1
|
Anguluri K, Sharma B, Bagherpour S, Calpena AC, Halbaut L, Amabilino DB, Kaur G, Chaudhary GR, Pérez-García L. Supramolecular gels for antimicrobial photodynamic therapy against E. coli and S. aureus. Photodiagnosis Photodyn Ther 2025; 52:104529. [PMID: 39988224 DOI: 10.1016/j.pdpdt.2025.104529] [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: 01/10/2025] [Revised: 02/14/2025] [Accepted: 02/21/2025] [Indexed: 02/25/2025]
Abstract
Singlet oxygen (SO) reacts with organic molecules, for example in degrading environmental contaminants and causing toxicity to cells in photodynamic therapy (PDT). The relevance of SO in the environmental and biomedical domains has fuelled research towards improved methodologies for its efficient generation. In this paper, we report the use of a bis-imidazolium-based amphiphile that forms supramolecular gels in water-ethanol mixtures encapsulating SO generating chromophores. The gels comprise twisted fibres that incorporate one of four different porphyrins: 5,10,15,20-tetrakis(N-methyl-4-pyridinium)porphyrin tetraiodide (TPPP), 5,10,15,20-tetrakis(4-carboxyphenyl)porphyrin (TCPP), 5,10,15,20-tetrakis(3-hydroxyphenyl)porphyrin (TPP-3OH) and 5,10,15,20-tetrakis(4-hydroxyphenyl)porphyrin (TPP-4OH). Rheological measurements confirmed viscoelastic properties characteristic of these types of supramolecular gels, suggesting their potential as effective local PDT delivery systems. Enhanced SO generation within the hydrogel matrix compared to the solution of the chromophores was observed in suspension in a spectrophotometer using uric acid as the molecular probe. The SO generation was also shown through antimicrobial PDT (aPDT) studies. The TPPP-containing gel showed the highest reduction in the colony forming unit (CFU) count, which is 94% against E. coli and 100% against S. aureus. These results indicate that the porphyrin gels based on a gemini amphiphile with a high level of SO production are of significant interest and have a lot of potential use in aPDT.
Collapse
Affiliation(s)
- Kavya Anguluri
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Avda. Joan XXIII 27-31, Universitat de Barcelona, 08028 Barcelona, Spain; Institut de Nanociència i Nanotecnologia IN2UB, Universitat de Barcelona, Barcelona, 08028 Spain
| | - Bunty Sharma
- Department of SAIF/CIL, Panjab University, Chandigarh, 160014, India
| | - Saman Bagherpour
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Avda. Joan XXIII 27-31, Universitat de Barcelona, 08028 Barcelona, Spain; Institut de Nanociència i Nanotecnologia IN2UB, Universitat de Barcelona, Barcelona, 08028 Spain
| | - Ana C Calpena
- Institut de Nanociència i Nanotecnologia IN2UB, Universitat de Barcelona, Barcelona, 08028 Spain; Departament de Farmàcia, Tecnologia Farmacèutica, i Fisicoquímica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain
| | - Lyda Halbaut
- Departament de Farmàcia, Tecnologia Farmacèutica, i Fisicoquímica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain
| | - David B Amabilino
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC), Carrer dels Til.lers, 08193 Bellaterra, Spain
| | - Gurpreet Kaur
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh, 160014, India
| | - Ganga Ram Chaudhary
- Department of SAIF/CIL, Panjab University, Chandigarh, 160014, India; Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh, 160014, India
| | - Lluïsa Pérez-García
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Avda. Joan XXIII 27-31, Universitat de Barcelona, 08028 Barcelona, Spain; Institut de Nanociència i Nanotecnologia IN2UB, Universitat de Barcelona, Barcelona, 08028 Spain.
| |
Collapse
|
2
|
Tao W, Li W, Jin R, Liang D, Weng W, Lin R, Yang S. BCP4: A novel antimicrobial peptide with potent efficacy against Bacillus cereus in rice porridge. Int J Food Microbiol 2025; 429:111001. [PMID: 39631214 DOI: 10.1016/j.ijfoodmicro.2024.111001] [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: 07/11/2024] [Revised: 11/12/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Bacillus cereus is a common foodborne pathogen that frequently contaminates rice products and produces cereulide toxins, presenting a significant risk to food safety and human health. In contrast, Bacillus subtilis is a promising source of antimicrobial peptides (AMPs). In this research, a novel AMP named BCP4 (KGKTLLQ) was discovered through the fermentation of shrimp waste with B. subtilis, which speculated that BCP4 might be generated through enzymatic hydrolysis catalyzed by endogenous enzymes naturally present in shrimp waste. BCP4 demonstrated potent antibacterial activity against B. cereus with a minimum bactericidal concentration (MBC) of 62.5 μg/mL and bacterial time-kill of 3 h. BCP4 surpassed the bactericidal efficiency of nisin (500 μg/mL), a commonly used AMP of microbial origin. BCP4 operates by causing damage to the bacterial cell wall and membrane, which allows the contents of the cell to flow out. BCP4 also penetrates the cell membrane and binds with DNA, effectively sterilizing the bacteria. Meanwhile, treatment of BCP4 with mammalian red blood cells revealed that it was nonhemolytic. Furthermore, the growth of B. cereus in rice porridge was significantly inhibited by BCP4 at a concentration of 62.5 μg/mL. This study provides a theoretical basis for using BCP4 to control B. cereus contamination.
Collapse
Affiliation(s)
- Weihong Tao
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Wenjie Li
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Ritian Jin
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China; Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Duo Liang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Wuyin Weng
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Rong Lin
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China; Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Jimei University, Xiamen, Fujian 361021, China.
| | - Shen Yang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China; Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Jimei University, Xiamen, Fujian 361021, China.
| |
Collapse
|
3
|
Heidarpanah S, Li K, Thibodeau A, Meniaï I, Parreira VR, Quessy S, Segura M, Fittipaldi N, Gaucher ML. Genomic Diversity and Virulence Factors of Clostridium perfringens Isolated from Healthy and Necrotic Enteritis-Affected Broiler Chicken Farms in Quebec Province. Microorganisms 2024; 12:2624. [PMID: 39770825 PMCID: PMC11677781 DOI: 10.3390/microorganisms12122624] [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: 11/26/2024] [Revised: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Avian necrotic enteritis due to the Gram-positive bacterium Clostridium perfringens has re-emerged following the ban on antibiotic growth promoters in many poultry producing countries. The limited number of previous studies has left important gaps in our understanding of the genetic diversity and virulence traits of the pathogen. To address these knowledge gaps, in this study, we sequenced the genomes of 41 Clostridium perfringens isolates recovered from commercial broiler chicken flocks in Quebec, Canada, including isolates from healthy birds and those affected by necrotic enteritis. We sought to understand the pangenome diversity and interrogated the genomes for key virulence factors involved in necrotic enteritis pathogenesis. On average, the genomes had a GC content of 28% and contained 3206 coding sequences. A variable presence of toxins, degradative hydrolytic enzymes, and collagen-binding proteins was also found. Through pangenome analysis, we revealed a total of 10,223 genes, 652 (6.4%) of which formed the core genome. Additionally, we identified 17 different plasmids, 12 antibiotic resistance genes, and nine prophage regions. Overall, our results demonstrated a relatively high genetic diversity among chicken Clostridium perfringens isolates collected from the same geographical location, offering new insights into potential virulence mechanisms and adaptation of the pathogen within poultry populations.
Collapse
Affiliation(s)
- Sara Heidarpanah
- Chaire de Recherche en Salubrité des Viandes (CRSV), Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.H.); (A.T.); (I.M.); (S.Q.)
- Swine and Poultry Infectious Diseases Research Centre (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada; (K.L.); (M.S.); (N.F.)
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Kevin Li
- Swine and Poultry Infectious Diseases Research Centre (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada; (K.L.); (M.S.); (N.F.)
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Groupe de Recherche sur les Maladies Infectieuses en Production Animale (GREMIP), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Alexandre Thibodeau
- Chaire de Recherche en Salubrité des Viandes (CRSV), Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.H.); (A.T.); (I.M.); (S.Q.)
- Swine and Poultry Infectious Diseases Research Centre (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada; (K.L.); (M.S.); (N.F.)
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Groupe de Recherche sur les Maladies Infectieuses en Production Animale (GREMIP), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Ilhem Meniaï
- Chaire de Recherche en Salubrité des Viandes (CRSV), Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.H.); (A.T.); (I.M.); (S.Q.)
- Swine and Poultry Infectious Diseases Research Centre (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada; (K.L.); (M.S.); (N.F.)
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Valeria R. Parreira
- Canadian Research Institute for Food Safety (CRIFS), Food Science Department, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Sylvain Quessy
- Chaire de Recherche en Salubrité des Viandes (CRSV), Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.H.); (A.T.); (I.M.); (S.Q.)
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Mariela Segura
- Swine and Poultry Infectious Diseases Research Centre (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada; (K.L.); (M.S.); (N.F.)
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Groupe de Recherche sur les Maladies Infectieuses en Production Animale (GREMIP), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Nahuel Fittipaldi
- Swine and Poultry Infectious Diseases Research Centre (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada; (K.L.); (M.S.); (N.F.)
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Groupe de Recherche sur les Maladies Infectieuses en Production Animale (GREMIP), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Marie-Lou Gaucher
- Chaire de Recherche en Salubrité des Viandes (CRSV), Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.H.); (A.T.); (I.M.); (S.Q.)
- Swine and Poultry Infectious Diseases Research Centre (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada; (K.L.); (M.S.); (N.F.)
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Groupe de Recherche sur les Maladies Infectieuses en Production Animale (GREMIP), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| |
Collapse
|
4
|
Huang J, Hong X, Lv Y, Wang Y, Han K, Zhu C, Xie L. Armored polymyxin B: a nanosystem for combating multidrug-resistant Gram-negative bacilli. RSC Adv 2024; 14:39700-39707. [PMID: 39691227 PMCID: PMC11650604 DOI: 10.1039/d4ra07577c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 11/28/2024] [Indexed: 12/19/2024] Open
Abstract
Bacterial infections cause high morbidity and mortality worldwide, and the emergence of drug-resistant bacteria further complicates the treatment of infections. Therefore, it is necessary to continuously develop new treatment methods. Polymyxin B (PMB), as the last line of defense, can combat most aerobic Gram-negative bacilli including common drug-resistant bacteria in clinical practice. However, the suboptimal lung tissue concentration of PMB and dose-dependent nephrotoxicity and neurotoxicity limit its clinical application. The nanodrug delivery system offers several key advantages, including high drug loading capacity, excellent biocompatibility, controlled release mechanisms, and targeted delivery. These features enhance the bioavailability of drugs while simultaneously reducing their toxicity and minimizing side effects. In this study, we designed a targeted nanodrug delivery system (PMB@HMnO2@NM) consisting of hollow mesoporous manganese dioxide (HMnO2) coated with neutrophil membrane (NM). In a mouse model of acute pneumonia induced by multidrug-resistant Pseudomonas aeruginosa, treatment with PMB@HMnO2@NM demonstrated the ability to target bacterial aggregation and specifically deliver the drug to the infected lung tissue. This targeted approach resulted in improved survival rates and reduced inflammatory damage without causing adverse effects. The findings of this study suggest the potential for developing a new class of multifunctional nanodrugs, providing new therapeutic strategies for multidrug-resistant (MDR) bacterial infections. Furthermore, these results provide a solid foundation for the design of biomimetic nanosized antibacterial drugs.
Collapse
Affiliation(s)
- Jianling Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Bengbu Medical University, Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Molecular Diagnosis Center Bengbu Anhui 233000 China
| | - Xiuwen Hong
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu 210011 China
| | - Yunxiang Lv
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Bengbu Medical University, Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Molecular Diagnosis Center Bengbu Anhui 233000 China
| | - Yueyue Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Bengbu Medical University, Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Molecular Diagnosis Center Bengbu Anhui 233000 China
| | - Kexing Han
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Bengbu Medical University, Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Molecular Diagnosis Center Bengbu Anhui 233000 China
| | - Chenghua Zhu
- Nanjing Pukou Hospital of TCM, Pukou Hospital of Chinese Medicine Affiliated to China Pharmaceutical University Nanjing 210000 China
| | - Lixu Xie
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University Wenhua Xi Road 107# Jinan 250012 China
| |
Collapse
|
5
|
Li H, E W, Zhao D, Liu H, Pei J, Du B, Liu K, Zhu X, Wang C. Response of Paenibacillus polymyxa SC2 to the stress of polymyxin B and a key ABC transporter YwjA involved. Appl Microbiol Biotechnol 2024; 108:17. [PMID: 38170316 DOI: 10.1007/s00253-023-12916-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/24/2023] [Accepted: 10/04/2023] [Indexed: 01/05/2024]
Abstract
Polymyxins are cationic peptide antibiotics and regarded as the "final line of defense" against multidrug-resistant bacterial infections. Meanwhile, some polymyxin-resistant strains and the corresponding resistance mechanisms have also been reported. However, the response of the polymyxin-producing strain Paenibacillus polymyxa to polymyxin stress remains unclear. The purpose of this study was to investigate the stress response of gram-positive P. polymyxa SC2 to polymyxin B and to identify functional genes involved in the stress response process. Polymyxin B treatment upregulated the expression of genes related to basal metabolism, transcriptional regulation, transport, and flagella formation and increased intracellular ROS levels, flagellar motility, and biofilm formation in P. polymyxa SC2. Adding magnesium, calcium, and iron alleviated the stress of polymyxin B on P. polymyxa SC2, furthermore, magnesium and calcium could improve the resistance of P. polymyxa SC2 to polymyxin B by promoting biofilm formation. Meanwhile, functional identification of differentially expressed genes indicated that an ABC superfamily transporter YwjA was involved in the stress response to polymyxin B of P. polymyxa SC2. This study provides an important reference for improving the resistance of P. polymyxa to polymyxins and increasing the yield of polymyxins. KEY POINTS: • Phenotypic responses of P. polymyxa to polymyxin B was performed and indicated by RNA-seq • Forming biofilm was a key strategy of P. polymyxa to alleviate polymyxin stress • ABC transporter YwjA was involved in the stress resistance of P. polymyxa to polymyxin B.
Collapse
Affiliation(s)
- Hui Li
- College of Life Sciences, National Engineering Research Center for Efficient Utilization of Soil and Fertilizer Resources, Shandong Engineering Research Center of Plant-Microbia Restoration for Saline-Alkali Land, Shandong Key Laboratory of Agricultural Microbiology, Shandong Agricultural University, Tai'an, 271018, China
| | - Wenhui E
- College of Life Sciences, National Engineering Research Center for Efficient Utilization of Soil and Fertilizer Resources, Shandong Engineering Research Center of Plant-Microbia Restoration for Saline-Alkali Land, Shandong Key Laboratory of Agricultural Microbiology, Shandong Agricultural University, Tai'an, 271018, China
| | - Dongying Zhao
- College of Life Sciences, National Engineering Research Center for Efficient Utilization of Soil and Fertilizer Resources, Shandong Engineering Research Center of Plant-Microbia Restoration for Saline-Alkali Land, Shandong Key Laboratory of Agricultural Microbiology, Shandong Agricultural University, Tai'an, 271018, China
| | - Haiyang Liu
- College of Life Sciences, National Engineering Research Center for Efficient Utilization of Soil and Fertilizer Resources, Shandong Engineering Research Center of Plant-Microbia Restoration for Saline-Alkali Land, Shandong Key Laboratory of Agricultural Microbiology, Shandong Agricultural University, Tai'an, 271018, China
| | - Jian Pei
- College of Life Sciences, National Engineering Research Center for Efficient Utilization of Soil and Fertilizer Resources, Shandong Engineering Research Center of Plant-Microbia Restoration for Saline-Alkali Land, Shandong Key Laboratory of Agricultural Microbiology, Shandong Agricultural University, Tai'an, 271018, China
| | - Binghai Du
- College of Life Sciences, National Engineering Research Center for Efficient Utilization of Soil and Fertilizer Resources, Shandong Engineering Research Center of Plant-Microbia Restoration for Saline-Alkali Land, Shandong Key Laboratory of Agricultural Microbiology, Shandong Agricultural University, Tai'an, 271018, China
| | - Kai Liu
- College of Life Sciences, National Engineering Research Center for Efficient Utilization of Soil and Fertilizer Resources, Shandong Engineering Research Center of Plant-Microbia Restoration for Saline-Alkali Land, Shandong Key Laboratory of Agricultural Microbiology, Shandong Agricultural University, Tai'an, 271018, China
| | - Xueming Zhu
- State Key Laboratory for Managing Biotic and Chemical Treats to the Quality and Safety of Agro-Products, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Chengqiang Wang
- College of Life Sciences, National Engineering Research Center for Efficient Utilization of Soil and Fertilizer Resources, Shandong Engineering Research Center of Plant-Microbia Restoration for Saline-Alkali Land, Shandong Key Laboratory of Agricultural Microbiology, Shandong Agricultural University, Tai'an, 271018, China.
| |
Collapse
|
6
|
Song D, Kim B, Kim M, Lee JK, Choi J, Lee H, Shin S, Shin D, Nam HY, Lee Y, Lee S, Kim Y, Seo J. Impact of Conjugation of the Reactive Oxygen Species (ROS)-Generating Catalytic Moiety with Membrane-Active Antimicrobial Peptoids: Promoting Multitarget Mechanism and Enhancing Selectivity. J Med Chem 2024; 67:15148-15167. [PMID: 39207209 DOI: 10.1021/acs.jmedchem.4c00775] [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: 09/04/2024]
Abstract
Antimicrobial peptides (AMPs) represent promising therapeutic modalities against multidrug-resistant bacterial infections. As a mimic of natural AMPs, peptidomimetic oligomers like peptoids (i.e., oligo-N-substituted glycines) have been utilized for antimicrobials with resistance against proteolytic degradation. Here, we explore the conjugation of catalytic metal-binding motifs─the amino terminal Cu(II) and Ni(II) binding (ATCUN) motif─with cationic amphipathic antimicrobial peptoids to enhance their efficacy. Upon complexation with Cu(II) or Ni(II), the conjugates catalyzed hydroxyl radical generation, and 22 and 22-Cu exhibited over 10-fold improved selectivity compared to the parent peptoid, likely due to reduced hydrophobicity. Cu-ATCUN-peptoids caused bacterial membrane disruption, aggregation of intracellular biomolecules, DNA oxidation, and lipid peroxidation, promoting multiple killing mechanisms. In a mouse sepsis model, 22 demonstrated antimicrobial and anti-inflammatory efficacy with low toxicity. This study suggests a strategy to improve the potency of membrane-acting antimicrobial peptoids by incorporating ROS-generating motifs, thereby adding oxidative damage as a killing mechanism.
Collapse
Affiliation(s)
- Dasom Song
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Byeongkwon Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Minsang Kim
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Jin Kyeong Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Jieun Choi
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Hyeju Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Sujin Shin
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Dongmin Shin
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Ho Yeon Nam
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Yunho Lee
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Seongsoo Lee
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju 61751, Republic of Korea
- Department of Systems Biotechnology, Chung-Ang University, Anseong, Gyeonggi 17546, Republic of Korea
- Department of Bio-Analysis Science, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Yangmee Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Jiwon Seo
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| |
Collapse
|
7
|
Huang J, Hong X, Chen S, He Y, Xie L, Gao F, Zhu C, Jin X, Yan H, Ye Y, Shao M, Du X, Feng G. Biomimetic Metal-Organic Framework Gated Nanoplatform for Sonodynamic Therapy against Extensively Drug Resistant Bacterial Lung Infection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402473. [PMID: 38962911 PMCID: PMC11434100 DOI: 10.1002/advs.202402473] [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: 03/08/2024] [Revised: 06/03/2024] [Indexed: 07/05/2024]
Abstract
Novel antimicrobial strategies are urgently needed to treat extensively drug-resistant (XDR) bacterial infections due to the high mortality rate and lack of effective therapeutic agents. Herein, nanoengineered human umbilical cord mesenchymal stem cells (hUC-MSCs), named PMZMU, are designed as a sonosensitizer for synergistic sonodynamic-nano-antimicrobial therapy against gram-negative XDR bacteria. PMZMU is composed of a bacterial targeting peptide (UBI29-41) modified hUC-MSCs membrane (MSCm), a sonosensitizer meso-tetra(4-car-boxyphenyl) porphine doped mesoporous organo-silica nanoparticle and an acidity-responsive metal-organic framework ZIF-8. This innovative formulation enables efficient loading of polymyxin B, reduces off-target drug release, increases circulation and targeting efficacy, and generates reactive oxygen species upon ultrasound irradiation. PMZMU exhibits remarkable in vitro inhibitory activity against four XDR bacteria: Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa (PA), and Escherichia coli. Taking advantage of the bacterial targeting ability of UBI29-41 and the inflammatory chemotaxis of hUC-MSC, PMZMU can be precisely delivered to lung infection sites thereby augmenting polymyxin B concentration. PMZMU-mediated sonodynamic therapy significantly reduces bacterial burden, relieves inflammatory damage by promoting the polarization of macrophages toward M2 phenotype, and improves survival rates without introducing adverse events. Overall, this study offers promising strategies for treating deep-tissue XDR bacterial infections, and guides the design and optimization of biomimetic nanomedicine.
Collapse
Affiliation(s)
- Jianling Huang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Xiuwen Hong
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Sixi Chen
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Yucong He
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Lixu Xie
- Department of Pulmonary and Critical Care Medicine, Qi Lu Hospital of Shandong University, Wen hua xi Road 107#, Jinan, 250012, China
| | - Fenglin Gao
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Chenghua Zhu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Xiao Jin
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Haihao Yan
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Yongxia Ye
- Department of Radiology, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, 210009, China
| | - Mingyue Shao
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Xingran Du
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, China
| | - Ganzhu Feng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| |
Collapse
|
8
|
Tüzemen NÜ, Önal U, Merdan O, Akca B, Ener B, Özakın C, Akalın H. Synergistic antibacterial activity of ceftazidime-avibactam in combination with colistin, gentamicin, amikacin, and fosfomycin against carbapenem-resistant Klebsiella pneumoniae. Sci Rep 2024; 14:17567. [PMID: 39080317 PMCID: PMC11289488 DOI: 10.1038/s41598-024-67347-5] [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: 01/30/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024] Open
Abstract
Carbapenem-resistant Klebsiella pneumoniae (CPKP) infections seriously threaten global public health. The main objective of this study was to assess the in-vitro synergistic activity of ceftazidime-avibactam (CZA) in combination with colistin (COL), amikacin (AK), gentamicin (GEN), and fosfomycin (FOS) against CPKP isolates. The secondary goal was to determine the antibiotic susceptibility performance of BD Phoenix. OXA-48 (49.1%) was the predominant carbapenemase, followed by KPC (29.1%). We used the broth microdilution (BMD) method to determine the minimum inhibitory concentrations (MICs) of CZA, COL, AK, and GEN. Meanwhile, the MICs of FOS were determined by the agar dilution (AD) method. To examine the antibacterial activity of CZA, we conducted a checkerboard assay (CBA) with COL, AK, GEN, and FOS against CRKP isolates. We randomly selected three strains and performed synergy testing via time-kill assay (TKA). CRKP isolates were 89.1% susceptible to CZA, 16.4% to COL, 21.8% to GEN, and 29.1% to AK using BMD, 47.3% to FOS by AD. The most synergistic effects were observed in the combination of CZA-COL (78.2%) and CZA-FOS (63.6%). Given the limited therapeutic options for treating severe CRKP infections, combining CZA with COL and FOS may enhance in-vitro activity against clinical CRKP isolates.
Collapse
Affiliation(s)
- Nazmiye Ülkü Tüzemen
- Faculty of Medicine, Department of Medical Microbiology, Bursa Uludag University, 16059 Görükle, Bursa, Turkey.
| | - Uğur Önal
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Bursa Uludag University, Bursa, Turkey
| | - Osman Merdan
- Faculty of Medicine, Department of Medical Microbiology, Bursa Uludag University, 16059 Görükle, Bursa, Turkey
- Institute of Virology, Technical University of Munich, Munich, Germany
| | - Bekir Akca
- Faculty of Medicine, Department of Medical Microbiology, Bursa Uludag University, 16059 Görükle, Bursa, Turkey
| | - Beyza Ener
- Faculty of Medicine, Department of Medical Microbiology, Bursa Uludag University, 16059 Görükle, Bursa, Turkey
| | - Cüneyt Özakın
- Faculty of Medicine, Department of Medical Microbiology, Bursa Uludag University, 16059 Görükle, Bursa, Turkey
| | - Halis Akalın
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Bursa Uludag University, Bursa, Turkey
| |
Collapse
|
9
|
Rudzite M, O’Toole GA. An energy coupling factor transporter of Streptococcus sanguinis impacts antibiotic susceptibility as well as metal and membrane homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.12.603315. [PMID: 39026867 PMCID: PMC11257530 DOI: 10.1101/2024.07.12.603315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Streptococcus sanguinis is a prevalent member of human microbiome capable of acting as a causative agent of oral and respiratory infections. S. sanguinis competitive success within the infection niche is dependent on acquisition of metal ions and vitamins. Among the systems that bacteria use for micronutrient uptake is the energy coupling factor (ECF) transporter system EcfAAT. Here we describe physiological changes arising from EcfAAT transporter disruption. We found that EcfAAT contributes to S. sanguinis antibiotic sensitivity as well as metal and membrane homeostasis. Specifically, our work found that disruption of EcfAAT results in increased polymyxin susceptibility. We performed assessment of cell-associated metal content and found depletion of iron, magnesium, and manganese. Furthermore, membrane composition analysis revealed significant enrichment in unsaturated fatty acid species resulting in increased membrane fluidity. Our results demonstrate how disruption of a single EcfAAT transporter can have broad consequences on bacterial cell homeostasis. ECF transporters are of interest within the context of infection biology in bacterial species other than streptococci, hence work described here will further the understanding of how micronutrient uptake systems contribute to bacterial pathogenesis.
Collapse
Affiliation(s)
- Marta Rudzite
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - G. A. O’Toole
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| |
Collapse
|
10
|
Leseigneur C, Mondange L, Pizarro-Cerdá J, Dussurget O. Staphylococcus aureus NAD kinase is required for envelop and antibiotic stress responses. Microbes Infect 2024; 26:105334. [PMID: 38556158 DOI: 10.1016/j.micinf.2024.105334] [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: 10/27/2023] [Revised: 03/09/2024] [Accepted: 03/28/2024] [Indexed: 04/02/2024]
Abstract
Global burden of infectious diseases and antimicrobial resistance are major public health issues calling for innovative control measures. Bacterial NAD kinase (NADK) is a crucial enzyme for production of NADP(H) and growth. In Staphylococcus aureus, NADK promotes pathogenesis by supporting production of key virulence determinants. Here, we find that knockdown of NADK by CRISPR interference sensitizes S. aureus to osmotic stress and to stresses induced by antibiotics targeting the envelop as well as replication, transcription and translation. Thus, NADK represents a promising target for the development of inhibitors which could be used in combination with current antibiotics.
Collapse
Affiliation(s)
- Clarisse Leseigneur
- Institut Pasteur, Université Paris Cité, Microbiology Department, CNRS UMR6047, Yersinia Research Unit, 28 rue du Dr Roux, 75015 Paris, France
| | - Lou Mondange
- Institut Pasteur, Université Paris Cité, Microbiology Department, CNRS UMR6047, Yersinia Research Unit, 28 rue du Dr Roux, 75015 Paris, France; Institut de Recherche Biomédicale des Armées, Microbiology and Infectious Diseases Department, Bacteriology Unit, 91220 Brétigny-sur-Orge, France
| | - Javier Pizarro-Cerdá
- Institut Pasteur, Université Paris Cité, Microbiology Department, CNRS UMR6047, Yersinia Research Unit, 28 rue du Dr Roux, 75015 Paris, France
| | - Olivier Dussurget
- Institut Pasteur, Université Paris Cité, Microbiology Department, CNRS UMR6047, Yersinia Research Unit, 28 rue du Dr Roux, 75015 Paris, France.
| |
Collapse
|
11
|
Medaglia S, Otri I, Bernardos A, Marcos MD, Aznar E, Sancenón F, Martínez-Máñez R. Synergistic antimicrobial photodynamic therapy using gated mesoporous silica nanoparticles containing curcumin and polymyxin B. Int J Pharm 2024; 654:123947. [PMID: 38408553 DOI: 10.1016/j.ijpharm.2024.123947] [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: 11/20/2023] [Revised: 02/14/2024] [Accepted: 02/23/2024] [Indexed: 02/28/2024]
Abstract
Photodynamic Therapy is a therapy based on combining a non-toxic compound, known as photosensitizer (PS), and irradiation with light of the appropriate wavelength to excite the PS molecule. The photon absorption by the PS leads to reactive oxygen species generation and a subsequent oxidative burst that causes cell damage and death. In this work, we report an antimicrobial nanodevice that uses the activity of curcumin (Cur) as a PS for antimicrobial Photodynamic Therapy (aPDT), based on mesoporous silica nanoparticles in which the action of the classical antibiotic PMB is synergistically combined with the aPDT properties of curcumin to combat bacteria. The synergistic effect of the designed gated device in combination with irradiation with blue LED light (470 nm) is evaluated against Escherichia coli, Pseudomonas aeruginosa and Staphylococcus epidermidis. The results show that the nanodevice exhibits a noteworthy antibacterial activity against these microorganisms, a much more significant effect than free Cur and PMB at equivalent concentrations. Thus, 0.1 µg/mL of MSNs-Cur-PMB eliminates a bacterial concentration of about 105 CFU/mL of E. coli, while 1 µg/mL of MSNs-Cur-PMB is required for P. aeruginosa and S. epidermidis. In addition, antibiofilm activity against the selected bacteria was also tested. We found that 0.1 mg/mL of MSNs-Cur-PMB inhibited 99 % biofilm formation for E. coli, and 1 mg/mL of MSNs-Cur-PMB achieved 90 % and 100 % inhibition of biofilm formation for S. epidermidis and P. aeruginosa, respectively.
Collapse
Affiliation(s)
- Serena Medaglia
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de Valencia, Universitat de València, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ismael Otri
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de Valencia, Universitat de València, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Andrea Bernardos
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de Valencia, Universitat de València, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; Departamento de Química, Universidad Politécnica de Valencia, Cami de Vera s/n, 46022 Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - María Dolores Marcos
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de Valencia, Universitat de València, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; Departamento de Química, Universidad Politécnica de Valencia, Cami de Vera s/n, 46022 Valencia, Spain; Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, Instituto de Investigación Sanitaria La Fe (IISLAFE), Av Fernando Abril Martorell 106, 46026 Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, Valencia, Spain.
| | - Elena Aznar
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de Valencia, Universitat de València, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; Departamento de Química, Universidad Politécnica de Valencia, Cami de Vera s/n, 46022 Valencia, Spain; Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, Instituto de Investigación Sanitaria La Fe (IISLAFE), Av Fernando Abril Martorell 106, 46026 Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, Valencia, Spain.
| | - Félix Sancenón
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de Valencia, Universitat de València, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; Departamento de Química, Universidad Politécnica de Valencia, Cami de Vera s/n, 46022 Valencia, Spain; Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, Instituto de Investigación Sanitaria La Fe (IISLAFE), Av Fernando Abril Martorell 106, 46026 Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Ramón Martínez-Máñez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de Valencia, Universitat de València, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; Departamento de Química, Universidad Politécnica de Valencia, Cami de Vera s/n, 46022 Valencia, Spain; Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, Instituto de Investigación Sanitaria La Fe (IISLAFE), Av Fernando Abril Martorell 106, 46026 Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, Valencia, Spain
| |
Collapse
|
12
|
Hale SJM, Cameron AJ, Lux CA, Biswas K, Kim R, O'Carroll M, Harris PWR, Douglas RG, Wagner Mackenzie B. Polymyxin B and ethylenediaminetetraacetic acid act synergistically against Pseudomonas aeruginosa and Staphylococcus aureus. Microbiol Spectr 2024; 12:e0170923. [PMID: 38168683 PMCID: PMC10845947 DOI: 10.1128/spectrum.01709-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 10/05/2023] [Indexed: 01/05/2024] Open
Abstract
Polymyxin B and ethylenediaminetetraacetic acid are antimicrobials possessing antibiofilm activity. They act by displacement and chelation, respectively, of divalent cations in bacterial membranes and may therefore act synergistically when applied in combination. If so, this combination of agents may be useful for the treatment of diseases like cystic fibrosis (CF), in which biofilms are present on the respiratory epithelium. We used checkerboard assays to investigate the synergy between these agents using reference strains Pseudomonas aeruginosa ATCC 27853 and Staphylococcus aureus ATCC 6538 in planktonic form. We then determined the efficacy of each agent against biofilms of both species grown on 96-pin lids and proceeded to combination testing against the P. aeruginosa reference strain and 10 clinical isolates from patients with CF. Synergism was observed for planktonic forms of both species and for biofilms of P. aeruginosa. The susceptibility of biofilms of P. aeruginosa clinical isolates to these agents was variable compared to the laboratory reference strain. This combination of agents may be useful in the management of biofilm-associated conditions, particularly those amenable to topical therapies. These results provide a basis upon which the antimicrobial and antibiofilm efficacy of preparations containing these agents may be enhanced.IMPORTANCEBacteria living in biofilms produce a protective matrix which makes them difficult to kill. Patients with severe respiratory disease often have biofilms. Polymyxin B is an antibiotic commonly used in topical medications, such as eye drops and nasal sprays. Ethylenediaminetetraacetic acid (EDTA) is used widely as a preservative in medication but also has antimicrobial properties. It has been hypothesized that Polymyxin B and EDTA could have a synergistic relationship: when used in combination their antimicrobial effect is enhanced. Here, we evaluated the levels at which Polymyxin B and EDTA work together to kill common pathogens Pseudomonas aeruginosa and Staphylococcus aureus. We found that Polymyxin B and EDTA were synergistic. This synergy may be useful in the management of planktonic infection with P. aeruginosa and S. aureus, or biofilm infection with P. aeruginosa. This synergy may be beneficial in the treatment of respiratory biofilms, in which P. aeruginosa biofilms are common.
Collapse
Affiliation(s)
- Samuel J M Hale
- Department of Surgery, Faculty of Medical and Health Sciences, The University of Auckland, Grafton, Auckland, New Zealand
| | - Alan J Cameron
- School of Chemical Sciences and School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Christian A Lux
- Department of Surgery, Faculty of Medical and Health Sciences, The University of Auckland, Grafton, Auckland, New Zealand
| | - Kristi Biswas
- Department of Surgery, Faculty of Medical and Health Sciences, The University of Auckland, Grafton, Auckland, New Zealand
| | - Raymond Kim
- Department of Surgery, Faculty of Medical and Health Sciences, The University of Auckland, Grafton, Auckland, New Zealand
| | - Mark O'Carroll
- Respiratory Services, Auckland City Hospital, Te Toka Tumai, Te Whatu Ora, Auckland, New Zealand
| | - Paul W R Harris
- School of Chemical Sciences and School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Richard G Douglas
- Department of Surgery, Faculty of Medical and Health Sciences, The University of Auckland, Grafton, Auckland, New Zealand
| | - Brett Wagner Mackenzie
- Department of Surgery, Faculty of Medical and Health Sciences, The University of Auckland, Grafton, Auckland, New Zealand
| |
Collapse
|
13
|
Salas-Orozco MF, Lorenzo-Leal AC, de Alba Montero I, Marín NP, Santana MAC, Bach H. Mechanism of escape from the antibacterial activity of metal-based nanoparticles in clinically relevant bacteria: A systematic review. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 55:102715. [PMID: 37907198 DOI: 10.1016/j.nano.2023.102715] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/05/2023] [Accepted: 10/05/2023] [Indexed: 11/02/2023]
Abstract
The emergency of antibiotic-resistant bacteria in severe infections is increasing, especially in nosocomial environments. The ESKAPE group is of special importance in the groups of multi-resistant bacteria due to its high capacity to generate resistance to antibiotics and bactericides. Therefore, metal-based nanomaterials are an attractive alternative to combat them because they have been demonstrated to damage biomolecules in the bacterial cells. However, there is a concern about bacteria developing resistance to NPs and their harmful effects due to environmental accumulation. Therefore, this systematic review aims to report the clinically relevant bacteria that have developed resistance to the NPs. According to the results of this systematic review, various mechanisms to counteract the antimicrobial activity of various NP types have been proposed. These mechanisms can be grouped into the following categories: production of extracellular compounds, metal efflux pumps, ROS response, genetic changes, DNA repair, adaptative morphogenesis, and changes in the plasma membrane.
Collapse
Affiliation(s)
- Marco Felipe Salas-Orozco
- Facultad de Estomatología, Doctorado en Ciencias Odontológicas, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico.
| | - Ana Cecilia Lorenzo-Leal
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | - Nuria Patiño Marín
- Facultad de Estomatología, Laboratorio de Investigación Clinica, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Miguel Angel Casillas Santana
- Maestría en Estomatología con Opcion Terminal en Ortodoncia, Facultad de Estomatología, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Horacio Bach
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
14
|
Upender I, Yoshida O, Schrecengost A, Ranson H, Wu Q, Rowley DC, Kishore S, Cywes C, Miller EL, Whalen KE. A marine-derived fatty acid targets the cell membrane of Gram-positive bacteria. J Bacteriol 2023; 205:e0031023. [PMID: 37905811 PMCID: PMC10662121 DOI: 10.1128/jb.00310-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 09/25/2023] [Indexed: 11/02/2023] Open
Abstract
IMPORTANCE With the lack of new antibiotics in the drug discovery pipeline, coupled with accelerated evolution of antibiotic resistance, new sources of antibiotics that target pathogens of clinical importance are paramount. Here, we use bacterial cytological profiling to identify the mechanism of action of the monounsaturated fatty acid (Z)-13-methyltetra-4-decenoic acid isolated from the marine bacterium Olleya marilimosa with antibacterial effects against Gram-positive bacteria. The fatty acid antibiotic was found to rapidly destabilize the cell membrane by pore formation and membrane aggregation in Bacillus subtilis, suggesting that this fatty acid may be a promising adjuvant used in combination to enhance antibiotic sensitivity.
Collapse
Affiliation(s)
- Isha Upender
- Department of Biology, Haverford College, Haverford, Pennsylvania, USA
| | - Olivia Yoshida
- Department of Biology, Haverford College, Haverford, Pennsylvania, USA
| | - Anna Schrecengost
- Department of Biology, Haverford College, Haverford, Pennsylvania, USA
| | - Hilary Ranson
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, USA
- College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
| | - Qihao Wu
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, USA
- College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
| | - David C. Rowley
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, USA
- College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
| | - Shreya Kishore
- Department of Biology, Haverford College, Haverford, Pennsylvania, USA
| | - Claire Cywes
- Department of Biology, Haverford College, Haverford, Pennsylvania, USA
| | - Eric L. Miller
- Department of Biology, Haverford College, Haverford, Pennsylvania, USA
| | - Kristen E. Whalen
- Department of Biology, Haverford College, Haverford, Pennsylvania, USA
| |
Collapse
|
15
|
Wu Y, Wu P, Wu R, Li H, Duan Y, Cai C, Liu Z, She P, Zhang D. Simeprevir restores the anti-Staphylococcus activity of polymyxins. AMB Express 2023; 13:122. [PMID: 37917339 PMCID: PMC10622387 DOI: 10.1186/s13568-023-01634-8] [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: 10/03/2023] [Accepted: 10/26/2023] [Indexed: 11/04/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) infection poses a severe threat to global public health due to its high mortality. Currently, polymyxins are mainly used for the treatment of Gram-negative bacterial-related infection, while exhibiting limited antibacterial activities against Staphylococcus aureus (S. aureus). However, the combination of antibiotics with antibiotic adjuvants is a feasible strategy for the hard-treated infection and toxicity reducing. We will investigate the antibacterial activity of simeprevir (SIM), which treated for genotype 1 and 4 chronic hepatitis C, combined with polymyxins against MRSA through high-throughput screening technology. In our study, the synergistic antibacterial effect of SIM and polymyxins against S. aureus in vitro was found by checkerboard assay and time-growth curve. The cytotoxicity of SIM combined with polymyxin B sulfate [PB(S)] or polymyxin E (PE) in vitro was evaluated using CCK-8, human RBC hemolysis and scratch assays. In addition, we investigated the eradication of biofilm formation of S. aureus by biofilm inhibition assay and the killing of persister cells. Moreover, we evaluated the therapeutic effect and in vivo toxicity of the combination against MRSA in murine subcutaneous abscess model. Furthermore, it was preliminarily found that SIM significantly enhanced the destruction of MRSA membrane by SYTOX Green and DISC3(5) probes. In summary, these results reveal that the therapy of SIM combined with polymyxins (especially PE) is promising for the treatment of MRSA infection.
Collapse
Affiliation(s)
- Yuan Wu
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Pingyun Wu
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Ruolan Wu
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Huilong Li
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Yao Duan
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Chaoni Cai
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Zixin Liu
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Pengfei She
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Di Zhang
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China.
| |
Collapse
|
16
|
Lv J, Wang S, Qi C, Li M, Sun Y, Yang Y, Zeng C, Shen R, Ma H. A fluorescent quaternary phosphonium main-chain-type polymer: an opportunity to fabricate functional materials with excellent antibacterial activity and bacterial imaging capability. J Mater Chem B 2023; 11:9237-9245. [PMID: 37702147 DOI: 10.1039/d3tb01240a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
The large-scale transmission and infection of pathogens worldwide have encouraged scientists to develop new antibacterial agents that do not succumb to bacterial resistance, which is not only of significant research interest but also challenging. In this work, we fabricated two main-chain (MC)-type cationic polymers (TPE-ammonium polymer and TPE-phosphonium polymer) through a one-step 100% atomic economic reaction. The two polymers demonstrated very promising antibacterial activity and their minimal inhibitory concentration (MIC) values are lower than that of most previously reported antibacterial agents. Especially, the phosphonium-doped MC polymer exhibited very small MICs of 0.24 and 0.98 μg mL-1 against S. aureus and E. coli, respectively. This excellent antibacterial performance by the TPE-phosphonium polymer is attributed to the advantages of the MC-type polymer such as its large molecular weight (Mn = 103 011) and stronger polarization effect from the P atom. More impressively, depending on the typical aggregation-induced emission (AIE) property and excellent antibacterial behaviors, the TPE-phosphonium polymer was successfully used for bacterial imaging and real-time monitoring of bacterial viability.
Collapse
Affiliation(s)
- Jiawei Lv
- Key Laboratory of Polymer Materials of Gansu Province, Key Laboratory of Eco-Environment-Related Polymer Materials Ministry of Education, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou 730070, P. R. China.
| | - Saicuo Wang
- China Agricultural Vet. Bio. Science and Technology Co., Ltd, Lanzhou 730046, P. R. China
| | - Chunxuan Qi
- AIE Research Centre, Shaanxi Key Laboratory of Phytochemistry, College of Chemistry and Chemical Engineering, Baoji University of Arts and Sciences, Baoji 721013, P. R. China
| | - Muheman Li
- Key Laboratory of Polymer Materials of Gansu Province, Key Laboratory of Eco-Environment-Related Polymer Materials Ministry of Education, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou 730070, P. R. China.
| | - Yuqing Sun
- Key Laboratory of Polymer Materials of Gansu Province, Key Laboratory of Eco-Environment-Related Polymer Materials Ministry of Education, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou 730070, P. R. China.
| | - Yuan Yang
- Key Laboratory of Polymer Materials of Gansu Province, Key Laboratory of Eco-Environment-Related Polymer Materials Ministry of Education, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou 730070, P. R. China.
| | - Cheng Zeng
- Key Laboratory of Polymer Materials of Gansu Province, Key Laboratory of Eco-Environment-Related Polymer Materials Ministry of Education, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou 730070, P. R. China.
| | - Richao Shen
- Key Laboratory of Polymer Materials of Gansu Province, Key Laboratory of Eco-Environment-Related Polymer Materials Ministry of Education, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou 730070, P. R. China.
| | - Hengchang Ma
- Key Laboratory of Polymer Materials of Gansu Province, Key Laboratory of Eco-Environment-Related Polymer Materials Ministry of Education, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou 730070, P. R. China.
| |
Collapse
|
17
|
Lembke HK, Espinasse A, Hanson MG, Grimme CJ, Tan Z, Reineke TM, Carlson EE. Cationic Polymers Enable Internalization of Negatively Charged Chemical Probes into Bacteria. ACS Chem Biol 2023; 18:2063-2072. [PMID: 37671702 PMCID: PMC10947785 DOI: 10.1021/acschembio.3c00351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
The bacterial cell envelope provides a protective barrier that is challenging for small molecules and biomolecules to cross. Given the anionic nature of both Gram-positive and Gram-negative bacterial cell envelopes, negatively charged molecules are particularly difficult to deliver into these organisms. Many strategies have been employed to penetrate bacteria, ranging from reagents such as cell-penetrating peptides, enzymes, and metal-chelating compounds to physical perturbations. While cationic polymers are known antimicrobial agents, polymers that promote the permeabilization of bacterial cells without causing high levels of toxicity and cell lysis have not yet been described. Here, we investigate four polymers that display a cationic poly(2-(dimethylamino)ethyl methacrylate (D) block for the internalization of an anionic adenosine triphosphate (ATP)-based chemical probe into Escherichia coli and Bacillus subtilis. We evaluated two polymer architectures, linear and micellar, to determine how shape and hydrophobicity affect internalization efficiency. We found that, in addition to these reagents successfully promoting probe internalization, the probe-labeled cells were able to continue to grow and divide. The micellar structures in particular were highly effective for the delivery of the negatively charged chemical probe. Finally, we demonstrated that these cationic polymers could act as general permeabilization reagents, promoting the entry of other molecules, such as antibiotics.
Collapse
Affiliation(s)
- Hannah K Lembke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Adeline Espinasse
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Christian J Grimme
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Zhe Tan
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Erin E Carlson
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
18
|
Silva JM, Teixeira AB, Reis AC. Silver-based gels for oral and skin infections: antimicrobial effect and physicochemical stability. Future Microbiol 2023; 18:985-996. [PMID: 37750752 DOI: 10.2217/fmb-2023-0034] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023] Open
Abstract
Aim: To systematically evaluate the literature on silver (Ag) gels and their antimicrobial efficacy and physicochemical stability. Materials & methods: A search was performed in PubMed/MEDLINE, LILACS, Web of Science, Scopus, Embase and Google Scholar. Results: Gels were formulated with Ag nanoparticles, Ag oxynitrate and colloidal Ag and showed antimicrobial activity for concentrations ranging from 0.002 to 30%. Gels showed stability of their chemical components, and their physicochemical properties, including viscosity, organoleptic characteristics, homogeneity, pH and spreadability, were suitable for topical application. Conclusion: Ag-based gels show antimicrobial action proportional to concentration, with higher action against Gram-negative bacteria and physicochemical stability for oral and skin infection applications.
Collapse
Affiliation(s)
- João Mc Silva
- Department of Dental Materials & Prosthesis, Ribeirão Preto Dental School, University of São Paulo, Avenida do Café s/n, Ribeirão Preto, São Paulo, 14040-904, Brazil
| | - Ana Bv Teixeira
- Department of Dental Materials & Prosthesis, Ribeirão Preto Dental School, University of São Paulo, Avenida do Café s/n, Ribeirão Preto, São Paulo, 14040-904, Brazil
| | - Andréa C Reis
- Department of Dental Materials & Prosthesis, Ribeirão Preto Dental School, University of São Paulo, Avenida do Café s/n, Ribeirão Preto, São Paulo, 14040-904, Brazil
| |
Collapse
|
19
|
Du J, Huang S, Wu M, Chen S, Zhou W, Zhan L, Huang X. Dlt operon regulates physiological function and cariogenic virulence in Streptococcus mutans. Future Microbiol 2023; 18:225-233. [PMID: 37097048 DOI: 10.2217/fmb-2022-0165] [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] [Indexed: 04/26/2023] Open
Abstract
Streptococcus mutans is one of the major cariogenic pathogens in the oral cavity. The dlt operon is responsible for the process of D-alanylation of lipoteichoic acid and is related to the virulence of S. mutans. The dlt operon contributes to the adhesion, biofilm formation, stress response, interspecies competitiveness and autolysis of S. mutans. In addition, we have summarized the possible regulatory networks of the dlt operon. This review highlights the significant role of the dlt operon in S. mutans and provides new ideas for ecological caries prevention.
Collapse
Affiliation(s)
- Jingyun Du
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key lab of Fujian College & University, School & Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Shan Huang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key lab of Fujian College & University, School & Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Minjing Wu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key lab of Fujian College & University, School & Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Shuai Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key lab of Fujian College & University, School & Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Wen Zhou
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key lab of Fujian College & University, School & Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Ling Zhan
- Division of Pediatric Dentistry, Department of Orofacial Sciences, Department of Preventive & Restorative Dental Sciences, University of California, San Francisco, CA, USA
| | - Xiaojing Huang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key lab of Fujian College & University, School & Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
20
|
Han X, Cui AL, Yang HX, Wu L, Wei R, Liu Q, Li ZR, Hu HY. Polymyxin-based fluorescent probes to combat Gram-negative antimicrobial resistance. Talanta 2023; 260:124576. [PMID: 37148689 DOI: 10.1016/j.talanta.2023.124576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 05/08/2023]
Abstract
Reliable diagnostic approaches especially those targeting critical Gram-negative bacteria are urgently needed for the prevention of antimicrobial resistance. Polymyxin B (PMB) which specifically targets the outer membrane of Gram-negative bacteria is the last-line antibiotic against life-threatening multidrug-resistant Gram-negative bacteria. However, increasing number of studies have reported the spread of PMB-resistant strains. With the aim to specifically detect Gram-negative bacteria and potentially reduce the irrational use of antibiotics, we herein rationally designed two Gram-negative bacteria specific fluorescent probes based on our previous activity-toxicity optimization of PMB. The in vitro probe PMS-Dns showed fast and selective labeling of Gram-negative pathogens in complex biological cultures. Subsequently, we constructed the caged in vivo fluorescent probe PMS-Cy-NO2 by conjugating bacterial nitroreductase (NTR)-activatable positive charged hydrophobic near-infrared (NIR) fluorophore with polymyxin scaffold. Significantly, PMS-Cy-NO2 exhibited excellent Gram-negative bacterial detection capability with the differentiation between Gram-positive and Gram-negative in a mouse skin infection model.
Collapse
Affiliation(s)
- Xiaowan Han
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - A-Long Cui
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - He-Xian Yang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Lingling Wu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Rao Wei
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Qian Liu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Zhuo-Rong Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Hai-Yu Hu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
21
|
Baran A, Kwiatkowska A, Potocki L. Antibiotics and Bacterial Resistance-A Short Story of an Endless Arms Race. Int J Mol Sci 2023; 24:ijms24065777. [PMID: 36982857 PMCID: PMC10056106 DOI: 10.3390/ijms24065777] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/10/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Despite the undisputed development of medicine, antibiotics still serve as first-choice drugs for patients with infectious disorders. The widespread use of antibiotics results from a wide spectrum of their actions encompassing mechanisms responsible for: the inhibition of bacterial cell wall biosynthesis, the disruption of cell membrane integrity, the suppression of nucleic acids and/or proteins synthesis, as well as disturbances of metabolic processes. However, the widespread availability of antibiotics, accompanied by their overprescription, acts as a double-edged sword, since the overuse and/or misuse of antibiotics leads to a growing number of multidrug-resistant microbes. This, in turn, has recently emerged as a global public health challenge facing both clinicians and their patients. In addition to intrinsic resistance, bacteria can acquire resistance to particular antimicrobial agents through the transfer of genetic material conferring resistance. Amongst the most common bacterial resistance strategies are: drug target site changes, increased cell wall permeability to antibiotics, antibiotic inactivation, and efflux pumps. A better understanding of the interplay between the mechanisms of antibiotic actions and bacterial defense strategies against particular antimicrobial agents is crucial for developing new drugs or drug combinations. Herein, we provide a brief overview of the current nanomedicine-based strategies that aim to improve the efficacy of antibiotics.
Collapse
Affiliation(s)
- Aleksandra Baran
- Department of Biotechnology, College of Natural Sciences, University of Rzeszów, Pigonia 1, 35-310 Rzeszow, Poland
| | - Aleksandra Kwiatkowska
- Institute of Physical Culture Studies, College of Medical Sciences, University of Rzeszów, ul. Towarnickiego 3, 35-959 Rzeszów, Poland
| | - Leszek Potocki
- Department of Biotechnology, College of Natural Sciences, University of Rzeszów, Pigonia 1, 35-310 Rzeszow, Poland
| |
Collapse
|
22
|
Saini M, Gaurav A, Kothari A, Omar BJ, Gupta V, Bhattacharjee A, Pathania R. Small Molecule IITR00693 (2-Aminoperimidine) Synergizes Polymyxin B Activity against Staphylococcus aureus and Pseudomonas aeruginosa. ACS Infect Dis 2023; 9:692-705. [PMID: 36716174 DOI: 10.1021/acsinfecdis.2c00622] [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: 01/31/2023]
Abstract
The rise of antibiotic resistance among skin-infecting pathogens poses an urgent threat to public health and has fueled the search for new therapies. Enhancing the potency of currently used antibiotics is an alternative for the treatment of infections caused by drug-resistant pathogens. In this study, we aimed to identify a small molecule that can potentiate currently used antibiotics. IITR00693 (2-aminoperimidine), a novel antibacterial small molecule, potentiates the antibacterial activity of polymyxin B against Staphylococcus aureus and Pseudomonas aeruginosa. Herein, we investigated in detail the mode of action of this interaction and the molecule's capability to combat soft-tissue infections caused by S. aureus and P. aeruginosa. A microdilution checkerboard assay was performed to determine the synergistic interaction between polymyxin B and IITR00693 in clinical isolates of S. aureus and P. aeruginosa. Time-kill kinetics, post-antibiotic effect, and resistance generation studies were performed to assess the pharmacodynamics of the combination. Assays based on different fluorescent probes were performed to decipher the mechanism of action of this combination. The in vivo efficacy of the IITR00693-polymyxin B combination was determined in a murine acute wound infection model. IITR00693 exhibited broad-spectrum antibacterial activity. IITR00693 potentiated polymyxin B and colistin against polymyxin-resistant S. aureus. IITR00693 prevented the generation of resistant mutants against multiple antibiotics. The IITR00693-polymyxin B combination decreased the S. aureus count by >3 log10 CFU in a murine acute wound infection model. IITR00693 is a potential and promising candidate for the treatment of soft-tissue infections along with polymyxins.
Collapse
Affiliation(s)
- Mahak Saini
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand247 667, India
| | - Amit Gaurav
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand247 667, India
| | - Ashish Kothari
- Department of Microbiology, All India Institute of Medical Sciences, Rishikesh, Uttarakhand249 201, India
| | - Balram Ji Omar
- Department of Microbiology, All India Institute of Medical Sciences, Rishikesh, Uttarakhand249 201, India
| | - Varsha Gupta
- Department of Microbiology, Government Medical College and Hospital Chandigarh, Chandigarh160 030, India
| | | | - Ranjana Pathania
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand247 667, India
| |
Collapse
|
23
|
Wei X, Gu Q, Feng Y, Zhang Y, Li Y, Zhang S, Zhang J, Wu S, Yang X, Ye Q, Ding Y, Wang J, Chen M, Wu Q. Sensitive and Selective Detection of Enterococcus faecalis Using a New Turn-on Fluorogenic β-glucosidase Substrate Combined with a Modified Selective Broth. Photochem Photobiol 2023; 99:68-77. [PMID: 35699359 DOI: 10.1111/php.13662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 06/02/2022] [Indexed: 01/25/2023]
Abstract
A new, simple-to-synthesize and sensitive turn-on fluorogenic substrate (CFMU-Glu) for β-glucosidase activity was developed. This probe was based on a 7-hydroxycoumarin derivative (CFMU) that could emit green fluorescence and had the low pKa value of 5.61 ± 0.01. CFMU-Glu could be used for sensitive monitoring of the almond βGLU and Enterococcus faecalis (E. faecalis) at the optimal pHs of 6.50 and 7.00, respectively. Moreover, a new sensitive and selective fluorogenic broth (PBF-B) for E. faecalis, utilizing CFMU-Glu and polymyxin B, was also developed. Polymyxin B was discovered to can significantly improve the detection selectivity and signal intensity. The proposed 4-four method using PBF-B and a microcentrifuge tube could provide fluorogenic detection limits of 5.01 × 104 and 1.0 × 105 CFU mL-1 by fluorescence microplate reader and naked eye, respectively; it could also provide a turn-on chromogenic detection limit of 1.0 × 106 CFU mL-1 by naked eye. The proposed method could detect 8 CFU mL-1 of E. faecalis in drinking water, Liangcha (herbal tea) and milk samples within 10 h, without pre-enrichment.
Collapse
Affiliation(s)
- Xianhu Wei
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Qihui Gu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Ying Feng
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Youxiong Zhang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Ying Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Shuhong Zhang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Jumei Zhang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Shi Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Xiaojuan Yang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Qinghua Ye
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Yu Ding
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Juan Wang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Moutong Chen
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| |
Collapse
|
24
|
Zhang H, Xu L, Gu X, Yu D, Li S. Amphiphilic di-cationic methylene blue for improving antibacterial photodynamic efficiency through high accumulation and low aggregation on bacterial cell surfaces. RSC Adv 2022; 13:239-250. [PMID: 36605628 PMCID: PMC9766197 DOI: 10.1039/d2ra06484g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
The aggregation state of photosensitizers on the surface of bacterial cells is an important scientific problem for antibacterial photodynamic therapy (APDT). High accumulation and high photoactive state maintenance of photosensitizers are the prerequisite of high APDT efficiency. In this study, an amphiphilic di-cationic methylene blue photosensitizer (C12-MB) was synthesized through quaternization, and its structure, interface properties, photophysical properties and antibacterial photodynamic properties were studied. The results showed that C12-MB could reduce 4.27 log10 CFU and 4.8 log10 CFU for P. aeruginosa and S. aureus under irradiation of light at 660 nm, higher than the parent methylene blue. Through a spectroscopic study on photosensitizer adsorption over the bacterial surface, C12-MB can be accumulated with higher concentration, and the photo-active monomer content is 73% and 70% over P. aeruginosa and S. aureus, higher than those of methylene blue: 25% and 49%, respectively. The higher content of non-aggregated photo-active monomer could contribute to higher antibacterial photodynamic efficiency. For C12-MB adsorbed over bacterial surfaces, planar packing inhibition and electrostatic repulsion could contribute to lower C12-MB aggregation, which provides an useful reference for the structural design of high-efficiency photosensitizers.
Collapse
Affiliation(s)
- Hao Zhang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University Nanjing 211816 P. R. China +86-25-58139386
| | - Lixian Xu
- Department of Dermatology, The Second Affiliated Hospital of Nanjing Medical University No. 121 Jiangjiayuan Road Nanjing 210000 P. R. China
| | - Xiaoxiao Gu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University Nanjing 211816 P. R. China +86-25-58139386
| | - Dinghua Yu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University Nanjing 211816 P. R. China +86-25-58139386
| | - Shuang Li
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University Nanjing 211816 P. R. China +86-25-58139386
| |
Collapse
|
25
|
Synthesis of an amphiphilic vancomycin aglycone derivative inspired by polymyxins: overcoming glycopeptide resistance in Gram-positive and Gram-negative bacteria in synergy with teicoplanin in vitro. Sci Rep 2022; 12:20921. [PMID: 36463278 PMCID: PMC9719540 DOI: 10.1038/s41598-022-24807-0] [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: 07/13/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022] Open
Abstract
Gram-negative bacteria possess intrinsic resistance to glycopeptide antibiotics so these important antibacterial medications are only suitable for the treatment of Gram-positive bacterial infections. At the same time, polymyxins are peptide antibiotics, structurally related to glycopeptides, with remarkable activity against Gram-negative bacteria. With the aim of breaking the intrinsic resistance of Gram-negative bacteria against glycopeptides, a polycationic vancomycin aglycone derivative carrying an n-decanoyl side chain and five aminoethyl groups, which resembles the structure of polymyxins, was prepared. Although the compound by itself was not active against the Gram-negative bacteria tested, it synergized with teicoplanin against Escherichia coli, Pseudomonas aeruginosa and Acinetobacter baumannii, and it was able to potentiate vancomycin against these Gram-negative strains. Moreover, it proved to be active against vancomycin- and teicoplanin-resistant Gram-positive bacteria.
Collapse
|
26
|
Qiu J, Li J, Du X, Zhou T, Xie B, He L. Synthesis and Characterization of Colistin-Functionalized Silica Materials for Rapid Capture of Bacteria in Water. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238292. [PMID: 36500384 PMCID: PMC9739998 DOI: 10.3390/molecules27238292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/10/2022] [Accepted: 11/17/2022] [Indexed: 11/29/2022]
Abstract
In this study, a new colistin-functionalized silica gel material (SiO2@NH2@COOH@CST) was synthesized after carboxylation on the surface of amino-modified silica. The main factors affecting the adsorptive properties of the material, such as the types of linkers, the linking methods, the reaction buffers and the particle sizes of carriers, were systematically investigated. The SiO2@NH2@COOH@CST was characterized by means of electron microscopy, Fourier-transform infrared spectroscopy, zeta potential measurements, etc. We demonstrated that the sorbent showed good adsorption of Gram-negative bacteria. The adsorption efficiency of E. coli on SiO2@NH2@COOH@CST was 5.2 × 1011 CFU/g, which was 3.5 times higher than that on SiO2@NH2@COOH, suggesting that electrostatic interactions between SiO2@NH2@COOH@CST and E. coli played a key role. The adsorption was quick, and was reached in 5 min. Both pseudo-first-order and pseudo-second-order kinetic models fit well with the dynamic adsorption process of SiO2@NH2@COOH@CST, indicating that physical adsorption and chemisorption might occur simultaneously during the adsorption process. SiO2@NH2@COOH@CST was successfully applied for the rapid capture of bacteria from water. The synthesized material could be used as a potential means of bacterial isolation and detection.
Collapse
Affiliation(s)
- Jingli Qiu
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- National Reference Laboratory of Veterinary Drug Residues (SCAU), College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Jianli Li
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Xiaoxi Du
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Tong Zhou
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
- National Reference Laboratory of Veterinary Drug Residues (SCAU), College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Bingbing Xie
- National Reference Laboratory of Veterinary Drug Residues (SCAU), College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Limin He
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- National Reference Laboratory of Veterinary Drug Residues (SCAU), College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Quality Supervision, Inspection and Testing Center for Domestic Animal Products Guangzhou, Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China
- Correspondence:
| |
Collapse
|
27
|
Wu YP, Liu DM, Liang MH, Huang YY, Lin J, Xiao LF. Genome-guided purification and characterization of polymyxin A1 from Paenibacillus thiaminolyticus SY20: A rarely explored member of polymyxins. Front Microbiol 2022; 13:962507. [DOI: 10.3389/fmicb.2022.962507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
Polymyxin A1 was a rarely investigated member in the polymyxins family produced by Bacillus aerosporus. As a cyclic non-ribosomal lipopeptide, it was purified from Paenibacillus thiaminolyticus for the first time. The producing strain SY20 was screened from Chinese natural fermented bamboo shoots and identified as P. thiaminolyticus SY20 using 16S rRNA homology along with whole genome sequencing. The optimum incubation time was 32 h by the growth kinetics of antimicrobial agent production. The proteinaceous nature of antimicrobial agents was characterized according to the physicochemical properties of the cell-free supernatant. Subsequently, the active antimicrobial agent was purified from the supernatant using ammonium sulfate–graded precipitation, ion-exchange chromatography, and C18-H chromatography. The active agent was identified as polymyxin A1 with a molecular weight 1156.7 Da and antimicrobial activity mainly against Gram-negative bacteria. The molecular structure, a cyclic heptapeptide and a tripeptide side chain acylated by a fatty acid at the amino terminus, was elucidated using the combination of liquid chromatography-tandem mass spectrometry (LC-MS/MS), matrix-assisted laser desorption ionization–time of flight mass spectrometry (MALDI-TOF MS), amino acid analysis, and whole genome mining tool. Meanwhile, the biosynthetic gene cluster of polymyxin A1 including five open reading frames (ORFs) was demonstrated in the genome. The compound should be further explored for its efficacy and toxicity in vivo to develop its application.
Collapse
|
28
|
Chiu S, Hancock AM, Schofner BW, Sniezek KJ, Soto-Echevarria N, Leon G, Sivaloganathan DM, Wan X, Brynildsen MP. Causes of polymyxin treatment failure and new derivatives to fill the gap. J Antibiot (Tokyo) 2022; 75:593-609. [PMID: 36123537 DOI: 10.1038/s41429-022-00561-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/08/2022]
Abstract
Polymyxins are a class of antibiotics that were discovered in 1947 from programs searching for compounds effective in the treatment of Gram-negative infections. Produced by the Gram-positive bacterium Paenibacillus polymyxa and composed of a cyclic peptide chain with a peptide-fatty acyl tail, polymyxins exert bactericidal effects through membrane disruption. Currently, polymyxin B and colistin (polymyxin E) have been developed for clinical use, where they are reserved as "last-line" therapies for multidrug-resistant (MDR) infections. Unfortunately, the incidences of strains resistant to polymyxins have been increasing globally, and polymyxin heteroresistance has been gaining appreciation as an important clinical challenge. These phenomena, along with bacterial tolerance to this antibiotic class, constitute important contributors to polymyxin treatment failure. Here, we review polymyxins and their mechanism of action, summarize the current understanding of how polymyxin treatment fails, and discuss how the next generation of polymyxins holds promise to invigorate this antibiotic class.
Collapse
Affiliation(s)
- Selena Chiu
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | - Anna M Hancock
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | - Bob W Schofner
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Katherine J Sniezek
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | | | - Gabrielle Leon
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | | | - Xuanqing Wan
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | - Mark P Brynildsen
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA.
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
29
|
Xiao X, Ji J, Wang H, Nangia S, Wang H, Libera M. Self-Defensive Antimicrobial Surfaces Using Polymyxin-Loaded Poly(styrene sulfonate) Microgels. ACS Biomater Sci Eng 2022; 8:4827-4837. [PMID: 36256955 DOI: 10.1021/acsbiomaterials.2c00783] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Self-defensive antimicrobial surfaces are of interest because they can inhibit bacterial colonization while minimizing unnecessary antimicrobial release in the absence of a bacterial challenge. One self-defensive approach uses self-assembly to first deposit a submonolayer coating of polyelectrolyte microgels and subsequently load those microgels by complexation with small-molecule antimicrobials. The microgel/antimicrobial complexation strength is a key parameter that controls the ability of the antimicrobial both to remain sequestered within the microgels when exposed to medium and to release in response to a bacterial challenge. Here we study the relative complexation strengths of two FDA-approved cationic antibiotics─colistin (polymyxin E) and polymyxin B─with microgels of poly(styrene sulfonate) (PSS). These polymyxins are similar cyclic polypeptides with +5 charge at pH 7.4. However, polymyxin B substitutes an aromatic ring for a dimethyl moiety in colistin, and this aromaticity can influence complexation via π and hydrophobic interactions. Coarse-grained molecular dynamics shows that the free-energy change associated with polymyxin B/PSS complexation is more negative than that of colistin/PSS complexation. Experimentally, in situ optical microscopy of microgel deswelling shows that both antibiotics load quickly from low-ionic-strength phosphate buffer. The enhanced polymyxin B/PSS complexation strength is then manifested by subsequent exposure to flowing antibiotic-free buffer with varying NaCl concentration. Microgels loaded with polymyxin B remain stably deswollen to higher salt concentrations than do colistin/PSS microgels. Importantly, exposing loaded microgels to E. coli in nutrient-free-flowing phosphate buffer shows that bacteria are killed by physical contact with the loaded microgels consistent with the contact-transfer mechanism of self-defensiveness. In vitro culture experiments show that these same surfaces, nevertheless, support the adhesion, spreading and proliferation of human fetal osteoblasts. These findings suggest a pathway to create a self-defensive antimicrobial surface effective under physiological conditions based on the nonmetabolic bacteria-triggered release of FDA-approved antibiotics.
Collapse
Affiliation(s)
- Xixi Xiao
- Department of Chemical Engineering and Materials Science, Stevens Institute of Technology, Hoboken, New Jersey07030, United States
| | - Jingjing Ji
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York13244, United States
| | - Haoyu Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey07030, United States
| | - Shikha Nangia
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York13244, United States
| | - Hongjun Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey07030, United States.,Center for Healthcare Innovation, Stevens Institute of Technology, Hoboken, New Jersey07030, United States
| | - Matthew Libera
- Department of Chemical Engineering and Materials Science, Stevens Institute of Technology, Hoboken, New Jersey07030, United States
| |
Collapse
|
30
|
Hydrophobic modification Thr of polymyxin E: Effect on activity and toxicity. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
31
|
Lee DH, Cha JH, Kim DW, Lee K, Kim YS, Oh HY, Cho YH, Cha CJ. Colistin-degrading proteases confer collective resistance to microbial communities during polymicrobial infections. MICROBIOME 2022; 10:129. [PMID: 35982474 PMCID: PMC9389796 DOI: 10.1186/s40168-022-01315-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/19/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND The increasing prevalence of resistance against the last-resort antibiotic colistin is a significant threat to global public health. Here, we discovered a novel colistin resistance mechanism via enzymatic inactivation of the drug and proposed its clinical importance in microbial communities during polymicrobial infections. RESULTS A bacterial strain of the Gram-negative opportunistic pathogen Stenotrophomonas maltophilia capable of degrading colistin and exhibiting a high-level colistin resistance was isolated from the soil environment. A colistin-degrading protease (Cdp) was identified in this strain, and its contribution to colistin resistance was demonstrated by growth inhibition experiments using knock-out (Δcdp) and complemented (Δcdp::cdp) mutants. Coculture and coinfection experiments revealed that S. maltophilia carrying the cdp gene could inactivate colistin and protect otherwise susceptible Pseudomonas aeruginosa, which may seriously affect the clinical efficacy of the drug for the treatment of cystic fibrosis patients with polymicrobial infection. CONCLUSIONS Our results suggest that Cdp should be recognized as a colistin resistance determinant that confers collective resistance at the microbial community level. Our study will provide vital information for successful clinical outcomes during the treatment of complex polymicrobial infections, particularly including S. maltophilia and other colistin-susceptible Gram-negative pathogens such as P. aeruginosa. Video abstract.
Collapse
Affiliation(s)
- Do-Hoon Lee
- Department of Systems Biotechnology and Center for Antibiotic Resistome, Chung-Ang University, Anseong, 17456, Republic of Korea
| | - Ju-Hee Cha
- Department of Systems Biotechnology and Center for Antibiotic Resistome, Chung-Ang University, Anseong, 17456, Republic of Korea
| | - Dae-Wi Kim
- Department of Systems Biotechnology and Center for Antibiotic Resistome, Chung-Ang University, Anseong, 17456, Republic of Korea
- Division of Life Sciences, Jeonbuk National University, Jeonju, 54896, Republic of Korea
| | - Kihyun Lee
- Department of Systems Biotechnology and Center for Antibiotic Resistome, Chung-Ang University, Anseong, 17456, Republic of Korea
| | - Yong-Seok Kim
- Department of Systems Biotechnology and Center for Antibiotic Resistome, Chung-Ang University, Anseong, 17456, Republic of Korea
| | - Hyo-Young Oh
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, 13488, Republic of Korea
| | - You-Hee Cho
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, 13488, Republic of Korea
| | - Chang-Jun Cha
- Department of Systems Biotechnology and Center for Antibiotic Resistome, Chung-Ang University, Anseong, 17456, Republic of Korea.
| |
Collapse
|
32
|
Study on the adsorption and dust suppression mechanism of urease-producing bacteria on coal dust. POWDER TECHNOL 2022. [DOI: 10.1016/j.powtec.2022.117801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
33
|
Hale SJM, Wagner Mackenzie B, Lux CA, Biswas K, Kim R, Douglas RG. Topical Antibiofilm Agents With Potential Utility in the Treatment of Chronic Rhinosinusitis: A Narrative Review. Front Pharmacol 2022; 13:840323. [PMID: 35770097 PMCID: PMC9234399 DOI: 10.3389/fphar.2022.840323] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
The role of bacterial biofilms in chronic and recalcitrant diseases is widely appreciated, and the treatment of biofilm infection is an increasingly important area of research. Chronic rhinosinusitis (CRS) is a complex disease associated with sinonasal dysbiosis and the presence of bacterial biofilms. While most biofilm-related diseases are associated with highly persistent but relatively less severe inflammation, the presence of biofilms in CRS is associated with greater severity of inflammation and recalcitrance despite appropriate treatment. Oral antibiotics are commonly used to treat CRS but they are often ineffective, due to poor penetration of the sinonasal mucosa and the inherently antibiotic resistant nature of bacteria in biofilms. Topical non-antibiotic antibiofilm agents may prove more effective, but few such agents are available for sinonasal application. We review compounds with antibiofilm activity that may be useful for treating biofilm-associated CRS, including halogen-based compounds, quaternary ammonium compounds and derivatives, biguanides, antimicrobial peptides, chelating agents and natural products. These include preparations that are currently available and those still in development. For each compound, antibiofilm efficacy, mechanism of action, and toxicity as it relates to sinonasal application are summarised. We highlight the antibiofilm agents that we believe hold the greatest promise for the treatment of biofilm-associated CRS in order to inform future research on the management of this difficult condition.
Collapse
Affiliation(s)
- Samuel J M Hale
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Brett Wagner Mackenzie
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Christian A Lux
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kristi Biswas
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Raymond Kim
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard G Douglas
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
34
|
Wang Y, Chen G, Liu R, Fang X, Li F, Wu L, Wu Y. Synergistically enhanced photothermal transition of a polyoxometalate/peptide assembly improved the antibiofilm and antibacterial activities. SOFT MATTER 2022; 18:2951-2958. [PMID: 35348178 DOI: 10.1039/d2sm00092j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
We successfully developed an antimicrobial assembly (Mo154/TK-14) using molybdenum-polyoxometalate and a positively charged peptide of TK-14. It was characterized and assayed using zeta-potential, dynamic light scattering (DLS), and TEM measurements. The Mo154/TK-14 assembly showed an enhanced 808 nm absorption and, therefore, improved the photothermal conversion efficiency of Mo154 (30.3%) to 38.6%. Consequently, in comparison to 5 μM Mo154 without irradiation, both the biofilm formation and bacterial viability of S. aureus were 24.6% and 20.2%, respectively, for the Mo154/TK-14 assembly; the biofilm formation and bacterial viability were further decreased to 7.7% and 4.4% under 808 nm irradiation, respectively. Therefore, the Mo154/TK-14 assembly reflects convincing antibacterial properties compared to Mo154. This is due to the synergistic effect between the peptide-binding enhanced 808 nm absorption and the improved PTT properties. The antimicrobial assembly offers a novel strategy for the rational design of light-responsive antibacterial materials.
Collapse
Affiliation(s)
- Yu Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, No. 2699 Qianjin Street, Changchun 130012, P. R. China.
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, No. 2 Liutiao Road, Changchun 130023, P. R. China
| | - Gang Chen
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, No. 2699 Qianjin Street, Changchun 130012, P. R. China.
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, No. 2 Liutiao Road, Changchun 130023, P. R. China
| | - Rongrong Liu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, College of Life Science, Jilin University, No. 2699 Qianjin Street, Changchun 130012, P. R. China
| | - Xuexun Fang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, College of Life Science, Jilin University, No. 2699 Qianjin Street, Changchun 130012, P. R. China
| | - Fei Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, No. 2699 Qianjin Street, Changchun 130012, P. R. China.
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, No. 2 Liutiao Road, Changchun 130023, P. R. China
| | - Lixin Wu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, No. 2699 Qianjin Street, Changchun 130012, P. R. China.
| | - Yuqing Wu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, No. 2699 Qianjin Street, Changchun 130012, P. R. China.
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, No. 2 Liutiao Road, Changchun 130023, P. R. China
| |
Collapse
|
35
|
The Microstructure, Antibacterial and Antitumor Activities of Chitosan Oligosaccharides and Derivatives. Mar Drugs 2022; 20:md20010069. [PMID: 35049924 PMCID: PMC8781119 DOI: 10.3390/md20010069] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/06/2022] [Accepted: 01/10/2022] [Indexed: 02/07/2023] Open
Abstract
Chitosan obtained from abundant marine resources has been proven to have a variety of biological activities. However, due to its poor water solubility, chitosan application is limited, and the degradation products of chitosan oligosaccharides are better than chitosan regarding performance. Chitosan oligosaccharides have two kinds of active groups, amino and hydroxyl groups, which can form a variety of derivatives, and the properties of these derivatives can be further improved. In this review, the key structures of chitosan oligosaccharides and recent studies on chitosan oligosaccharide derivatives, including their synthesis methods, are described. Finally, the antimicrobial and antitumor applications of chitosan oligosaccharides and their derivatives are discussed.
Collapse
|
36
|
Yin J, Cheng D, Zhu Y, Liang Y, Yu Z. Development of a whole-cell biosensor for detection of antibiotics targeting bacterial cell envelope in Bacillus subtilis. Appl Microbiol Biotechnol 2022; 106:789-798. [DOI: 10.1007/s00253-022-11762-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 12/15/2021] [Accepted: 01/05/2022] [Indexed: 12/29/2022]
|
37
|
Vimberg V, Buriánková K, Mazumdar A, Branny P, Novotná GB. Role of membrane proteins in bacterial resistance to antimicrobial peptides. Med Res Rev 2021; 42:1023-1036. [PMID: 34796517 DOI: 10.1002/med.21869] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 09/19/2021] [Accepted: 10/21/2021] [Indexed: 11/07/2022]
Abstract
Several natural antimicrobial peptides (AMPs), including the novel semisynthetic lipoglycopeptide antibiotics telavancin, dalbavancin, and oritavancin, have been approved for clinical use to address the growing problem of multiple antibiotic-resistant Gram-positive bacterial infections. Nevertheless, the efficacy of these antibiotics has already been compromised. The SARS-CoV-2 pandemic led to the increased clinical use of all antibiotics, further promoting the development of bacterial resistance. Therefore, it is critical to gain a deeper understanding of the role of resistance mechanisms to minimize the consequential risks of long-term antibiotic use and misuse. Here, we summarize for the first time the current knowledge of resistance mechanisms that have been shown to cause resistance to clinically used AMPs, with particular focus on membrane proteins that have been reported to interfere with the activity of AMPs by affecting the binding of AMPs to bacteria.
Collapse
Affiliation(s)
- Vladimir Vimberg
- Laboratory for Biology of Secondary Metabolism, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Karolína Buriánková
- Laboratory of Cell Signaling, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Aninda Mazumdar
- Laboratory for Biology of Secondary Metabolism, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Pavel Branny
- Laboratory of Cell Signaling, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Gabriela B Novotná
- Laboratory for Biology of Secondary Metabolism, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
38
|
Mohapatra SS, Dwibedy SK, Padhy I. Polymyxins, the last-resort antibiotics: Mode of action, resistance emergence, and potential solutions. J Biosci 2021. [PMID: 34475315 PMCID: PMC8387214 DOI: 10.1007/s12038-021-00209-8] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Infections caused by multi-drug resistant (MDR) bacterial pathogens are a leading cause of mortality and morbidity across the world. Indiscriminate use of broad-spectrum antibiotics has seriously affected this situation. With the diminishing discovery of novel antibiotics, new treatment methods are urgently required to combat MDR pathogens. Polymyxins, the cationic lipopeptide antibiotics, discovered more than half a century ago, are considered to be the last-line of antibiotics available at the moment. This antibiotic shows a great bactericidal effect against Gram-negative bacteria. Polymyxins primarily target the bacterial membrane and disrupt them, causing lethality. Because of their membrane interacting mode of action, polymyxins cause nephrotoxicity and neurotoxicity in humans, limiting their usability. However, recent modifications in their chemical structure have been able to reduce the toxic effects. The development of better dosing regimens has also helped in getting better clinical outcomes in the infections caused by MDR pathogens. Since the mid-1990s the use of polymyxins has increased manifold in clinical settings, resulting in the emergence of polymyxin-resistant strains. The risk posed by the polymyxin-resistant nosocomial pathogens such as the Enterobacteriaceae group, Pseudomonas aeruginosa, and Acinetobacter baumannii, etc. is very serious considering these pathogens are resistant to almost all available antibacterial drugs. In this review article, the mode of action of the polymyxins and the genetic regulatory mechanism responsible for the emergence of resistance are discussed. Specifically, this review aims to update our current understanding in the field and suggest possible solutions that can be pursued for future antibiotic development. As polymyxins primarily target the bacterial membranes, resistance to polymyxins arises primarily by the modification of the lipopolysaccharides (LPS) in the outer membrane (OM). The LPS modification pathways are largely regulated by the bacterial two-component signal transduction (TCS) systems. Therefore, targeting or modulating the TCS signalling mechanisms can be pursued as an alternative to treat the infections caused by polymyxin-resistant MDR pathogens. In this review article, this aspect is also highlighted.
Collapse
Affiliation(s)
- Saswat S Mohapatra
- Molecular Microbiology Lab, Department of Bioscience and Bioinformatics, Khallikote University, Konisi, Berhampur, 761 008 Odisha India
| | - Sambit K Dwibedy
- Molecular Microbiology Lab, Department of Bioscience and Bioinformatics, Khallikote University, Konisi, Berhampur, 761 008 Odisha India
| | - Indira Padhy
- Molecular Microbiology Lab, Department of Bioscience and Bioinformatics, Khallikote University, Konisi, Berhampur, 761 008 Odisha India
| |
Collapse
|
39
|
Khatun M, Damgaard BM, Andersen JB, Røntved CM. Effect of polymyxin B on ex vivo tumor necrosis factor-alpha responsiveness of blood leukocytes in Danish Holstein Friesian cows. Vet Immunol Immunopathol 2021; 238:110293. [PMID: 34284224 DOI: 10.1016/j.vetimm.2021.110293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/14/2021] [Accepted: 07/14/2021] [Indexed: 10/20/2022]
Abstract
Whole blood stimulation assay (WBA) with killed gram-positive and gram-negative udder pathogens were used to investigate the interference of the endotoxin-binding antibiotic polymyxin B (PMB) on the ex vivo TNF-α response. Blood samples were collected from first to third lactating dairy cows in their early lactation (<50 days in milk, n = 32) period. The WBA was stimulated with both inactivated bacteria (e.g., dead Escherichia coli, Staphylococcus aureus, Streptococcus dysgalactiae, Streptococcus uberis), at a concentration of 2.5 × 106/mL; and pathogen-associated molecular pattern molecules, namely E. coli LPS (10 μg/mL), and S. aureus peptidoglycan (PG, 10 μg/mL). The PMB was added at a concentration of 0, 12.5, 25, 50, 100, and 200 μg/mL to each stimulant, respectively. All bacteria stimulants resulted in an increased TNF-α response compared to the negative control. The PMB affected the TNF-α responses of gram-positive (except S. dysgalactaie), gram-negative bacteria; and bacterial cell wall components at a PMB concentration of 25-50 μg/mL. The LPS and E. coli had similar TNF-α response but PG had a lower TNF-α response than gram-positive bacteria. The doses of PMB (≥ 25 μg/mL) should be used with caution when using different types of pathogens or should be avoided in ex vivo TNF-α studies.
Collapse
Affiliation(s)
- Momena Khatun
- Aarhus University, Faculty of Science and Technology, Department of Animal Science, Blichers Allé 20, P.O. Box 50, DK-8300, Tjele, Denmark.
| | - Birthe M Damgaard
- Aarhus University, Faculty of Science and Technology, Department of Animal Science, Blichers Allé 20, P.O. Box 50, DK-8300, Tjele, Denmark.
| | - Jens B Andersen
- Aarhus University, Faculty of Science and Technology, Department of Animal Science, Blichers Allé 20, P.O. Box 50, DK-8300, Tjele, Denmark.
| | - Christine M Røntved
- Aarhus University, Faculty of Science and Technology, Department of Animal Science, Blichers Allé 20, P.O. Box 50, DK-8300, Tjele, Denmark; Christine Røntved, CMR On-Site RD, Graverhusvej 53, Langholt, 9310, Vodskov, Denmark.
| |
Collapse
|
40
|
Panta PR, Doerrler WT. A link between pH homeostasis and colistin resistance in bacteria. Sci Rep 2021; 11:13230. [PMID: 34168215 PMCID: PMC8225787 DOI: 10.1038/s41598-021-92718-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/14/2021] [Indexed: 12/15/2022] Open
Abstract
Colistin resistance is complex and multifactorial. DbcA is an inner membrane protein belonging to the DedA superfamily required for maintaining extreme colistin resistance of Burkholderia thailandensis. The molecular mechanisms behind this remain unclear. Here, we report that ∆dbcA displays alkaline pH/bicarbonate sensitivity and propose a role of DbcA in extreme colistin resistance of B. thailandensis by maintaining cytoplasmic pH homeostasis. We found that alkaline pH or presence of sodium bicarbonate displays a synergistic effect with colistin against not only extremely colistin resistant species like B. thailandensis and Serratia marcescens, but also a majority of Gram-negative and Gram-positive bacteria tested, suggesting a link between cytoplasmic pH homeostasis and colistin resistance across species. We found that lowering the level of oxygen in the growth media or supplementation of fermentable sugars such as glucose not only alleviated alkaline pH stress, but also increased colistin resistance in most bacteria tested, likely by avoiding cytoplasmic alkalinization. Our observations suggest a previously unreported link between pH, oxygen, and colistin resistance. We propose that maintaining optimal cytoplasmic pH is required for colistin resistance in a majority of bacterial species, consistent with the emerging link between cytoplasmic pH homeostasis and antibiotic resistance.
Collapse
Affiliation(s)
- Pradip R Panta
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - William T Doerrler
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
41
|
Rónavári A, Igaz N, Adamecz DI, Szerencsés B, Molnar C, Kónya Z, Pfeiffer I, Kiricsi M. Green Silver and Gold Nanoparticles: Biological Synthesis Approaches and Potentials for Biomedical Applications. Molecules 2021; 26:844. [PMID: 33562781 PMCID: PMC7915205 DOI: 10.3390/molecules26040844] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/30/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023] Open
Abstract
The nanomaterial industry generates gigantic quantities of metal-based nanomaterials for various technological and biomedical applications; however, concomitantly, it places a massive burden on the environment by utilizing toxic chemicals for the production process and leaving hazardous waste materials behind. Moreover, the employed, often unpleasant chemicals can affect the biocompatibility of the generated particles and severely restrict their application possibilities. On these grounds, green synthetic approaches have emerged, offering eco-friendly, sustainable, nature-derived alternative production methods, thus attenuating the ecological footprint of the nanomaterial industry. In the last decade, a plethora of biological materials has been tested to probe their suitability for nanomaterial synthesis. Although most of these approaches were successful, a large body of evidence indicates that the green material or entity used for the production would substantially define the physical and chemical properties and as a consequence, the biological activities of the obtained nanomaterials. The present review provides a comprehensive collection of the most recent green methodologies, surveys the major nanoparticle characterization techniques and screens the effects triggered by the obtained nanomaterials in various living systems to give an impression on the biomedical potential of green synthesized silver and gold nanoparticles.
Collapse
Affiliation(s)
- Andrea Rónavári
- Department of Applied and Environmental Chemistry, University of Szeged, Rerrich Béla tér 1., H-6720 Szeged, Hungary; (A.R.); (Z.K.)
| | - Nóra Igaz
- Department of Biochemistry and Molecular Biology and Doctoral School of Biology, University of Szeged, Közép fasor 52., H-6726 Szeged, Hungary; (N.I.); (D.I.A.)
| | - Dóra I. Adamecz
- Department of Biochemistry and Molecular Biology and Doctoral School of Biology, University of Szeged, Közép fasor 52., H-6726 Szeged, Hungary; (N.I.); (D.I.A.)
| | - Bettina Szerencsés
- Department of Microbiology and Doctoral School of Biology, University of Szeged, Közép fasor 52., H-6726 Szeged, Hungary; (B.S.); (I.P.)
| | - Csaba Molnar
- Broad Institute of MIT and Harvard, Cambridge, 415 Main St, Cambridge, MA 02142, USA;
| | - Zoltán Kónya
- Department of Applied and Environmental Chemistry, University of Szeged, Rerrich Béla tér 1., H-6720 Szeged, Hungary; (A.R.); (Z.K.)
- MTA-SZTE Reaction Kinetics and Surface Chemistry Research Group, Rerrich Béla tér 1., H-6720 Szeged, Hungary
| | - Ilona Pfeiffer
- Department of Microbiology and Doctoral School of Biology, University of Szeged, Közép fasor 52., H-6726 Szeged, Hungary; (B.S.); (I.P.)
| | - Monika Kiricsi
- Department of Biochemistry and Molecular Biology and Doctoral School of Biology, University of Szeged, Közép fasor 52., H-6726 Szeged, Hungary; (N.I.); (D.I.A.)
| |
Collapse
|
42
|
Mohapatra SS, Dwibedy SK, Padhy I. Polymyxins, the last-resort antibiotics: Mode of action, resistance emergence, and potential solutions. J Biosci 2021; 46:85. [PMID: 34475315 PMCID: PMC8387214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/03/2021] [Indexed: 04/04/2024]
Abstract
Infections caused by multi-drug resistant (MDR) bacterial pathogens are a leading cause of mortality and morbidity across the world. Indiscriminate use of broad-spectrum antibiotics has seriously affected this situation. With the diminishing discovery of novel antibiotics, new treatment methods are urgently required to combat MDR pathogens. Polymyxins, the cationic lipopeptide antibiotics, discovered more than half a century ago, are considered to be the last-line of antibiotics available at the moment. This antibiotic shows a great bactericidal effect against Gram-negative bacteria. Polymyxins primarily target the bacterial membrane and disrupt them, causing lethality. Because of their membrane interacting mode of action, polymyxins cause nephrotoxicity and neurotoxicity in humans, limiting their usability. However, recent modifications in their chemical structure have been able to reduce the toxic effects. The development of better dosing regimens has also helped in getting better clinical outcomes in the infections caused by MDR pathogens. Since the mid1990s the use of polymyxins has increased manifold in clinical settings, resulting in the emergence of polymyxin-resistant strains. The risk posed by the polymyxin-resistant nosocomial pathogens such as the Enterobacteriaceae group, Pseudomonas aeruginosa, and Acinetobacter baumannii, etc. is very serious considering these pathogens are resistant to almost all available antibacterial drugs. In this review article, the mode of action of the polymyxins and the genetic regulatory mechanism responsible for the emergence of resistance are discussed. Specifically, this review aims to update our current understanding in the field and suggest possible solutions that can be pursued for future antibiotic development. As polymyxins primarily target the bacterial membranes, resistance to polymyxins arises primarily by the modification of the lipopolysaccharides (LPS) in the outer membrane (OM). The LPS modification pathways are largely regulated by the bacterial two-component signal transduction (TCS) systems. Therefore, targeting or modulating the TCS signalling mechanisms can be pursued as an alternative to treat the infections caused by polymyxin-resistant MDR pathogens. In this review article, this aspect is also highlighted.
Collapse
Affiliation(s)
- Saswat S Mohapatra
- Molecular Microbiology Lab, Department of Bioscience and Bioinformatics, Khallikote University, Konisi, Berhampur, 761 008 Odisha India
| | - Sambit K Dwibedy
- Molecular Microbiology Lab, Department of Bioscience and Bioinformatics, Khallikote University, Konisi, Berhampur, 761 008 Odisha India
| | - Indira Padhy
- Molecular Microbiology Lab, Department of Bioscience and Bioinformatics, Khallikote University, Konisi, Berhampur, 761 008 Odisha India
| |
Collapse
|
43
|
Sisay M, Hagos B, Edessa D, Tadiwos Y, Mekuria AN. Polymyxin-induced nephrotoxicity and its predictors: a systematic review and meta-analysis of studies conducted using RIFLE criteria of acute kidney injury. Pharmacol Res 2020; 163:105328. [PMID: 33276108 DOI: 10.1016/j.phrs.2020.105328] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/18/2020] [Accepted: 11/23/2020] [Indexed: 01/16/2023]
Abstract
Polymyxins are last-resort antibiotics re-emerged to treat infections caused by multidrug resistant (MDR) and extensively drug-resistant (XDR) Gram-negative bacterial infections. However, polymyxin-associated nephrotoxicity has become the main safety concern. Therefore, we conducted this systematic review and meta-analysis on polymyxin-induced nephrotoxicity and its predictors using studies conducted based on the validated RIFLE (Risk, Injury, Failure, Loss of Function and End-stage renal disease) criteria of acute kidney damage. Literature search was carried out through visiting legitimate databases and indexing services including PubMed, MEDLINE (Ovid®), EMBASE (Ovid®), and Scopus to retrieve relevant studies. Following screening and eligibility evaluation, relevant data were extracted from included studies and analyzed using STATA 15.0 and Rev-Man 5.3. Inverse variance method with random effects pooling model was used for the analysis of outcome measures at 95% confidence interval. Besides, meta-regression, meta-influence, and publication bias analyses were conducted. A total of 48 studies involving 6,199 adult patients aged ≥ 18 years were included for systematic review and meta-analysis. The pooled incidence of polymyxin-induced nephrotoxicity was found to be 45% (95% CI: 41- 49%; I2 = 92.52%). Stratifying with RIFLE severity scales, pooled estimates of polymyxin-treated patients identified as 'risk', 'injury' and 'failure' were 17% (95% CI: 14-20%), 13% (95% CI: 11-15%), and 10% (95% CI: 9-11%), respectively. Besides, the pooled incidence of colistin-induced nephrotoxicity was about 48% (95% CI: 42-54%), whereas that of polymyxin B was 38% (95% CI: 32-44%). Likewise, colistin had 37% increased risk of developing nephrotoxicity compared to the polymyxin B treated cohorts (RR = 1.37, 95% CI: 1.13-1.67; I2 = 57%). Older age (AOR = 1.03, 95% CI: 1.01-1.05), daily dose (AOR = 1.46, 95% CI: 1.09-1.96), underlying diabetes mellitus (AOR = 1.81, 95% CI: 1.25-2.63), and concomitant nephrotoxic drugs (AOR = 2.31, 95% CI: 1.79-3.00) were independent risk factors for polymyxin-induced nephrotoxicity. Patients with high serum albumin level were less likely (AOR = 0.69, 95% CI: 0.56-0.85] to experience nephrotoxicity compared to those with low albumin level. Despite the resurgence of these antibiotics for the chemotherapy of MDR/XDR-Gram-negative superbugs, the high incidence of nephrotoxicity has become a contemporary clinical concern. Being elderly, high daily dose, having underlying diseases such as diabetes, and use of concomitant nephrotoxic drugs were independent predictors of nephrotoxicity. Therefore, therapeutic drug monitoring should be done to these patients to outweigh the potential benefits of polymyxin therapy from its risk.
Collapse
Affiliation(s)
- Mekonnen Sisay
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia.
| | - Bisrat Hagos
- Department of Social Pharmacy, School of Pharmacy, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia.
| | - Dumessa Edessa
- Department of Clinical Pharmacy, School of Pharmacy, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia.
| | - Yohannes Tadiwos
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia.
| | - Abraham Nigussie Mekuria
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia.
| |
Collapse
|
44
|
Abstract
Invasive fungal infections in humans are generally associated with high mortality, making the choice of antifungal drug crucial for the outcome of the patient. The limited spectrum of antifungals available and the development of drug resistance represent the main concerns for the current antifungal treatments, requiring alternative strategies. Antimicrobial peptides (AMPs), expressed in several organisms and used as first-line defenses against microbial infections, have emerged as potential candidates for developing new antifungal therapies, characterized by negligible host toxicity and low resistance rates. Most of the current literature focuses on peptides with antibacterial activity, but there are fewer studies of their antifungal properties. This review focuses on AMPs with antifungal effects, including their in vitro and in vivo activities, with the biological repercussions on the fungal cells, when known. The classification of the peptides is based on their mode of action: although the majority of AMPs exert their activity through the interaction with membranes, other mechanisms have been identified, including cell wall inhibition and nucleic acid binding. In addition, antifungal compounds with unknown modes of action are also described. The elucidation of such mechanisms can be useful to identify novel drug targets and, possibly, to serve as the templates for the synthesis of new antimicrobial compounds with increased activity and reduced host toxicity.
Collapse
|
45
|
Dezanet C, Kempf J, Mingeot-Leclercq MP, Décout JL. Amphiphilic Aminoglycosides as Medicinal Agents. Int J Mol Sci 2020; 21:E7411. [PMID: 33049963 PMCID: PMC7583001 DOI: 10.3390/ijms21197411] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 09/27/2020] [Accepted: 10/02/2020] [Indexed: 12/25/2022] Open
Abstract
The conjugation of hydrophobic group(s) to the polycationic hydrophilic core of the antibiotic drugs aminoglycosides (AGs), targeting ribosomal RNA, has led to the development of amphiphilic aminoglycosides (AAGs). These drugs exhibit numerous biological effects, including good antibacterial effects against susceptible and multidrug-resistant bacteria due to the targeting of bacterial membranes. In the first part of this review, we summarize our work in identifying and developing broad-spectrum antibacterial AAGs that constitute a new class of antibiotic agents acting on bacterial membranes. The target-shift strongly improves antibiotic activity against bacterial strains that are resistant to the parent AG drugs and to antibiotic drugs of other classes, and renders the emergence of resistant Pseudomonas aeruginosa strains highly difficult. Structure-activity and structure-eukaryotic cytotoxicity relationships, specificity and barriers that need to be crossed in their development as antibacterial agents are delineated, with a focus on their targets in membranes, lipopolysaccharides (LPS) and cardiolipin (CL), and the corresponding mode of action against Gram-negative bacteria. At the end of the first part, we summarize the other recent advances in the field of antibacterial AAGs, mainly published since 2016, with an emphasis on the emerging AAGs which are made of an AG core conjugated to an adjuvant or an antibiotic drug of another class (antibiotic hybrids). In the second part, we briefly illustrate other biological and biochemical effects of AAGs, i.e., their antifungal activity, their use as delivery vehicles of nucleic acids, of short peptide (polyamide) nucleic acids (PNAs) and of drugs, as well as their ability to cleave DNA at abasic sites and to inhibit the functioning of connexin hemichannels. Finally, we discuss some aspects of structure-activity relationships in order to explain and improve the target selectivity of AAGs.
Collapse
Affiliation(s)
- Clément Dezanet
- Molecular Pharmacochemistry Department, University Grenoble Alpes, CNRS, 470 Rue de la Chimie, F-38000 Grenoble, France; (C.D.); (J.K.)
| | - Julie Kempf
- Molecular Pharmacochemistry Department, University Grenoble Alpes, CNRS, 470 Rue de la Chimie, F-38000 Grenoble, France; (C.D.); (J.K.)
| | - Marie-Paule Mingeot-Leclercq
- Cellular and Molecular Pharmacology Unit, Louvain Drug Research Institute, Catholic University of Louvain, Avenue E. Mounier 73, UCL B1.73.05, 1200 Brussels, Belgium
| | - Jean-Luc Décout
- Molecular Pharmacochemistry Department, University Grenoble Alpes, CNRS, 470 Rue de la Chimie, F-38000 Grenoble, France; (C.D.); (J.K.)
| |
Collapse
|
46
|
Deslouches B, Montelaro RC, Urish KL, Di YP. Engineered Cationic Antimicrobial Peptides (eCAPs) to Combat Multidrug-Resistant Bacteria. Pharmaceutics 2020; 12:pharmaceutics12060501. [PMID: 32486228 PMCID: PMC7357155 DOI: 10.3390/pharmaceutics12060501] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/25/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022] Open
Abstract
The increasing rate of antibiotic resistance constitutes a global health crisis. Antimicrobial peptides (AMPs) have the property to selectively kill bacteria regardless of resistance to traditional antibiotics. However, several challenges (e.g., reduced activity in the presence of serum and lack of efficacy in vivo) to clinical development need to be overcome. In the last two decades, we have addressed many of those challenges by engineering cationic AMPs de novo for optimization under test conditions that typically inhibit the activities of natural AMPs, including systemic efficacy. We reviewed some of the most promising data of the last two decades in the context of the advancement of the field of helical AMPs toward clinical development.
Collapse
Affiliation(s)
- Berthony Deslouches
- Department of Environmental and Occupational Health, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15261, USA;
- Correspondence: ; Tel.: +1-412-624-0103
| | - Ronald C. Montelaro
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA;
| | - Ken L. Urish
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Yuanpu P. Di
- Department of Environmental and Occupational Health, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15261, USA;
| |
Collapse
|