specifically bind with phosphatidylglycerol to destroy the cell membrane. Importantly, 2f exhibited potent in vivo antibacterial activity in a mouse subcutaneous methicillin-resistant S. aureus (MRSA) infection model.
Collapse
Affiliation(s)
- Dachao Yan
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Hongtao Kong
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Ye Qu
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Ruirui Li
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Maxwell Ampomah-Wireko
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Jifeng Liu
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Shangshang Qin
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Zhenya Wang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Wen Li
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - En Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
- Pingyuan Laboratory (Zhengzhou University), Zhengzhou 450001, China
| |
Collapse
|
40
|
Wu D, Du X, Xue Q, Zhou J, Ping K, Cao Y, Liu S, Zhu Q. Supramolecular Porphyrin Photosensitizers Based on Host-Guest Recognition for In Situ Bacteria-Responsive Near-Infrared Photothermal Therapy. Adv Healthc Mater 2024:e2401662. [PMID: 39388515 DOI: 10.1002/adhm.202401662] [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: 05/06/2024] [Revised: 09/30/2024] [Indexed: 10/12/2024]
Abstract
Antibiotic resistance resulting from the overuse of antibiotics sets a high challenge for brutal antimicrobial treatment. Although photothermal therapy (PTT) overcomes the awkward situation of antibiotic resistance, it usually mistakenly kills the beneficial bacteria strains when eliminating pernicious bacteria. Specifically recognizing and damaging the target pathogens is urgently required for PTT-mediated sterilization strategy. Based on the host-guest recognition between cucurbit[10]uril (CB[10]) and porphyrins, two water-soluble supramolecular porphyrins are designed and implement selective bactericidal effect via in situ bacteria-responsive near-infrared (NIR) PTT. With the help of CB[10], the π-π stacking and hydrophobic interactions of porphyrins are efficiently inhibited, thus contributing to a good photostability and a high photothermal conversion efficiency. Attributing to the matching reduction potential between facultative anaerobic Escherichia coli (E. coli) and porphyrins, they are selectively in situ reduced into supramolecular phlorin and supramolecular chlorin by E. coli, successfully achieving a selective sterilization against E. coli. In vivo, the in situ bacteria-responsive NIR PTT systems also promote the quick recovery of E. coli-infected abscesses and trauma on mice without inducing obvious systemic toxicity, providing a new alternative to the current antibiotics and helping relieve the global public health crisis of abusive antibiotics.
Collapse
Affiliation(s)
- Dan Wu
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, 310014, P. R. China
| | - Xianlong Du
- Bethune First Clinical Medical College, Jilin University, Changchun, 130012, P. R. China
| | - Qiangqiang Xue
- Shanxi Provincial Department of Science and Technology, Taiyuan, 030021, P. R. China
| | - Jie Zhou
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, 310014, P. R. China
| | - Kunmin Ping
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, 310014, P. R. China
| | - Yibin Cao
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, 310014, P. R. China
| | - Shuang Liu
- Emergency Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, P. R. China
| | - Qing Zhu
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology Hangzhou, Hangzhou, 310014, P. R. China
| |
Collapse
|
41
|
Singh V, Nandanwar H. IMT-P8 potentiates Gram-positive specific antibiotics in intrinsically resistant Gram-negative bacteria. Antimicrob Agents Chemother 2024; 68:e0075324. [PMID: 39235250 PMCID: PMC11459922 DOI: 10.1128/aac.00753-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/06/2024] [Indexed: 09/06/2024] Open
Abstract
Gram-negative bacteria (GNB) pose a major global public health challenge as they exhibit a remarkable level of resistance to antibiotics. One of the factors responsible for promoting resistance against a wide range of antibiotics is the outer membrane (OM) of Gram-negative bacteria. The OM acts as a barrier that prevents the entry of numerous antibiotics by reducing their influx (due to membrane impermeability) and enhancing their efflux (with the help of efflux pumps). Our study focuses on analyzing the effect of IMT-P8, a cell-penetrating peptide (CPP), to enhance the influx of various Gram-positive specific antibiotics in multi-drug resistant Gram-negative pathogens. In the mechanistic experiments, IMT-P8 permeabilizes the OM at the same concentrations at which it enhances the activity of various antibiotics against GNB. Cytoplasmic membrane permeabilization was also observed at these concentrations, indicating that IMT-P8 acts on both the outer and cytoplasmic membranes. IMT-P8 interferes with the intrinsic resistance mechanism of GNB and has the potential to make Gram-positive specific antibiotics effective against GNB. IMT-P8 extends the post-antibiotic effect and in combination with antibiotics shows anti-persister activity. The IMT-P8/fusidic acid combination is effective in eliminating intracellular pathogens. IMT-P8 with negligible toxicity displayed good efficacy in murine lung and thigh infection models. Based on these findings, IMT-P8 is a potential antibiotic adjuvant to treat Gram-negative bacterial infections that pose a health hazard.
Collapse
Affiliation(s)
- Vidhu Singh
- Clinical Microbiology & Antimicrobial Research Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Hemraj Nandanwar
- Clinical Microbiology & Antimicrobial Research Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| |
Collapse
|
42
|
Matthyssen T, Li W, Holden JA, Lenzo JC, Hadjigol S, O’Brien-Simpson NM. Dimerization and lysine substitution of melittin have differing effects on bacteria. Front Pharmacol 2024; 15:1443497. [PMID: 39434904 PMCID: PMC11492869 DOI: 10.3389/fphar.2024.1443497] [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: 06/04/2024] [Accepted: 09/09/2024] [Indexed: 10/23/2024] Open
Abstract
Introduction Melittin is a potent antimicrobial peptide from bee venom that is effective against both Gram-positive and Gram-negative bacteria. However, it is extremely toxic to mammalian cells and, as yet, has no clinical use. Modifications to its amino acid sequence, cyclization, truncation, and dimerization have been attempted in order to reduce its toxicity whilst maintaining its antimicrobial activity. Methods In this study, we targeted the three lysine residues present in melittin and substituted them with lysine homologs containing shorter side chains (ornithine, Orn, diaminobutyric acid, Dab, and diaminopropanoic acid, Dap) and made both parallel and antiparallel melittin dimers to observe how lysine substitution and dimerization affects its activity and toxicity. The antibacterial activity of melittin and its analogs was tested against S. aureus (Gram-positive bacteria) and E. coli (Gram-negative bacteria), and cytotoxicity was tested against the mammalian cell lines HEK293 and H4IIE. Results Overall, dimerization and lysine substitution exhibited improved antimicrobial activity toward E. coli and limited improvement toward S. aureus. However, mammalian cell toxicity was only marginally reduced compared to native melittin. Interestingly, the parallel dimer was found to be marginally more active than the antiparallel dimer, indicating orientation maybe important for activity, although both dimers were less effective than the native and Lys-analog peptides toward S. aureus. Of the Lys substitutions, Dab and Dap improved melittin's activity toward E. coli. Discussion Dimerization and Lys substitution of melittin improved the antimicrobial activity toward Gram-negative bacteria but did not significantly improve its activity toward Gram-positive bacteria. Some analogs also displayed reduced toxicity toward HEK293 and H4IIE cells but overall remained toxic at bactericidal concentrations. Our data indicates that although highly antibacterial, melittin's toxicity is the major drawback in its potential use.
Collapse
Affiliation(s)
- Tamara Matthyssen
- ACTV Research Group, Melbourne Dental School, Division of Basic and Clinical Oral Sciences, Royal Dental Hospital and The Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne, Melbourne, VIC, Australia
| | - Wenyi Li
- ACTV Research Group, Melbourne Dental School, Division of Basic and Clinical Oral Sciences, Royal Dental Hospital and The Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - James A. Holden
- Melbourne Dental School, Centre for Oral Health Research, Royal Dental Hospital and The Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne, Melbourne, VIC, Australia
| | - Jason C. Lenzo
- Western Australian Health Translation Network, Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
| | - Sara Hadjigol
- ACTV Research Group, Melbourne Dental School, Division of Basic and Clinical Oral Sciences, Royal Dental Hospital and The Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne, Melbourne, VIC, Australia
| | - Neil M. O’Brien-Simpson
- ACTV Research Group, Melbourne Dental School, Division of Basic and Clinical Oral Sciences, Royal Dental Hospital and The Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
43
|
Matin M, Koszarska M, Atanasov AG, Król-Szmajda K, Jóźwik A, Stelmasiak A, Hejna M. Bioactive Potential of Algae and Algae-Derived Compounds: Focus on Anti-Inflammatory, Antimicrobial, and Antioxidant Effects. Molecules 2024; 29:4695. [PMID: 39407623 PMCID: PMC11477577 DOI: 10.3390/molecules29194695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
Algae, both micro- and macroalgae, are recognized for their rich repository of bioactive compounds with potential therapeutic applications. These marine organisms produce a variety of secondary metabolites that exhibit significant anti-inflammatory, antioxidant, and antimicrobial properties, offering promising avenues for the development of new drugs and nutraceuticals. Algae-derived compounds, including polyphenols, carotenoids, lipids, and polysaccharides, have demonstrated efficacy in modulating key inflammatory pathways, reducing oxidative stress, and inhibiting microbial growth. At the molecular level, these compounds influence macrophage activity, suppress the production of pro-inflammatory cytokines, and regulate apoptotic processes. Studies have shown that algae extracts can inhibit inflammatory signaling pathways such as NF-κB and MAPK, reduce oxidative damage by activating Nrf2, and offer an alternative to traditional antibiotics by combatting bacterial infections. Furthermore, algae's therapeutic potential extends to addressing diseases such as cardiovascular disorders, neurodegenerative conditions, and cancer, with ongoing research exploring their efficacy in preclinical animal models. The pig model, due to its physiological similarities to humans, is highlighted as particularly suitable for validating the bioactivities of algal compounds in vivo. This review underscores the need for further investigation into the specific mechanisms of action and clinical applications of algae-derived biomolecules.
Collapse
Affiliation(s)
- Maima Matin
- Department of Biotechnology and Nutrigenomics, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, 05-552 Jastrzębiec, Poland; (M.M.); (M.K.); (A.G.A.); (K.K.-S.); (A.J.)
| | - Magdalena Koszarska
- Department of Biotechnology and Nutrigenomics, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, 05-552 Jastrzębiec, Poland; (M.M.); (M.K.); (A.G.A.); (K.K.-S.); (A.J.)
| | - Atanas G. Atanasov
- Department of Biotechnology and Nutrigenomics, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, 05-552 Jastrzębiec, Poland; (M.M.); (M.K.); (A.G.A.); (K.K.-S.); (A.J.)
- Ludwig Boltzmann Institute Digital Health and Patient Safety, Medical University of Vienna, 1090 Vienna, Austria
| | - Karolina Król-Szmajda
- Department of Biotechnology and Nutrigenomics, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, 05-552 Jastrzębiec, Poland; (M.M.); (M.K.); (A.G.A.); (K.K.-S.); (A.J.)
| | - Artur Jóźwik
- Department of Biotechnology and Nutrigenomics, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, 05-552 Jastrzębiec, Poland; (M.M.); (M.K.); (A.G.A.); (K.K.-S.); (A.J.)
| | - Adrian Stelmasiak
- Department of Technique and Food Development, Institute of Human Nutrition Sciences, University of Life Sciences of Warsaw, 02-787 Warsaw, Poland;
| | - Monika Hejna
- Department of Biotechnology and Nutrigenomics, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, 05-552 Jastrzębiec, Poland; (M.M.); (M.K.); (A.G.A.); (K.K.-S.); (A.J.)
| |
Collapse
|
44
|
Miller WR, Arias CA. ESKAPE pathogens: antimicrobial resistance, epidemiology, clinical impact and therapeutics. Nat Rev Microbiol 2024; 22:598-616. [PMID: 38831030 DOI: 10.1038/s41579-024-01054-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2024] [Indexed: 06/05/2024]
Abstract
The rise of antibiotic resistance and a dwindling antimicrobial pipeline have been recognized as emerging threats to public health. The ESKAPE pathogens - Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter spp. - were initially identified as critical multidrug-resistant bacteria for which effective therapies were rapidly needed. Now, entering the third decade of the twenty-first century, and despite the introduction of several new antibiotics and antibiotic adjuvants, such as novel β-lactamase inhibitors, these organisms continue to represent major therapeutic challenges. These bacteria share several key biological features, including adaptations for survival in the modern health-care setting, diverse methods for acquiring resistance determinants and the dissemination of successful high-risk clones around the world. With the advent of next-generation sequencing, novel tools to track and combat the spread of these organisms have rapidly evolved, as well as renewed interest in non-traditional antibiotic approaches. In this Review, we explore the current epidemiology and clinical impact of this important group of bacterial pathogens and discuss relevant mechanisms of resistance to recently introduced antibiotics that affect their use in clinical settings. Furthermore, we discuss emerging therapeutic strategies needed for effective patient care in the era of widespread antimicrobial resistance.
Collapse
Affiliation(s)
- William R Miller
- Department of Internal Medicine, Division of Infectious Diseases, Houston Methodist Hospital, Houston, TX, USA
- Center for Infectious Diseases, Houston Methodist Research Institute, Houston, TX, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Cesar A Arias
- Department of Internal Medicine, Division of Infectious Diseases, Houston Methodist Hospital, Houston, TX, USA.
- Center for Infectious Diseases, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
45
|
Chen C, Yin Y, Lu P, Han T, Wang H, Li C. Establishing the Comprehensive Structure-Activity Relationship of the Natural Antibiotic Kibdelomycin/Amycolamicin. Angew Chem Int Ed Engl 2024:e202415439. [PMID: 39344479 DOI: 10.1002/anie.202415439] [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: 08/13/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/01/2024]
Abstract
Kibdelomycin (KBD) and amycolamicin (AMM) are potent natural antibiotics effective against antibiotic-resistant Gram-positive pathogens, including vancomycin-intermediate Staphylococcus aureus (S. aureus, VISA), methicillin-resistant S. aureus (MRSA), and quinolone-resistant S. aureus (QRSA). Their antibacterial activity stems from an unprecedented dual mechanism: the lower binding sites occupy the adenosine triphosphate (ATP) binding pocket of bacterial type II topoisomerases, while the upper binding sites disrupt the enzyme dimer interface. This dual action, combined with their unique chemical structures, positions KBD and AMM as promising scaffolds for developing new antibiotics. However, the structure-activity relationship (SAR) of KBD/AMM remains underexplored due to their highly complex chemical structures. In this study, we utilized total synthesis to produce KBD/AMM analogs with various site modifications and evaluated their antimicrobial activities. Our findings establish the first comprehensive SAR for KBD/AMM, paving the way for the development of novel KBD/AMM-based antibiotics.
Collapse
Affiliation(s)
- Chenglong Chen
- College of Life Sciences, Beijing Normal University, No. 19, Xinjiekouwai St, Haidian District, Beijing, 100875, P.R. China
- National Institute of Biological Sciences, Beijing, 7 Science Park Road ZGC Life Science Park, Beijing, 102206, P.R. China
| | - Yuyao Yin
- Department of Clinical Laboratory, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing, P.R. China
| | - Panrui Lu
- National Institute of Biological Sciences, Beijing, 7 Science Park Road ZGC Life Science Park, Beijing, 102206, P.R. China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 7 Science Park Road ZGC Life Science Park, Beijing, 102206, P.R. China
| | - Ting Han
- National Institute of Biological Sciences, Beijing, 7 Science Park Road ZGC Life Science Park, Beijing, 102206, P.R. China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 7 Science Park Road ZGC Life Science Park, Beijing, 102206, P.R. China
| | - Hui Wang
- Department of Clinical Laboratory, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing, P.R. China
| | - Chao Li
- National Institute of Biological Sciences, Beijing, 7 Science Park Road ZGC Life Science Park, Beijing, 102206, P.R. China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 7 Science Park Road ZGC Life Science Park, Beijing, 102206, P.R. China
| |
Collapse
|
46
|
Stephen C, Palmer D, Mishanina TV. Opportunities for Riboswitch Inhibition by Targeting Co-Transcriptional RNA Folding Events. Int J Mol Sci 2024; 25:10495. [PMID: 39408823 PMCID: PMC11476745 DOI: 10.3390/ijms251910495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/27/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Antibiotic resistance is a critical global health concern, causing millions of prolonged bacterial infections every year and straining our healthcare systems. Novel antibiotic strategies are essential to combating this health crisis and bacterial non-coding RNAs are promising targets for new antibiotics. In particular, a class of bacterial non-coding RNAs called riboswitches has attracted significant interest as antibiotic targets. Riboswitches reside in the 5'-untranslated region of an mRNA transcript and tune gene expression levels in cis by binding to a small-molecule ligand. Riboswitches often control expression of essential genes for bacterial survival, making riboswitch inhibitors an exciting prospect for new antibacterials. Synthetic ligand mimics have predominated the search for new riboswitch inhibitors, which are designed based on static structures of a riboswitch's ligand-sensing aptamer domain or identified by screening a small-molecule library. However, many small-molecule inhibitors that bind an isolated riboswitch aptamer domain with high affinity in vitro lack potency in vivo. Importantly, riboswitches fold and respond to the ligand during active transcription in vivo. This co-transcriptional folding is often not considered during inhibitor design, and may explain the discrepancy between a low Kd in vitro and poor inhibition in vivo. In this review, we cover advances in riboswitch co-transcriptional folding and illustrate how intermediate structures can be targeted by antisense oligonucleotides-an exciting new strategy for riboswitch inhibitor design.
Collapse
Affiliation(s)
| | | | - Tatiana V. Mishanina
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA (D.P.)
| |
Collapse
|
47
|
Yang R, Cui L, Xu S, Zhong Y, Xu T, Liu J, Lan Z, Qin S, Guo Y. Membrane-Targeting Amphiphilic Honokiol Derivatives Containing an Oxazole Moiety as Potential Antibacterials against Methicillin-Resistant Staphylococcus aureus. J Med Chem 2024; 67:16858-16872. [PMID: 39259708 DOI: 10.1021/acs.jmedchem.4c01860] [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: 09/13/2024]
Abstract
Infections with methicillin-resistant Staphylococcus aureus (MRSA) are becoming increasingly serious, making the development of novel antimicrobials urgent. Here, we synthesized some amphiphilic honokiol derivatives bearing an oxazole moiety and investigated their antibacterial and hemolytic activities. Bioactivity evaluation showed that E17 possessed significant in vitro antibacterial activity against S. aureus and MRSA, along with low hemolytic activity. Moreover, E17 exhibited rapid bactericidal properties and was not susceptible to resistance. Mechanistic studies indicated that E17 interacts with phosphatidylglycerol and cardiolipin of bacterial cell membranes, leading to changes in cell membrane permeability and polarization, increased intracellular ROS, and leakage of DNA and proteins, thus accelerating bacterial death. Transcriptome analysis further demonstrated that E17 has membrane-targeting effects, affecting the expression of genes related to cell membranes and ABC transporter proteins. Notably, in vivo activity showed that E17 has prominent anti-MRSA efficacy, comparable to vancomycin, and is expected to be a new anti-MRSA drug candidate.
Collapse
Affiliation(s)
- Ruige Yang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan Province 421001, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Liping Cui
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Shengnan Xu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan Province 421001, China
| | - Yan Zhong
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan Province 421001, China
| | - Ting Xu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan Province 421001, China
| | - Jifeng Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Zhenwei Lan
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Shangshang Qin
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Yong Guo
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan Province 421001, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| |
Collapse
|
48
|
Adhikari S, Lee HH, Kim DH. 'Primary' antibiotics in wastewater treatment plants. iScience 2024; 27:110789. [PMID: 39286505 PMCID: PMC11403462 DOI: 10.1016/j.isci.2024.110789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
There are anywhere from 5 to 8 priority antibiotics in typical wastewater treatment plants (WWTPs) whose concentrations exceed the maximum allowed, out of 12 priority antibiotics designated by the World Health Organization as the species to pose severe health hazard than others. If the priority antibiotics to deal with could be reduced to just one or two, such reduction would greatly simplify the construction and operation of the treatment plants. Introduced here is a concept of 'primary' antibiotic, the abatement of which ensures mitigation of all the other priority antibiotics in the wastewater. A criterion for determining primary antibiotic is developed. For a demonstration of the approach, the wastewater systems treated with solar-based photocatalysts are considered. The criterion reveals that the primary antibiotic in the typical European WWTP as well as in the typical municipal and hospital wastewater is ciprofloxacin, whereas the typical industrial wastewater contains ciprofloxacin and oxytetracycline.
Collapse
Affiliation(s)
- Sangeeta Adhikari
- School of Chemical Engineering, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea
| | - Hong H Lee
- School of Chemical and Biological Engineering, Seoul National University, Gwanak-ro, Seoul 08826, Republic of Korea
| | - Do-Heyoung Kim
- School of Chemical Engineering, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea
| |
Collapse
|
49
|
Zhang X, Li Y, Zhao Z, Ding J, Shan H, Ren R, Du C. An Intelligent Hydrogel Platform with Triple-Triggered On-Demand Release for Accelerating Diabetic Wound Healing. SMALL METHODS 2024:e2401127. [PMID: 39300860 DOI: 10.1002/smtd.202401127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/27/2024] [Indexed: 09/22/2024]
Abstract
The hydrogel platform with intelligent drug delivery system possesses great potential in the treatment of diabetic wounds. Nevertheless, the intelligent elimination of reactive oxygen species (ROS) remains a formidable challenge in facilitating diabetic wound healing. Herein, a hydrogel platform with triple-triggered on-demand release is constructed to intelligently scavenge ROS and modulate the wound microenvironment to accelerate diabetic wound healing through the release of antioxidative factors. Specifically, the gelatin (Gel) is modified with phenylboronic acid (PBA) to obtain a glucose-sensitive Gel derivative (Gel-BA), which is mixed with oxidized dextran (ODex) and the strong antioxidant myricetin (MY) to swiftly generate a hydrogel platform (OGM). Significantly, the smart release of MY from the hybrid hydrogel under inflammatory conditions intelligently eliminates ROS, effectively alleviating oxidative stress and promoting angiogenic reprogramming of the wound immune microenvironments by activating the Nrf2 pathway. In summary, in vitro and in vivo studies reveal that the OGM hydrogel platform significantly promotes cell proliferation, migration, and tube formation and greatly accelerates diabetic wound healing, offering a local-specific triple-response drug release strategy for the treatment of diabetic wound management.
Collapse
Affiliation(s)
- Xueliang Zhang
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, 200433, P. R. China
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Yingying Li
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Zhenfang Zhao
- High & New Technology Research Center, Henan Academy of Sciences, Zhengzhou, 450002, P. R. China
| | - Jin Ding
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, 200433, P. R. China
| | - Haojie Shan
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Ruizhen Ren
- Department of Endocrinology, Yantai Yuhuangding Hospital, Yantai, 264000, P. R. China
| | - Chang Du
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, 200433, P. R. China
| |
Collapse
|
50
|
Liu DY, Ruan YJ, Wang XL, Hu XY, Wang PF, Wen MM, Zhang CZ, Xiao YH, Liu XG. Concise synthesis of 3- C-glycosyl isocoumarins and 2-glycosyl-4 H-chromen-4-ones. Chem Commun (Camb) 2024; 60:10390-10393. [PMID: 39224044 DOI: 10.1039/d4cc03004d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
A new Ru-catalyzed C-H activation/cyclization reaction for the synthesis of 3-C-glycosyl isocoumarins and 2-glycosyl-4H-chromen-4-ones with carbonyl sulfoxonium ylide glycogen are reported. In this catalytic system, benzoic acid and its derivatives react with carbonyl sulfoxonium ylide glycogen to yield isocoumarin C-glycosides, while 2-hydroxybenzaldehyde substrates react to produce chromone C-glycosides. These reactions were characterized by mild reaction conditions, broad substrate scope, high functional-group compatibility, and high stereoselectivity to yield several high-value isocoumarins and chromone skeleton-containing C-glycosides. The methods were successfully implemented in the context of large-scale reactions and the late-stage modification of complex natural products.
Collapse
Affiliation(s)
- Deng-Yin Liu
- The Zhongzhou Laboratory for Integrative Biology, School of Pharmacy, Henan University, Kaifeng, Henan 475004, China.
| | - Yu-Jun Ruan
- The Zhongzhou Laboratory for Integrative Biology, School of Pharmacy, Henan University, Kaifeng, Henan 475004, China.
| | - Xiao-Li Wang
- The Zhongzhou Laboratory for Integrative Biology, School of Pharmacy, Henan University, Kaifeng, Henan 475004, China.
| | - Xin-Yue Hu
- The Zhongzhou Laboratory for Integrative Biology, School of Pharmacy, Henan University, Kaifeng, Henan 475004, China.
| | - Peng-Fei Wang
- The Zhongzhou Laboratory for Integrative Biology, School of Pharmacy, Henan University, Kaifeng, Henan 475004, China.
| | - Miao-Miao Wen
- The Zhongzhou Laboratory for Integrative Biology, School of Pharmacy, Henan University, Kaifeng, Henan 475004, China.
| | - Cong-Zhen Zhang
- The Zhongzhou Laboratory for Integrative Biology, School of Pharmacy, Henan University, Kaifeng, Henan 475004, China.
| | - Yu-He Xiao
- The Zhongzhou Laboratory for Integrative Biology, School of Pharmacy, Henan University, Kaifeng, Henan 475004, China.
| | - Xu-Ge Liu
- The Zhongzhou Laboratory for Integrative Biology, School of Pharmacy, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|