1
|
Cheng Q, Cheung Y, Xu C, Chan EWC, Chan KF, Chen S. Overall mutational scanning unveils the essential active residues for the mechanistic action of MCR-1. Microbiol Res 2025; 291:127982. [PMID: 39608179 DOI: 10.1016/j.micres.2024.127982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/11/2024] [Accepted: 11/20/2024] [Indexed: 11/30/2024]
Abstract
Polymyxins, including colistin and polymyxin B, serve as crucial last-resort antibiotics for managing infections caused by carbapenem-resistant Enterobacterales (CRE). However, the rapid spread of the mobilized colistin resistance gene (mcr-1) challenged the efficacy of treatment by polymyxins. The mcr-1 gene encoded a transmembrane phosphoethanolamine (PEA) transferase enzyme, MCR-1. MCR-1 could catalyze the transfer of PEA moiety of phosphatidylethanolamine (PE) to the 1' (or 4')-phosphate group of the lipid A. Despite the determination of several structures of the soluble domain of MCR-1, the structural and biochemical mechanisms of integral MCR-1 remain less understood. In this study, we utilized an alanine scanning mutagenesis approach to systematically investigate the functional attributes of distinct regions within MCR-1. We identified fifteen critical residues that are indispensable for the enzymatic activity of MCR-1 and are pivotal for its ability to confer resistance to colistin. Furthermore, molecular docking of MCR-1 complexed with the phosphoethanolamine (PE) substrate revealed the presence of a channel-shaped cavity, a characteristic feature shared with other phosphoethanolamine transferases. Despite MCR-1 exhibiting a low sequence identity with both MCR homologues and other phosphoethanolamine (PEA) transferases, several conserved sites were identified, including Y97, M105, K333, H395, L477, and H478, suggesting a potentially shared catalytic mechanism among them for modifying LPS-lipid A. Overall, these findings provide a deep understanding of the catalytic mechanism of MCR-1 for colistin resistance. Moreover, these findings provide a robust structural and functional foundation, enabling the rational design of targeted inhibitors and restoring colistin activity against serious infections with carbapenem-resistant Enterobacterales (CRE).
Collapse
Affiliation(s)
- Qipeng Cheng
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Metabolic Diseases, College of Life Sciences, Anhui Normal University, Wuhu, Anhui, China; State Key Laboratory of Chemical Biology and Drug Discovery, Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Yanchu Cheung
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Chen Xu
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Edward Wai Chi Chan
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Kin Fai Chan
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Sheng Chen
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong.
| |
Collapse
|
2
|
Cui XD, Liu SB, Wang RY, He DD, Pan YS, Yuan L, Zhai YJ, Hu GZ. Investigation on the reversal effect of closantel on colistin resistance in MCR-1 positive Escherichia coli based on dose-response relationship. J Antimicrob Chemother 2024:dkae441. [PMID: 39658100 DOI: 10.1093/jac/dkae441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/26/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND The lack of research on the dose-response relationship of adjuvants in reversing colistin resistance will lead to a lack of scientific theoretical basis for determining the dosage of adjuvants in clinical combination therapy plans or their compound formulations. OBJECTIVES This study investigates the dose-response relationship of the deworming drug closantel (CST) on the reversal of colistin resistance in mcr-1-positive Escherichia coli (E. coli). METHODS Firstly, the reversal effect of different concentrations of CST on colistin resistance in mcr-1-positive E. coli was analysed using broth microdilution method, checkerboard method and time-killing curves. Then, the inhibitory effect of CST on the development of colistin resistance, as well as the haemolytic and cytotoxic properties of CST, was analysed. Finally, the in vivo efficacy of the combination of CST and colistin was evaluated. RESULTS Both the checkerboard assays and the time-killing curves indicate that there is a special dose-response relationship between CST and its reversal effect on colistin resistance, which is not concentration-dependent. High reversal efficiency can be achieved within a low concentration range. However, as the CST concentration increases, the ability to reverse colistin resistance remains unchanged or decreases, which resulted in a gradual decrease in reversal efficiency. Additionally, CST can inhibit the development of colistin resistance and reduce the cytotoxicity of colistin. Importantly, in a mouse model of E. coli infection, the combination of CST and colistin showed a significant therapeutic effect. CONCLUSIONS This study indicates a special dose-response relationship between CST and its reversal effect on colistin resistance, which was not concentration-dependent.
Collapse
Affiliation(s)
- Xiao-Die Cui
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Shuo-Bo Liu
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Rui-Yun Wang
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Dan-Dan He
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Yu-Shan Pan
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Li Yuan
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Ya-Jun Zhai
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Gong-Zheng Hu
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, China
| |
Collapse
|
3
|
Mao YN, Ma YJ, Wang GQ. Synergistic Antibacterial Effect of Lactic Acid Bacteria and Baicalin Against Staphylococcus aureus In Vitro and In Vivo. Foodborne Pathog Dis 2024. [PMID: 39527139 DOI: 10.1089/fpd.2024.0086] [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: 11/16/2024] Open
Abstract
Pathogenic bacteria such as Staphylococcus aureus (S. aureus) are the principal cause of cow mastitis, which primarily impacts milk yield and results in significant financial losses for the animal husbandry industry. Lactic acid bacteria-cell-free supernatant (LAB-CFS) and baicalin (BAI) both have a number of biological effects, including decreasing inflammation. The combined use of LAB-CFS and BAI does not appear to have been used to protective against mastitis, however, and the underlying mechanisms are yet unknown. In this study, in vitro activity of LAB-CFS and BAI alone and in combination was determined (checkerboard experiments, time-kill curves, and flow cytometry to investigate membrane permeability) and examined the protective effects of LAB-CFS and BAI on S. aureus-induced mastitis in mice and the impact of NF-κB signaling pathways on the emergence of mastitis. We discovered that when LAB-CFS and BAI were used together, S. aureus was more effectively treated than when LAB-CFS and BAI were used separately. Flow cytometry demonstrated that LAB-CFS and BAI work together to kill bacteria. In vivo, the usage of LAB-CFS and BAI decreased the activity of myeloperoxidase, as well as IL-6, IL-1β, and TNF-α secretion and the levels of TLR2 and p65 (NF-κB) expression. These findings suggested that LAB-CFS and BAI had a preventive effect against mastitis brought on by S. aureus. Therefore, the NF-κB signaling pathway is thought to be the likely mechanism through which LAB-CFS and BAI reduced S. aureus-induced inflammation in the mammary of cows. For the treatment of cow mastitis, LAB-CFS and BAI are likely to replace antibiotics.
Collapse
Affiliation(s)
- Yan-Ni Mao
- Veterinary Pharmacology Lab, School of Animal Science and Technology, Ningxia University, Yinchuan, China
- Guyuan Branch, Ning Xia Academy of Agriculture and Forestry Sciences, Guyuan, China
| | - Yan-Jun Ma
- Veterinary Pharmacology Lab, School of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Gui-Qin Wang
- Veterinary Pharmacology Lab, School of Animal Science and Technology, Ningxia University, Yinchuan, China
| |
Collapse
|
4
|
Cheng P, Sun Y, Wang B, Liang S, Yang Y, Gui S, Zhang K, Qu S, Li L. Mechanism of synergistic action of colistin with resveratrol and baicalin against mcr-1-positive Escherichia coli. Biomed Pharmacother 2024; 180:117487. [PMID: 39332187 DOI: 10.1016/j.biopha.2024.117487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/11/2024] [Accepted: 09/20/2024] [Indexed: 09/29/2024] Open
Abstract
The rising incidence of colistin (COL) resistance poses a significant challenge, undermining the therapeutic efficacy of COL against life-threatening bacterial infections. Therefore, the urgent identification and development of new therapeutics are imperative. It has been proven that combinations of antibiotics and promising non-antibiotic agents could be a potential strategy to combat infections caused by MDR pathogens. Due to various antimicrobial properties, medicinal plants have attracted significant attention, which could be promising adjuvant. In this study, we investigated the synergistic effects of combining COL with resveratrol (RST) and baicalin (BAI) against mcr-1-positive Escherichia coli through antibiotic susceptibility testing, checkerboard method and time-killing assays. The mechanisms of combination treatment were analyzed using SEM, fluorometric assays and transcriptome analysis. The molecular docking assay was conducted to elucidate potential interactions between RST, BAI and the MCR-1 protein. Finally, we assessed the in vivo efficacy of combination against mcr-1-positive Escherichia coli. The results demonstrated that the combination of RST, BAI and COL showed significant synergistic activity both in vitro and in vivo. Further mechanistic study revealed that the combination could increase the membrane-damaging ability of COL, disrupt the homeostasis of proton motive force (PMF), inhibit the activity of efflux pumps and impair ATP supply. The molecular docking revealed that RST and BAI could bind to MCR-1 stably, indicating the combination of RST and BAI may be an effective MCR-1 inhibitor. Our findings demonstrated that the combination of RST and BAI might be potential COL adjuvant, providing an alternative approach to address mcr-1-positive Escherichia coli infections.
Collapse
Affiliation(s)
- Ping Cheng
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Yingying Sun
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Botao Wang
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Shuying Liang
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Yuqi Yang
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Shixin Gui
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Kai Zhang
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Shaoqi Qu
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China.
| | - Lin Li
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China.
| |
Collapse
|
5
|
Nuske MR, Zhong J, Huang R, Sarojini V, Chen JLY, Squire CJ, Blaskovich MAT, Leung IKH. Adjuvant strategies to tackle mcr-mediated polymyxin resistance. RSC Med Chem 2024:d4md00654b. [PMID: 39539347 PMCID: PMC11556429 DOI: 10.1039/d4md00654b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
The emergence of the mobile colistin resistance (mcr) gene is a demonstrable threat contributing to the worldwide antibiotic resistance crisis. The gene is encoded on plasmids and can easily spread between different bacterial strains. mcr encodes a phosphoethanolamine (pEtN) transferase, which catalyses the transfer of the pEtN moiety from phosphatidylethanolamine to lipid A, the head group of lipopolysaccharides (LPS). This neutralises the overall negative charge of the LPS and prevents the binding of polymyxins to bacterial membranes. We believe that the development of polymyxin adjuvants could be a promising approach to prolong the use of this important class of last-resort antibiotics. This review discusses recent progress in the identification, design and development of adjuvants to restore polymyxin sensitivity in these resistant bacteria, and focuses on both MCR inhibitors as well as alternative approaches that modulate polymyxin resistance.
Collapse
Affiliation(s)
- Madison R Nuske
- School of Chemistry, The University of Melbourne Parkville Victoria 3010 Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne Parkville Victoria 3010 Australia
| | - Junlang Zhong
- School of Chemistry, The University of Melbourne Parkville Victoria 3010 Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne Parkville Victoria 3010 Australia
| | - Renjie Huang
- School of Chemical Sciences, The University of Auckland Auckland 1010 New Zealand
| | | | - Jack L Y Chen
- Centre for Biomedical and Chemical Sciences, School of Science, Auckland University of Technology Auckland 1010 New Zealand
- Department of Biotechnology, Chemistry and Pharmaceutical Sciences, Università degli Studi di Siena 53100 Siena Italy
| | - Christopher J Squire
- School of Biological Sciences, The University of Auckland Auckland 1010 New Zealand
| | - Mark A T Blaskovich
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland St. Lucia Queensland 4072 Australia
| | - Ivanhoe K H Leung
- School of Chemistry, The University of Melbourne Parkville Victoria 3010 Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne Parkville Victoria 3010 Australia
- School of Chemical Sciences, The University of Auckland Auckland 1010 New Zealand
| |
Collapse
|
6
|
Liu H, Zhang Y, Zhong Z, Gong Y, Yu P, Yang Y, Zhang Y, Zhou T, Chen L. Immunomodulator AS101 restores colistin susceptibility of clinical colistin-resistant Escherichia coli and Klebsiella pneumoniae in vitro and in vivo. Int J Antimicrob Agents 2024; 64:107285. [PMID: 39111708 DOI: 10.1016/j.ijantimicag.2024.107285] [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: 05/06/2024] [Revised: 07/08/2024] [Accepted: 07/22/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVES Colistin (COL) was once considered to be the last line of defence against multidrug-resistant bacteria belonging to the family Enterobacteriaceae. Due to the misuse of COL, COL-resistant (COL-R) Enterobacteriaceae have emerged. To address this clinical issue and combat COL resistance, novel approaches are urgently needed. METHODS In this study, the in vitro and in vivo antimicrobial and antibiofilm effects of the immunomodulator AS101 were investigated in combination with COL against COL-R Escherichia coli (E. coli) and Klebsiella pneumoniae (K. pneumoniae). RESULTS Checkerboard assay, time-kill assay, and scanning electron microscopy confirmed the in vitro antimicrobial phenotype, whereas, crystal violet staining and multidimensional confocal laser scanning microscopy with live/dead staining confirmed the antibiofilm capability of the combination therapy. Moreover, the Galleria mellonella infection model and the mouse infection model indicated the high in vivo efficacy of the combination therapy. Additionally, cytotoxicity experiments performed using human kidney-derived HK-2 cells and haemolysis assays performed using human erythrocytes collectively demonstrated safety at effective combination concentrations. Furthermore, quantification of the expression of inflammatory cytokines via enzyme-linked immunosorbent assay confirmed the anti-inflammatory advantage of combination therapy. At the mechanistic level, changes in outer and inner membrane permeability and accumulation of ROS levels, which might be potential mechanisms for synergistic antimicrobial effects. CONCLUSIONS This study found that AS101 can restore COL susceptibility in clinical COL-R E. coli and K. pneumoniae and also has synergistic antibiofilm and anti-inflammatory capabilities. This study provided a novel strategy to combat clinical infections caused by COL-R E. coli and K. pneumoniae.
Collapse
Affiliation(s)
- Haifeng Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Ying Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Zeyong Zhong
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yanchun Gong
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Pingting Yu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Yuhan Yang
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yichi Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Tieli Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Lijiang Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China.
| |
Collapse
|
7
|
Li J, Zhang QY, Lu QY, Liu QZ, Guo L, Li M, Sun QY. Baicalin relieves complement alternative pathway activation-induced lung inflammation through inhibition of NF-κB pathway. BMC Complement Med Ther 2024; 24:334. [PMID: 39272057 PMCID: PMC11395835 DOI: 10.1186/s12906-024-04622-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 08/19/2024] [Indexed: 09/15/2024] Open
Abstract
INTRODUCTION Acute lung injury (ALI) as one kind of acute pulmonary inflammatory disorder, manifests primarily as damage to alveolar epithelial cells and microvascular endothelial cells. Activation of the complement system is a common pathological mechanism in ALI induced by diverse factors, with the complement alternative pathway assuming a pivotal role. Baicalin, a flavonoid derived from the root of Scutellaria baicalensis Georgi, exhibits noteworthy biological activities. The present study attempted the interventional effects and underlying mechanisms of baicalin in microangiopathy in ALI induced by complement alternative pathway activation. METHODS Activation of the complement alternative pathway by cobra venom factor (CVF). HMEC cells were pretreated with baicalin and then exposed to complement activation products. The expression of inflammatory mediators was detected by ELISA, and the intranuclear transcriptional activity of NF-κB was assessed by a dual fluorescent kinase reporter gene assay kit. Before establishing the ALI mouse model, baicalin or PDTC was gavaged for 7 d. CVF was injected into the tail vein to establish the ALI model. The levels of inflammatory mediators in BALF and serum were determined by ELISA. HE staining and immunohistochemistry evaluated pathological changes, complement activation product deposition, and NF-κB p65 phosphorylation in lung tissue. RESULTS Baicalin reduced complement alternative activation product-induced expression of HMEC cells adhesion molecules (ICAM-1, VCAM-1, E-selectin) and cytokines (IL-6, TNF-α) as well as upregulation of NF-κB intranuclear transcriptional activity. Baicalin intervention reduced the number of inflammatory cells and protein content in the BALF and decreased the levels of IL-6, TNF-α, and ICAM-1 in serum and IL-6, TNF-α, ICAM-1, and P-selectin in BLAF. In addition, baicalin attenuated inflammatory cell infiltration in the lung of ALI mice and reduced the deposition of complement activation products (C5a, C5b-9) and phosphorylation of NF-κB p65 in lung tissue. CONCLUSION Baicalin relieves complement alternative pathway activation-induced lung inflammation by inhibition of NF-κB pathway, delaying the progression of ALI.
Collapse
Affiliation(s)
- Jiao Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
| | - Qi-Yun Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
| | - Qing-Yu Lu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
| | - Qiao-Zhou Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
| | - Li Guo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
| | - Min Li
- Guizhou Provincial People's Hospital, No. 83, Zhongshan East Road, Nanming District, Guiyang, 550000, China.
| | - Qian-Yun Sun
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China.
- Natural Products Research Center of Guizhou Province, Guiyang, China.
| |
Collapse
|
8
|
Zerrouki H, Hamieh A, Hadjadj L, Rolain JM, Baron SA. The effect of combinations of a glyphosate-based herbicide with various clinically used antibiotics on phenotypic traits of Gram-negative species from the ESKAPEE group. Sci Rep 2024; 14:21006. [PMID: 39251613 PMCID: PMC11383965 DOI: 10.1038/s41598-024-68968-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 07/30/2024] [Indexed: 09/11/2024] Open
Abstract
The emission of glyphosate and antibiotic residues from human activities threatens the diversity and functioning of the microbial community. This study examines the impact of a glyphosate-based herbicide (GBH) and common antibiotics on Gram-negative bacteria within the ESKAPEE group (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter spp. and Escherichia coli). Ten strains, including type and multidrug-resistant strains for each species were analysed and eight antibiotics (cefotaxime, meropenem, aztreonam, ciprofloxacin, gentamicin, tigecycline, sulfamethoxazole-trimethoprim, and colistin) were combined with the GBH. While most combinations yielded additive or indifferent effects in 70 associations, antagonistic effects were observed with ciprofloxacin and gentamicin in five strains. GBH notably decreased the minimum inhibitory concentration of colistin in eight strains and displayed synergistic activity with meropenem against metallo-β-lactamase (MBL)-producing strains. Investigation into the effect of GBH properties on outer membrane permeability involved exposing strains to a combination of this GBH and vancomycin. Results indicated that GBH rendered strains sensitive to vancomycin, which is typically ineffective against Gram-negative bacteria. Furthermore, we examined the impact of GBH in combination with three carbapenem agents on 14 strains exhibiting varying carbapenem-resistance mechanisms to assess its effect on carbapenemase activity. The GBH efficiently inhibited MBL activity, demonstrating similar effects to EDTA (ethylenediaminetetraacetic acid). Chelating effect of GBH may have multifaceted impacts on bacterial cells, potentially by increasing outer membrane permeability and inactivating metalloenzyme activity.
Collapse
Affiliation(s)
- Hanane Zerrouki
- MEPHI, Faculté de Médecine et de Pharmacie, Aix Marseille University, 19-21 Boulevard Jean Moulin, 13385, Marseille Cedex 05, France
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13385, Marseille Cedex 05, France
| | - Aïcha Hamieh
- MEPHI, Faculté de Médecine et de Pharmacie, Aix Marseille University, 19-21 Boulevard Jean Moulin, 13385, Marseille Cedex 05, France
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13385, Marseille Cedex 05, France
| | - Linda Hadjadj
- MEPHI, Faculté de Médecine et de Pharmacie, Aix Marseille University, 19-21 Boulevard Jean Moulin, 13385, Marseille Cedex 05, France
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13385, Marseille Cedex 05, France
| | - Jean-Marc Rolain
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13385, Marseille Cedex 05, France.
- APHM, MEPHI, Faculté de Médecine et de Pharmacie, Aix Marseille University, 19-21 Boulevard Jean Moulin, 13385, Marseille Cedex 05, France.
| | - Sophie Alexandra Baron
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13385, Marseille Cedex 05, France.
- APHM, MEPHI, Faculté de Médecine et de Pharmacie, Aix Marseille University, 19-21 Boulevard Jean Moulin, 13385, Marseille Cedex 05, France.
| |
Collapse
|
9
|
Li Y, Niu H, Huang L, Zhang L, Mao L, Wan P, Ma Z, Peng X, Wan K, Zeng Z. Research Note: Synergistic effect of isopropoxy benzene guanidine and colistin against mcr-1-positive Escherichia coli in vitro and in duck intestine infection models. Poult Sci 2024; 103:104018. [PMID: 39043027 PMCID: PMC11318535 DOI: 10.1016/j.psj.2024.104018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/01/2024] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Colistin (CST) is considered as "agent of last resort" against gram-negative bacteria as feed additive. Its clinical effectiveness has reduced since the emergence of mcr-1 gene in ducks. Isopropoxy benzene guanidine (IBG), a new guanidine derivative, showed positive effects on improving animal weights and alleviating intestinal pathogens, therefore, the objective of this study was to evaluate the effect of this compound supplement with CST in ducks and explore the possibilities in feed additive. A total of fifteen duck-origin Escherichia coli carrying the mcr-1 gene were included in this study. A checkerboard microdilution assay was used to evaluate the in vitro antibacterial activity of IBG combined with CST against mcr-1-positive E. coli. A 3-by-2 time-kill array of IBG (16, 32, and 64 μg/mL) and CST (1/2 MIC and 1/4 MIC) over 24 hours was utilized to characterize the activity of the agents alone and in combination against E. coli strain 1 in vitro. The intestinal colonization model was used to evaluate the in vivo effect of IBG combined with CST. These results indicated that the combination of IBG plus CST showed a synergistic effect against all clinical isolates (FICI < 0.5). The bacterial burden was reduced by more than 2 log10 CFU/mL when E. coli strain 1 was tested with 1/2 MIC CST plus 64 μg/mL IBG for 24 h. Further experiments in vivo demonstrated that the CST combined with IBG was able to increase duck weights, reduced intestinal pathogenic E. coli and showed a synergistic antibacterial effect. Combination of CST (4 mg/kg b.w.) plus IBG (32 or 64 mg/kg b.w.) achieved 1.84 to 3.29 log10 CFU/g killing after 7 d of therapy, which was significantly different from that in the challenge control group (p<0.05). In summary, our study demonstrated the potential use of IBG as feed additive for veterinary purposes in ducks and provided new insights into overcoming resistance in the future.
Collapse
Affiliation(s)
- Yafei Li
- Institute of Quality Standard and Monitoring Technology for Agro-Products of Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; Guangdong Provincial Key Laboratory of Quality & Safety Risk Assessment for Agro-Products, Guangzhou 510640, China
| | - Huijun Niu
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Lang Huang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Lingxuan Zhang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Lingxiang Mao
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Peng Wan
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhenbao Ma
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Xianfeng Peng
- Guangzhou Insighter Biotechnology Co., Ltd., Guangzhou 510663, China
| | - Kai Wan
- Institute of Quality Standard and Monitoring Technology for Agro-Products of Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; Guangdong Provincial Key Laboratory of Quality & Safety Risk Assessment for Agro-Products, Guangzhou 510640, China
| | - Zhenling Zeng
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
10
|
Ashrafudoulla M, Yun H, Ashikur Rahman M, Jung SJ, Jie-Won Ha A, Anamul Hasan Chowdhury M, Shaila S, Akter S, Park SH, Ha SD. Prophylactic efficacy of baicalin and carvacrol against Salmonella Typhimurium biofilm on food and food contact surfaces. Food Res Int 2024; 187:114458. [PMID: 38763690 DOI: 10.1016/j.foodres.2024.114458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/28/2024] [Accepted: 05/01/2024] [Indexed: 05/21/2024]
Abstract
This study examines the antimicrobial and antibiofilm effectiveness of baicalin and carvacrol against Salmonella enterica ser. Typhimurium on food contact surfaces and chicken meat. The minimum inhibitory concentrations (MIC) for baicalin and carvacrol were found to be 100 μg/mL and 200 μg/mL, respectively, which aligns with findings from previous studies. The compounds exhibited a concentration-dependent decrease in microbial populations and biofilm formation. When used together, they displayed a remarkable synergistic effect, greatly augmenting their antibacterial activity. The assessment of food quality demonstrated that these treatments have no negative impact on the sensory characteristics of chicken meat. The impact of the structure on biofilms was observed through the use of Field Emission Scanning Electron Microscopy (FE-SEM) and Confocal Laser Scanning Microscopy (CLSM), revealing disrupted biofilm architectures and decreased cell viability. Crucially, RT-PCR analysis revealed a marked downregulation of quorum sensing (luxS), virulence (hilA), and stress response (rpoS) genes, highlighting the multifaceted antimicrobial mechanism of action. This gene-specific suppression suggests a targeted disruption of bacterial communication and virulence pathways, offering insight into the comprehensive antibiofilm strategy. This provides further insight into the molecular mechanisms that contribute to their antibiofilm effects.
Collapse
Affiliation(s)
- Md Ashrafudoulla
- School of Food Science and Technology, Chung-Ang University, 72-1 Nae-Ri, Anseong, Gyeonggi-do 456-756, Republic of Korea; National Institute of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Hyojae Yun
- School of Food Science and Technology, Chung-Ang University, 72-1 Nae-Ri, Anseong, Gyeonggi-do 456-756, Republic of Korea
| | - Md Ashikur Rahman
- School of Food Science and Technology, Chung-Ang University, 72-1 Nae-Ri, Anseong, Gyeonggi-do 456-756, Republic of Korea
| | - Soo-Jin Jung
- School of Food Science and Technology, Chung-Ang University, 72-1 Nae-Ri, Anseong, Gyeonggi-do 456-756, Republic of Korea
| | - Angela Jie-Won Ha
- School of Food Science and Technology, Chung-Ang University, 72-1 Nae-Ri, Anseong, Gyeonggi-do 456-756, Republic of Korea; Grand Hyatt Jeju, 12 Noyeon-ro, Jeju-si, Jeju-do, South Korea
| | - Md Anamul Hasan Chowdhury
- School of Food Science and Technology, Chung-Ang University, 72-1 Nae-Ri, Anseong, Gyeonggi-do 456-756, Republic of Korea
| | - Shanjida Shaila
- School of Food Science and Technology, Chung-Ang University, 72-1 Nae-Ri, Anseong, Gyeonggi-do 456-756, Republic of Korea
| | - Shirin Akter
- School of Food Science and Technology, Chung-Ang University, 72-1 Nae-Ri, Anseong, Gyeonggi-do 456-756, Republic of Korea
| | - Si Hong Park
- Food Science and Technology, Oregon State University, Corvallis, OR, USA
| | - Sang-Do Ha
- School of Food Science and Technology, Chung-Ang University, 72-1 Nae-Ri, Anseong, Gyeonggi-do 456-756, Republic of Korea.
| |
Collapse
|
11
|
Cui XD, Liu XK, Ma XY, Li SH, Zhang JK, Han RJ, Yi KF, Liu JH, Pan YS, He DD, Hu GZ, Zhai YJ. Restoring colistin sensitivity in colistin-resistant Salmonella and Escherichia coli: combinatorial use of berberine and EDTA with colistin. mSphere 2024; 9:e0018224. [PMID: 38738873 PMCID: PMC11332338 DOI: 10.1128/msphere.00182-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 04/07/2024] [Indexed: 05/14/2024] Open
Abstract
The appearance and prevalence of multidrug-resistance (MDR) Gram-negative bacteria (GNB) have limited our antibiotic capacity to control bacterial infections. The clinical efficacy of colistin (COL), considered as the "last resort" for treating GNB infections, has been severely hindered by its increased use as well as the emergence and prevalence of mobile colistin resistance (MCR)-mediated acquired drug resistance. Identifying promising compounds to restore antibiotic activity is becoming an effective strategy to alleviate the crisis of increasing MDR. We first demonstrated that the combination of berberine (BBR) and EDTA substantially restored COL sensitivity against COL-resistant Salmonella and Escherichia coli. Molecular docking indicated that BBR can interact with MCR-1 and the efflux pump system AcrAB-TolC, and BBR combined with EDTA downregulated the expression level of mcr-1 and tolC. Mechanically, BBR combined with EDTA could increase bacterial membrane damage, inhibit the function of multidrug efflux pump, and promote oxidative damage, thereby boosting the action of COL. In addition, transcriptome analysis found that the combination of BBR and EDTA can accelerate the tricarboxylic acid cycle, inhibit cationic antimicrobial peptide (CAMP) resistance, and attenuate Salmonella virulence. Notably, the combination of BBR and EDTA with COL significantly reduced the bacterial load in the liver and spleen of a mice model infected with Salmonella. Our findings revealed that BBR and EDTA can be used as adjuvants collectively with COL to synergistically reverse the COL resistance of bacteria. IMPORTANCE Colistin is last-resort antibiotic used to treat serious clinical infections caused by MDR bacterial pathogens. The recent emergence of transferable plasmid-mediated COL resistance gene mcr-1 has raised the specter of a rapid worldwide spread of COL resistance. Coupled with the fact of barren antibiotic development pipeline nowadays, a critical approach is to revitalize existing antibiotics using antibiotic adjuvants. Our research showed that berberine combined with EDTA effectively reversed COL resistance both in vivo and in vitro through multiple modes of action. The discovery of berberine in combination with EDTA as a new and safe COL adjuvant provides a therapeutic regimen for combating Gram-negative bacteria infections. Our findings provide a potential therapeutic option using existing antibiotics in combination with antibiotic adjuvants and address the prevalent infections caused by MDR Gram-negative pathogens worldwide.
Collapse
Affiliation(s)
- Xiao-die Cui
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Xiao-kang Liu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Xiao-yuan Ma
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Shuai-hua Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Jun-kai Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Rong-jia Han
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Kai-fang Yi
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Jian-hua Liu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Yu-shan Pan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Dan-dan He
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Gong-zheng Hu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Ya-jun Zhai
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| |
Collapse
|
12
|
Bing C, Mengjuan A, Xinyu M, Chixin Z, Xinyao T, Yan S, Zhi L. Efflux pump inhibitor chlorpromazine effectively increases the susceptibility of Escherichia coli to antimicrobial peptide Brevinin-2CE. Future Microbiol 2024; 19:771-782. [PMID: 38683168 PMCID: PMC11290751 DOI: 10.2217/fmb-2023-0272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/21/2024] [Indexed: 05/01/2024] Open
Abstract
Aim: The response of E. coli ATCC8739 to Brevinin-2CE (B2CE) was evaluated as a strategy to prevent the development of antimicrobial peptide (AMP)-resistant bacteria. Methods: Gene expression levels were detected by transcriptome sequencing and RT-PCR. Target genes were knocked out using CRISPR-Cas9. MIC was measured to evaluate strain resistance. Results: Expression of acrZ and sugE were increased with B2CE stimulation. ATCC8739ΔacrZ and ATCC8739ΔsugE showed twofold and fourfold increased sensitivity, respectively. The survival rate of ATCC8739 was reduced in the presence of B2CE/chlorpromazine (CPZ). Combinations of other AMPs with CPZ also showed antibacterial effects. Conclusion: The results indicate that combinations of AMPs/efflux pump inhibitors (EPIs) may be a potential approach to combat resistant bacteria.
Collapse
Affiliation(s)
- Cao Bing
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, PR China
| | - An Mengjuan
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, PR China
| | - Ma Xinyu
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, PR China
| | - Zhu Chixin
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, PR China
| | - Tan Xinyao
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, PR China
| | - Sun Yan
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, PR China
| | - Li Zhi
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, PR China
| |
Collapse
|
13
|
Zhang M, Li ZE, Duan MH, Dai Y, Jin YG, Liu Y, Zhang YN, Li XP, Yang F. Effects of chitooligosaccharide on the in vitro antibacterial activity against avian Escherichia coli and the pharmacokinetics of florfenicol in healthy chickens. Poult Sci 2024; 103:103373. [PMID: 38150832 PMCID: PMC10788265 DOI: 10.1016/j.psj.2023.103373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/29/2023] Open
Abstract
This study investigates the combined effects of chitooligosaccharide (COS) and florfenicol (FLO) on the inhibition of Escherichia coli in vitro, as well as the pharmacokinetic interactions between these compounds in healthy chickens. The minimum inhibitory concentration (MIC) of COS and FLO alone and the fractional inhibitory concentration index (FICI) after combined treatment were determined using the broth microdilution method and checkerboard method, respectively. Additionally, we evaluated the pharmacokinetic interactions between the 2 types of COS and FLO in healthy chickens. Thirty chickens were randomly divided into 3 groups: Florfenicol group (30 mg/kg), COS J85 group (COS J85 20 mg/kg + florfenicol 30 mg/kg), COS H85 group (COS H85 20 mg/kg + florfenicol 30 mg/kg). Either FLO or COS was orally administered by gavage. The concentrations of FLO in chicken plasma were measured at different time points after the drug withdrawal using high-performance liquid chromatography (HPLC), and pharmacokinetic parameters were calculated by a compartmental method. The results showed that COS J85 and COS H85, when combined with FLO, had FICI values of 0.1875 to 0.75 and 0.3125 to 1, respectively, indicating good synergistic or additive effects against Escherichia coli. The pharmacokinetics of FLO alone and in combination with COS followed a 1-compartment model with first-order absorption and elimination. Furthermore, the pharmacokinetic analysis revealed that the elimination half-life (t1/2ke) of florfenicol was significantly increased in the COS H85 group compared to oral administration of florfenicol alone (P < 0.05). Other pharmacokinetic parameters did not show significant changes (P > 0.05), except between the 2 combined treatment groups, where statistical differences were observed for various parameters, excluding the area under the concentration-time curve from the time of dosing to infinity (AUC) and peak concentration (Cmax).
Collapse
Affiliation(s)
- Mei Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Ze-En Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Ming-Hui Duan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Yan Dai
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Yang-Guang Jin
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Yue Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Yan-Ni Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Xing-Ping Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Fan Yang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China.
| |
Collapse
|
14
|
Feng H, Zhang J, Zhang K, Wang X, Guo Z, Wang L, Li J. Synergistic anti-infectious bronchitis virus activity of Phillygenin combined Baicalin by modulating respiratory microbiota and improving metabolic disorders. Poult Sci 2024; 103:103371. [PMID: 38150830 PMCID: PMC10788278 DOI: 10.1016/j.psj.2023.103371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/03/2023] [Accepted: 12/07/2023] [Indexed: 12/29/2023] Open
Abstract
Phillygenin (PHI) and Baicalin (Bai) are the major chemical ingredients extracted from Forsythia suspensa and Scutellaria baicalensis, respectively. The mixture of Forsythia suspensa and Scutellaria baicalensis according to the theories of Traditional Chinese Veterinary Medicine, compounded formulation can effectively exert heat-clearing and detoxifying effect, but the synergistic anti-IBV activity of PHI combined with Bai was unclear. Here, the protection of PHI combined with Bai on avian infectious bronchitis virus (IBV) M41 infection and the change of respiratory microbiota and metabolomics profiles in broilers that infected with IBV were investigated. According to the experimental findings, the combination of PHI and Bai effectively alleviated broilers' slowing-growth weight and respiratory symptoms. This was accompanied by a reduction in viral copies and histopathological changes, as well as an increase of antiviral protein (G3BP1) level in tracheas and anti-IBV antibody levels in serum. In addition, 16s RNA sequencing revealed that IBV infection significantly changed respiratory microbiota composition at different taxonomic levels and respiratory metabolism composition in broilers. Interestingly, PHI combined with Bai modulated the composition of respiratory microfloras, especially the abundance of Firmicutes and Lactobacillaceae were upregulated, as well as the abundance of Proteobacteria was downregulated. The metabolomics results indicated that PHI combined with Bai involved in glucose, lipids, amino acids and nucleotide metabolism during IBV infection. In summary, PHI combined with Bai exhibited a synergistic effect on preventing infectious bronchitis (IB), with the protection being closely associated with the composition of respiratory microbiota and metabolites. Therefore, adding the mixture of PHI and Bai to the chicken drinking water is recommended to prevent and control IB in clinical.
Collapse
Affiliation(s)
- Haipeng Feng
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730050, China
| | - Jingyan Zhang
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730050, China
| | - Kang Zhang
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730050, China
| | - Xuezhi Wang
- Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Zhiting Guo
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730050, China
| | - Lei Wang
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730050, China
| | - Jianxi Li
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730050, China.
| |
Collapse
|
15
|
Xiao G, Li J, Sun Z. The Combination of Antibiotic and Non-Antibiotic Compounds Improves Antibiotic Efficacy against Multidrug-Resistant Bacteria. Int J Mol Sci 2023; 24:15493. [PMID: 37895172 PMCID: PMC10607837 DOI: 10.3390/ijms242015493] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 10/29/2023] Open
Abstract
Bacterial antibiotic resistance, especially the emergence of multidrug-resistant (MDR) strains, urgently requires the development of effective treatment strategies. It is always of interest to delve into the mechanisms of resistance to current antibiotics and target them to promote the efficacy of existing antibiotics. In recent years, non-antibiotic compounds have played an important auxiliary role in improving the efficacy of antibiotics and promoting the treatment of drug-resistant bacteria. The combination of non-antibiotic compounds with antibiotics is considered a promising strategy against MDR bacteria. In this review, we first briefly summarize the main resistance mechanisms of current antibiotics. In addition, we propose several strategies to enhance antibiotic action based on resistance mechanisms. Then, the research progress of non-antibiotic compounds that can promote antibiotic-resistant bacteria through different mechanisms in recent years is also summarized. Finally, the development prospects and challenges of these non-antibiotic compounds in combination with antibiotics are discussed.
Collapse
Affiliation(s)
| | | | - Zhiliang Sun
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (G.X.); (J.L.)
| |
Collapse
|
16
|
Thai VC, Stubbs KA, Sarkar-Tyson M, Kahler CM. Phosphoethanolamine Transferases as Drug Discovery Targets for Therapeutic Treatment of Multi-Drug Resistant Pathogenic Gram-Negative Bacteria. Antibiotics (Basel) 2023; 12:1382. [PMID: 37760679 PMCID: PMC10525099 DOI: 10.3390/antibiotics12091382] [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: 08/04/2023] [Revised: 08/25/2023] [Accepted: 08/26/2023] [Indexed: 09/29/2023] Open
Abstract
Antibiotic resistance caused by multidrug-resistant (MDR) bacteria is a major challenge to global public health. Polymyxins are increasingly being used as last-in-line antibiotics to treat MDR Gram-negative bacterial infections, but resistance development renders them ineffective for empirical therapy. The main mechanism that bacteria use to defend against polymyxins is to modify the lipid A headgroups of the outer membrane by adding phosphoethanolamine (PEA) moieties. In addition to lipid A modifying PEA transferases, Gram-negative bacteria possess PEA transferases that decorate proteins and glycans. This review provides a comprehensive overview of the function, structure, and mechanism of action of PEA transferases identified in pathogenic Gram-negative bacteria. It also summarizes the current drug development progress targeting this enzyme family, which could reverse antibiotic resistance to polymyxins to restore their utility in empiric therapy.
Collapse
Affiliation(s)
- Van C. Thai
- The Marshall Center for Infectious Diseases Research and Training, School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia; (V.C.T.); (M.S.-T.)
| | - Keith A. Stubbs
- School of Molecular Sciences, University of Western Australia, Crawley, WA 6009, Australia;
| | - Mitali Sarkar-Tyson
- The Marshall Center for Infectious Diseases Research and Training, School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia; (V.C.T.); (M.S.-T.)
| | - Charlene M. Kahler
- The Marshall Center for Infectious Diseases Research and Training, School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia; (V.C.T.); (M.S.-T.)
| |
Collapse
|
17
|
Guo J, Pan Z, Fan L, Zhong Y, Pang R, Su Y. Effect of Three Different Amino Acids Plus Gentamicin Against Methicillin-Resistant Staphylococcus aureus. Infect Drug Resist 2023; 16:4741-4754. [PMID: 37496695 PMCID: PMC10366528 DOI: 10.2147/idr.s411658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/05/2023] [Indexed: 07/28/2023] Open
Abstract
Background The issue of methicillin-resistant Staphylococcus aureus (MRSA) resistant to many antibiotics and causing serious infectious diseases is a growing healthcare concern. Purpose In recent years, exogenous administration of metabolites in combination with antibiotics can re-sensitize resistant bacteria to antibiotics; however, their effects vary, and their underlying mechanism of action remains elusive. Methods We assessed the bactericidal effects of the three amino acids in combination with gentamicin in vitro and in vivo. Subsequently, we explored the role of these amino acids on the metabolomics of MRSA using Liquid chromatography-tandem mass spectrometry (LC-MS/MS). Furthermore, we performed the downstream analyses using MetaboAnalyst and Interactive Pathways Explorer. Results Exogenous threonine showed the best bactericidal efficacy with gentamicin, followed by glycine, wherein serine had no effect. Amino acid treatments mainly up-regulated the metabolites, increased the amino acid abundance, and significantly activated metabolisms; these effects were consistent with the bactericidal efficacy of the three amino acids. Most amino acids participated in the tricarboxylic acid cycle, and threonine supplementation increased the activities of citrate synthase, isocitrate dehydrogenase and α-ketoglutarate dehydrogenase, whereas glycine increased activities of citrate synthase and α-ketoglutarate dehydrogenase, and serine did not affect the activities of any of the three key enzymes. We identified 24 biomarkers in the three groups, among which glutamic acid and cysteine showed a gradient decrease and increase, respectively. Subsequent analyses revealed that glutamic acid but not cysteine promoted the bactericidal effect of gentamicin synergistically. Conclusion Threonine has the best synergistic effect in reversing bacterial resistance compared to glycine and serine. We show that different amino acids combined with an antibiotic mainly affect amino acid metabolism and act via different metabolic regulatory mechanisms, which could help develop effective strategies for tackling MRSA infections.
Collapse
Affiliation(s)
- Juan Guo
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Zhiyu Pan
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Lvyuan Fan
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Yilin Zhong
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Rui Pang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, People’s Republic of China
| | - Yubin Su
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, People’s Republic of China
| |
Collapse
|
18
|
Espinal P, Fusté E, Sierra JM, Jiménez-Galisteo G, Vinuesa T, Viñas M. Progress towards the clinical use of antimicrobial peptides: challenges and opportunities. Expert Opin Biol Ther 2023:1-10. [PMID: 37366927 DOI: 10.1080/14712598.2023.2226796] [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: 03/22/2023] [Accepted: 06/14/2023] [Indexed: 06/28/2023]
Abstract
INTRODUCTION To overcome the challenge of multidrug resistance, natural and synthetic peptides are candidates to become the basis of innovative therapeutics, featuring diverse mechanisms of action. Traditionally, the time elapsed from medical discoveries to their application is long. The urgency derived from the emergence of antibiotic resistance recommends an acceleration of research to put the new weapons in the hands of clinicians. AREAS COVERED This narrative review introduces ideas and suggestions of new strategies that may be used as a basis upon which to recommend reduced development times and to facilitate the arrival of new molecules in the fight against microbes. EXPERT OPINION Although studies on new innovative antimicrobial treatments are being conducted, sooner rather than later, more clinical trials, preclinical and translational research are needed to promote the development of innovative antimicrobial treatments for multidrug resistant infections. The situation is worrying, no less than that generated by pandemics such as the ones we have just experienced and conflicts such as world wars. Although from the point of view of human perception, resistance to antibiotics may not seem as serious as these other situations, it is possibly the hidden pandemic that most jeopardizes the future of medicine.
Collapse
Affiliation(s)
- Paula Espinal
- Laboratory of Molecular Microbiology & Antimicrobials, Department of Pathology & Experimental Therapeutics, Medical School, Campus Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ester Fusté
- Laboratory of Molecular Microbiology & Antimicrobials, Department of Pathology & Experimental Therapeutics, Medical School, Campus Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
- Department of Public Health, Mental Health, And Maternal and Child Health Nursing, University of Barcelona and IDIBELL, Campus Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Josep M Sierra
- Laboratory of Molecular Microbiology & Antimicrobials, Department of Pathology & Experimental Therapeutics, Medical School, Campus Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Guadalupe Jiménez-Galisteo
- Laboratory of Molecular Microbiology & Antimicrobials, Department of Pathology & Experimental Therapeutics, Medical School, Campus Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Teresa Vinuesa
- Laboratory of Molecular Microbiology & Antimicrobials, Department of Pathology & Experimental Therapeutics, Medical School, Campus Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Miguel Viñas
- Laboratory of Molecular Microbiology & Antimicrobials, Department of Pathology & Experimental Therapeutics, Medical School, Campus Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|