1
|
Zehra M, Shafiq J, Asghar S, Vankwani S, Hasan SM, Khan RMA, Mirza MR, Ahmed A. Proteomic profiling and pre-clinical efficacy of antimicrobial lithium complex and colistin combination against multi-drug resistant Acinetobacter baumannii. Microb Pathog 2025; 200:107335. [PMID: 39864760 DOI: 10.1016/j.micpath.2025.107335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 01/04/2025] [Accepted: 01/23/2025] [Indexed: 01/28/2025]
Abstract
Multi-drug resistant (MDR) Acinetobacter baumannii accounts for high mortality rates in hospital-acquired infections. Colistin is the last resort treatment despite nephrotoxic effects and the emergence of colistin resistant A. baumannii is an emerging issue. To tackle this dilemma, metal complexes can be used to potentiate colistin as combination therapy. However, mechanistic and in vivo studies are lacking to present them as compelling therapeutic options. In this study, a lithium complex ([Li(phen)2sal]) based on salicylic acid and 1,10-phenanthroline was used in synergy with colistin to test its antimicrobial and anti-biofilm potential against MDR A. baumannii. Furthermore, proteomics via mass spectrometry, flow cytometry and scanning electron microscopy was performed to study the cellular targets of the treatment. Combination therapy was also tested against pneumonia model in mice to observe pre-clinical efficacy. The lithium complex showed synergistic and additive interaction with colistin and inhibited >85 % of bacterial cells and biofilm formation in A. baumannii strains. Proteomic analysis revealed that combination therapy downregulated significantly more membrane proteins as compared to the individual doses. Flow cytometry indicated that combination therapy caused hyperpolarization in bacteria which led to the cellular damage as observed in scanning electron microscopy. Combination therapy was non-toxic in mice and reduced the clinical score to 0 with bacterial load lessened to 5.56 ± 0.90 log10 CFU in 48 h. Therefore, parenchymal sections had lesser inflammatory regions with intact alveoli. Consequently, combination therapy can be an alternative therapeutic approach with antimicrobial, anti-biofilm, and pre-clinical efficacy against MDR A. baumannii infection.
Collapse
Affiliation(s)
- Moatter Zehra
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Jazib Shafiq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Sidrah Asghar
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Soma Vankwani
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Syed Mehmood Hasan
- Department of Pathology, Jinnah Sindh Medical University, Karachi, Pakistan
| | - Rao Muhammad Abid Khan
- Department of Microbiology, Sindh Institute of Urology and Transplantation, Karachi, Pakistan
| | - Munazza Raza Mirza
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| | - Ayaz Ahmed
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
2
|
Ahmad M, Aduru SV, Smith RP, Zhao Z, Lopatkin AJ. The role of bacterial metabolism in antimicrobial resistance. Nat Rev Microbiol 2025:10.1038/s41579-025-01155-0. [PMID: 39979446 DOI: 10.1038/s41579-025-01155-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2025] [Indexed: 02/22/2025]
Abstract
The relationship between bacterial metabolism and antibiotic treatment is complex. On the one hand, antibiotics leverage cell metabolism to function. On the other hand, increasing research has highlighted that the metabolic state of the cell also impacts all aspects of antibiotic biology, from drug efficacy to the evolution of antimicrobial resistance (AMR). Given that AMR is a growing threat to the current global antibiotic arsenal and ability to treat infectious diseases, understanding these relationships is key to improving both public and human health. However, quantifying the contribution of metabolism to antibiotic activity and subsequent bacterial evolution has often proven challenging. In this Review, we discuss the complex and often bidirectional relationships between metabolism and the various facets of antibiotic treatment and response. We first summarize how antibiotics leverage metabolism for their function. We then focus on the converse of this relationship by specifically delineating the unique contribution of metabolism to three distinct but related arms of antibiotic biology: antibiotic efficacy, AMR evolution and AMR mechanisms. Finally, we note the relevance of metabolism in clinical contexts and explore the future of metabolic-based strategies for personalized antimicrobial therapies. A deeper understanding of these connections is crucial for the broader scientific community to address the growing crisis of AMR and develop future effective therapeutics.
Collapse
Affiliation(s)
- Mehrose Ahmad
- Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Sai Varun Aduru
- Department of Chemical Engineering, University of Rochester, Rochester, NY, USA
| | - Robert P Smith
- Cell Therapy Institute, Kiran Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
- Department of Medical Education, Kiran Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Zirui Zhao
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Allison J Lopatkin
- Department of Chemical Engineering, University of Rochester, Rochester, NY, USA.
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
3
|
Salazar-Alemán DA, Turner RJ. Escherichia coli growing under antimicrobial gallium nitrate stress reveals new processes of tolerance and toxicity. Sci Rep 2025; 15:1389. [PMID: 39789098 PMCID: PMC11718255 DOI: 10.1038/s41598-025-85772-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025] Open
Abstract
Metals have been used throughout history to manage disease. With the rising incidence of antibiotic-resistant bacterial strains, metal-based antimicrobials (MBAs) have re-emerged as an alternative to combat infections. Gallium nitrate has shown promising efficacy against several pathogens. Although its main toxicity mechanisms have focused on oxidative stress and its "trojan horse" iron mimetic strategy, there are still many knowledge gaps in the full-systems response elicited to counteract its toxic effects, especially in non-acute toxicity models that evaluate longer exposure times. In this study, we explore the transcriptomic response profile of Escherichia coli K12 BW25113 when challenged to grow planktonically for 10 h in the presence of a sublethal inhibitory concentration of gallium nitrate. 581 genes were significantly up-regulated, and 791 down-regulated. Some of the affected biological systems identified in our analysis include iron homeostasis, sulfate metabolism, oxidative and nitrosative stress response, cysteine biosynthesis, anaerobic respiration, toxin-antitoxin interactions, and DNA repair. Altogether, this work provides a valuable snapshot of how E. coli acclimates to this MBA and expands the current knowledge of mechanisms of sensitivity and tolerance. This is a significant step in understanding how bacteria can adjust their physiology to coexist with sublethal concentrations of toxic metals.
Collapse
Affiliation(s)
| | - Raymond J Turner
- Department of Biological Sciences, University of Calgary, Calgary, AB, T2N 1N4, Canada.
| |
Collapse
|
4
|
Choi SR, Kulkarni S, Arnett E, Schlesinger LS, Britigan BE, Narayanasamy P. Efficacy and Possible Mechanism(s) of Action of Gallium Tetraphenylporphyrin Nanoparticles against HIV-TB Coinfection in an In Vitro Granuloma Structure Model. ACS Infect Dis 2024; 10:4279-4290. [PMID: 39499869 DOI: 10.1021/acsinfecdis.4c00639] [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] [Indexed: 11/07/2024]
Abstract
Coinfection of Mycobacterium tuberculosis (Mtb) and human immunodeficiency virus-1 (HIV) is a significant public health concern. Treatment is challenging due to prolonged duration of therapy and drug interactions between antiretroviral therapy (ART) and anti-TB drugs. Noniron gallium meso-tetraphenyl porphyrin (GaTP), a heme mimetic, has shown broad antimicrobial activity. Here, we investigated the efficacy of nanoparticle encapsulating GaTP (GaNP) for the treatment of HIV and Mtb coinfection or single infection in in vitro granuloma structures. GaNP significantly reduced viable Mtb within primary human in vitro granuloma structures infected with Mtb H37Rv-lux and significantly reduced levels of HIV in CD4+ T cells infected with the virus axenically. Similarly, GaNP exhibited significant antimicrobial activity against HIV/Mtb-coinfected granuloma structures created in vitro, which contain the primary immune cells seen in human TB granulomas, including CD4+ T cells and macrophages, as assessed by a luciferase assay for Mtb and p24 ELISA for HIV detection. Furthermore, mechanistic studies revealed that GaTP increases the level of reactive oxygen species and inhibits catalase in Mtb. A significant increase in Mtb nitrate reductase activity was also observed when Mtb was incubated with GaTP and sodium nitrate. Overall, increased oxidative stress and nitrite levels induced by GaTP are consistent with the possibility that GaTP inhibits Mtb aerobic respiration, which leads to incomplete O2 reduction and a shift to respiration using exogenous NO3. These cumulative data continue to support the potential for developing the noniron heme analog GaTP and its nanoparticle GaNP as new therapeutic approaches for the treatment of HIV/Mtb coinfection.
Collapse
Affiliation(s)
- Seoung-Ryoung Choi
- Department of Pathology, Microbiology and Immunology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Smita Kulkarni
- Host-Pathogen Interactions Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, Texas 78227, United States
| | - Eusondia Arnett
- Host-Pathogen Interactions Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, Texas 78227, United States
| | - Larry S Schlesinger
- Host-Pathogen Interactions Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, Texas 78227, United States
| | - Bradley E Britigan
- Department of Pathology, Microbiology and Immunology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Prabagaran Narayanasamy
- Department of Pathology, Microbiology and Immunology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
5
|
Hibbert TM, Whiteley M, Renshaw SA, Neill DR, Fothergill JL. Emerging strategies to target virulence in Pseudomonas aeruginosa respiratory infections. Crit Rev Microbiol 2024; 50:1037-1052. [PMID: 37999716 DOI: 10.1080/1040841x.2023.2285995] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023]
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that is responsible for infections in people living with chronic respiratory conditions, such as cystic fibrosis (CF) and non-CF bronchiectasis (NCFB). Traditionally, in people with chronic respiratory disorders, P. aeruginosa infection has been managed with a combination of inhaled and intravenous antibiotic therapies. However, due in part to the prolonged use of antibiotics in these people, the emergence of multi-drug resistant P. aeruginosa strains is a growing concern. The development of anti-virulence therapeutics may provide a new means of treating P. aeruginosa lung infections whilst also combatting the AMR crisis, as these agents are presumed to exert reduced pressure for the emergence of drug resistance as compared to antibiotics. However, the pipeline for developing anti-virulence therapeutics is poorly defined, and it is currently unclear as to whether in vivo and in vitro models effectively replicate the complex pulmonary environment sufficiently to enable development and testing of such therapies for future clinical use. Here, we discuss potential targets for P. aeruginosa anti-virulence therapeutics and the effectiveness of the current models used to study them. Focus is given to the difficulty of replicating the virulence gene expression patterns of P. aeruginosa in the CF and NCFB lung under laboratory conditions and to the challenges this poses for anti-virulence therapeutic development.
Collapse
Affiliation(s)
- Tegan M Hibbert
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK
| | - Marvin Whiteley
- School of Biological Sciences, Georgia Institute of Technology, Centre for Microbial Dynamics and Infection, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Stephen A Renshaw
- The Bateson Centre and Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Daniel R Neill
- Division of Molecular Microbiology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Joanne L Fothergill
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK
| |
Collapse
|
6
|
Liu C, Cui C, Tan X, Miao J, Wang W, Ren H, Wu H, Zheng C, Ren H, Kang W. pH-mediated potentiation of gallium nitrate against Pseudomonas aeruginosa. Front Microbiol 2024; 15:1464719. [PMID: 39380683 PMCID: PMC11458400 DOI: 10.3389/fmicb.2024.1464719] [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: 07/15/2024] [Accepted: 09/11/2024] [Indexed: 10/10/2024] Open
Abstract
The emergence of multidrug-resistant Pseudomonas aeruginosa isolates is a growing concern for public health, necessitating new therapeutic strategies. Gallium nitrate [Ga(NO3)3], a medication for cancer-related hypercalcemia, has attracted great attention due to its ability to inhibit P. aeruginosa growth and biofilm formation by disrupting iron metabolism. However, the antibacterial efficacy of Ga(NO3)3 is not always satisfactory. It is imperative to investigate the factors that affect the bactericidal effects of Ga(NO3)3 and to identify new ways to enhance its efficacy. This study focused on the impact of pH on P. aeruginosa resistance to Ga(NO3)3, along with the underlying mechanism. The results indicate that acidic conditions could increase the effectiveness of Ga(NO3)3 against P. aeruginosa by promoting the production of pyochelin and gallium uptake. Subsequently, using glutamic acid, a clinically compatible acidic amino acid, the pH was significantly lowered and enhanced the bactericidal and inhibitory efficacy of Ga(NO3)3 against biofilm formation by P. aeruginosa, including a reference strain PA14 and several multidrug-resistant clinical isolates. Furthermore, we used an abscess mouse model to evaluate this combination in vivo; the results show that the combination of glutamic acid and Ga(NO3)3 significantly improved P. aeruginosa clearance. Overall, the present study demonstrates that acidic conditions can increase the sensitivity of P. aeruginosa to Ga(NO3)3. Combining glutamic acid and Ga(NO3)3 is a potential strategy for the treatment of P. aeruginosa infections.
Collapse
Affiliation(s)
- Chang Liu
- School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Chenxuan Cui
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Shijiazhuang Qiaoxi Distinct Center for Disease Control and Prevention, Shijiazhuang, China
| | - Xiaoxin Tan
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Junjie Miao
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Wei Wang
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Han Ren
- Clinical Laboratory, Xinle Traditional Chinese Medicine Hospital, Shijiazhuang, China
| | - Hua Wu
- Clinical Laboratory, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Cuiying Zheng
- Clinical Laboratory, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Huan Ren
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Weijun Kang
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| |
Collapse
|
7
|
de Assis ASJ, Pegoraro GM, Duarte ICS, Delforno TP. Gallium: a decisive "Trojan Horse" against microorganisms. Antonie Van Leeuwenhoek 2024; 118:3. [PMID: 39269546 DOI: 10.1007/s10482-024-02015-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 08/19/2024] [Indexed: 09/15/2024]
Abstract
Controlling multidrug-resistant microorganisms (MRM) has a long history with the extensive and inappropriate use of antibiotics. At the cost of these drugs being scarce, new possibilities have to be explored to inhibit the growth of microorganisms. Thus, metallic compounds have shown to be promising as a viable alternative to contain pathogens resistant to conventional antimicrobials. Gallium (Ga3+) can be highlighted, which is an antimicrobial agent capable of disrupting the essential activities of microorganisms, such as metabolism, cellular respiration and DNA synthesis. It was observed that this occurs due to the similar properties between Ga3+ and iron (Fe3+), which is a fundamental ion for the correct functioning of bacterial activities. The mimetic effect performed by Ga3+ prevents iron transporters from distinguishing both ions and results in the substitution of Fe3+ for Ga3+ and in adverse metabolic disturbances in rapidly growing cells. This review focuses on analyzing the development of research involving Ga3+, elucidating the intracellular incorporation of the "Trojan Horse", summarizing the mechanism of interaction between gallium and iron and comparing the most recent and broad-spectrum studies using gallium-based compounds with antimicrobial scope.
Collapse
Affiliation(s)
- Amanda Stefanie Jabur de Assis
- Center of Science and Technology for Sustainability (CCTS), Laboratory of Applied Microbiology, Federal University of São Carlos (UFSCar), Rodovia João Leme dos Santos, km 110, Itinga,, Sorocaba, SP, 18052-780, Brazil.
| | - Guilherme Manassés Pegoraro
- Center of Science and Technology for Sustainability (CCTS), Laboratory of Applied Microbiology, Federal University of São Carlos (UFSCar), Rodovia João Leme dos Santos, km 110, Itinga,, Sorocaba, SP, 18052-780, Brazil
| | - Iolanda Cristina Silveira Duarte
- Center of Human and Biological Sciences (CCHB), Federal University of São Carlos (UFSCar), Rodovia João Leme dos Santos, km 110, Sorocaba, SP, Brazil
| | | |
Collapse
|
8
|
Liu P, Shui X, Shi M, Kang M, Liu Y, Yang X, Zhang G. The comparative study of two new Schiff bases derived from 5-(thiophene-2-yl)isoxazole as "Off-On-Off" fluorescence sensors for the sequential detection of Ga 3+ and Fe 3+ ions. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 315:124247. [PMID: 38599023 DOI: 10.1016/j.saa.2024.124247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 03/22/2024] [Accepted: 04/02/2024] [Indexed: 04/12/2024]
Abstract
Two new Schiff bases, TIC ((E)-N'-(2-hydroxybenzylidene)-5-(thiophene-2-yl)isoxazole-3-carbohydrazide) and TIE ((E)-N'-(3-ethoxy-2-hydroxybenzylidene)-5-(thiophene-2-yl)isoxazole-3-carbohydrazide), have been designed and synthesized as chemosensors for distinct recognition of Ga3+ and Fe3+ ions. TIE demonstrated a prominent "turn on" response characterized by clear distinguished fluorescence when coordination with Ga3+ ions in the DMSO/H2O buffer solution. In comparison, TIC also showed "turn on" response of blue fluorescence which was more selective and sensitive than that of TIE due to the steric hindrance of ethoxy group of TIE. The newly formed complexes TIC-Ga3+ and TIE-Ga3+ may act as selective "turn-off" fluorescent probes towards Fe3+ ions. Limits of detection of TIC and TIE towards Ga3+ ions were 7.8809 × 10-9 M and 2.6277 × 10-8 M, respectively. Limits of detection of TIC-Ga3+ and TIE-Ga3+ towards Fe3+ ions were 8.6562 × 10-9 M and 3.3764 × 10-7 M, respectively. The molar ratio of the complex between the sensor and Ga3+ or Fe3+ ions were all 1:2 determined through Job's Plot, mass spectrometry, and theoretical calculations. Both sensors were utilized for the determination of target ions in environment water samples, and the portable paper sensors for detecting Ga3+ ions have been successfully developed.
Collapse
Affiliation(s)
- Peng Liu
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Xiaoxing Shui
- Henan Sanmenxia Aoke Chemical Industry Co. Ltd., Sanmenxia 472000, China.
| | - Manman Shi
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Mingyi Kang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Yuanying Liu
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Xiaofeng Yang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Guangyou Zhang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China.
| |
Collapse
|
9
|
Liu JJ, Wang J, Chen QY, Chen F, Wang GJ. Photoresponsive Nanoarchitectonics Based on Copper-Porphyrins for Near-Infrared-Enhanced Bacterial Treatment. ACS OMEGA 2024; 9:24513-24519. [PMID: 38882152 PMCID: PMC11170729 DOI: 10.1021/acsomega.4c00496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/15/2024] [Accepted: 05/15/2024] [Indexed: 06/18/2024]
Abstract
Antibiotic resistance is one of the biggest challenges that causes incurable diseases and endangers public health. Metal-porphyrin-modified nanoarchitectonics can enhance the bacterial affinity and destruction of cell walls. Herein, a new photoresponsive nanoarchitectonics (BPGa@COF-Cu) was synthesized by doping Ga(III) on the surface of black phosphorus (BP) and subsequently loaded into a Cu(II)-based covalent-organic framework (COF-Cu). The COF-Cu was induced by the coupling reaction of terephthalic chloride with amino-substituted porphyrin derivatives (THPP), followed by the coordination of the Cu(II) ion. The material BPGa@COF-Cu is a nanoball, and the mean radius is ca. 250 nm. The photochemical properties of BPGa@COF-Cu show that it efficiently catalyzes H2O2 into ·OH. BPGa@COF-Cu can also produce both singlet oxygen and heat upon 808 nm irradiation. Further, BPGa@COF-Cu was employed to inhibit bacteria, and the results showed that it can destroy the membrane of bacteria. The MIC (minimal inhibition concentration) of BPGa@COF-Cu against E. coli was 1 μg/mL. All the data suggest that BPGa@COF-Cu is a multiple nanoarchitectonics for bacterial treatment.
Collapse
Affiliation(s)
- Jing-Jing Liu
- School of Chemistry and Chemical Engineering, Jiangsu University, Jingkou District, Xuefu Road, Zhenjiang 212013, People's Republic of China
| | - Jun Wang
- School of Chemistry and Chemical Engineering, Jiangsu University, Jingkou District, Xuefu Road, Zhenjiang 212013, People's Republic of China
| | - Qiu-Yun Chen
- School of Chemistry and Chemical Engineering, Jiangsu University, Jingkou District, Xuefu Road, Zhenjiang 212013, People's Republic of China
| | - Feng Chen
- School of Chemistry and Chemical Engineering, Jiangsu University, Jingkou District, Xuefu Road, Zhenjiang 212013, People's Republic of China
| | - Gao-Ji Wang
- School of Chemistry and Chemical Engineering, Jiangsu University, Jingkou District, Xuefu Road, Zhenjiang 212013, People's Republic of China
| |
Collapse
|
10
|
de Oliveira GV, Soares MV, Cordeiro LM, da Silva AF, Venturini L, Ilha L, Baptista FBO, da Silveira TL, Soares FAA, Iglesias BA. Toxicological assessment of photoactivated tetra-cationic porphyrin molecules under white light exposure in a Caenorhabditis elegans model. Toxicology 2024; 504:153793. [PMID: 38574843 DOI: 10.1016/j.tox.2024.153793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024]
Abstract
Photodynamic therapy (PDT) utilizes the potential of photosensitizing substances to absorb light energy and produce reactive oxygen species. Tetra-cationic porphyrins, which have organic or coordination compounds attached to their periphery, are heterocyclic derivatives with well-described antimicrobial and antitumoral properties. This is due to their ability to produce reactive oxygen species and their photobiological properties in solution. Consequently, these molecules are promising candidates as new and more effective photosensitizers with biomedical, environmental, and other biomedical applications. Prior to human exposure, it is essential to establish the toxicological profile of these molecules using in vivo models. In this study, we used Caenorhabditis elegans, a small free-living nematode, as a model for assessing toxic effects and predicting toxicity in preclinical research. We evaluated the toxic effects of porphyrins (neutral and tetra-cationic) on nematodes under dark/light conditions. Our findings demonstrate that tetra-methylated porphyrins (3TMeP and 4TMeP) at a concentration of 3.3 µg/mL (1.36 and 0.93 µM) exhibit high toxicity (as evidenced by reduced survival, development, and locomotion) under dark conditions. Moreover, photoactivated tetra-methylated porphyrins induce higher ROS levels compared to neutral (3TPyP and 4TPyP), tetra-palladated (3PdTPyP and 4PdTPyP), and tetra-platinated (3PtTPyP and 4PtTPyP) porphyrins, which may be responsible for the observed toxic effects.
Collapse
Affiliation(s)
- Gabriela Vitória de Oliveira
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Marcell Valandro Soares
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Larissa Marafiga Cordeiro
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Aline Franzen da Silva
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Luiza Venturini
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Larissa Ilha
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Fabiane Bicca Obetine Baptista
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Tássia Limana da Silveira
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Félix Alexandre Antunes Soares
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil.
| | - Bernardo Almeida Iglesias
- Laboratory of Bioinorganic and Porphyrinic Materials, Department of Chemistry, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil.
| |
Collapse
|
11
|
Rosa LD, Oliveira CB, Chaúque BJM, Grando TH, Gressler LT, Bottari N, Monteiro SG. Gallium maltolate, a promising low toxicity drug with curative effect on mice chronically infected with Trypanosoma evansi. Acta Trop 2024; 252:107148. [PMID: 38354996 DOI: 10.1016/j.actatropica.2024.107148] [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: 12/20/2023] [Revised: 02/05/2024] [Accepted: 02/11/2024] [Indexed: 02/16/2024]
Abstract
Trypanosoma evansi is a flagellate protozoan that infects a wide range of hosts, especially horses. Clinically, the infection is characterized by rapid weight loss, anemia and mobility disorders. This study evaluated the efficacy of treatment gallium maltolate (GaM) in rats infected with T. evansi in the acute and chronic phases of the disease and its influence on the enzyme and blood parameters. 48 animals (Rattus norvegicus) were divided into 8 groups (A-H) of 6 animals each, namely: A: (negative control) uninfected; B: acutely infected positive control; C: chronically infected positive control; D: acutely infected, treated with GaM for 7 days post infection (p.i.); E: acutely infected treated with GaM for 3 days before infection (b.i) and 7 days p.i.; F: chronically infected, treated with GaM for 7 days p.i.; G: chronically infected, treated with GaM for 3 days b.i. and 7 days p.i.; and H: uninfected treated with GaM for 10 days. Acute infected animals (B, D and E) had a progressive increase in parasitemia and were died or euthanized before completing treatment days (5th days p.i.) as they had high parasitemia (over 100 field trypanosomes in the blood smear). Thus, it can be concluded that GaM was not effective against an acute infection. In untreated chronically infected animals (C) the parasitemia also increased progressively and they were euthanized on the 7th day p.i.. The chronically infected and treated animals (F and G) showed low parasitemia and after treatment became negative, showing no trypanosomes in the bloodstream until the 50th day of the experiment. Thus, we conclude that GaM was effective against chronic infections. In uninfected and treated animals (H) hematological, biochemical and enzymatic parameters had no significant changes when compared to the negative control group (A) demonstrating the low toxicity of GaM.
Collapse
Affiliation(s)
- Luciana Dalla Rosa
- Department of Microbiology, Immunology and Parasitology, Universidade Federal do Rio Grande do Sul, Rua Ramiro, 2600 - Santa Cecília, Building 21116, Porto Alegre, Rio Grande do Sul, Brazil.
| | | | - Beni Jequicene Mussengue Chaúque
- Department of Microbiology, Immunology and Parasitology, Universidade Federal do Rio Grande do Sul, Rua Ramiro, 2600 - Santa Cecília, Building 21116, Porto Alegre, Rio Grande do Sul, Brazil; Postgraduate Program in Biological Sciences, Pharmacology and Therapeutics, UFRGS, Rio Grande do Sul, Brazil; Center of Studies in Science and Technology (NECET), Biology Course, Universidade Rovuma, Niassa Branch, Lichinga, Mozambique
| | | | | | | | | |
Collapse
|
12
|
Zhao Q, Yan J, Wang J, Liu R, Bartlam M. Structural analysis of the ferric-binding protein KfuA from Klebsiella pneumoniae. Biochem Biophys Res Commun 2023; 679:52-57. [PMID: 37669596 DOI: 10.1016/j.bbrc.2023.08.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 08/31/2023] [Indexed: 09/07/2023]
Abstract
Iron acquisition is an essential process of cell physiology for biological systems. In Klebsiella pneumoniae, the siderophore and ferric-acquisition ABC (ATP-Binding-Cassette) transporter KfuABC is utilized for iron uptake. Initial recognition of the various ferric sources in periplasm and transportation across the cytoplasmic membrane is performed by the substrate-binding protein (SBP) KfuA. Here we report the 2.0 Å resolution crystal structure of KfuA from K. pneumoniae, which crystallizes in the space group P1211 with a single monomer in the asymmetric unit. A bound metal ion reveals the residues required for binding ferric ions. Binding analysis shows that ferric iron and the iron-mimicking gallium bind with high affinity to KfuA. Growth curves show that gallium inhibits growth of K. pneumoniae whereas ferric iron enhances it. This work suggests a mechanism whereby gallium effectively competes with ferric iron, disrupting iron-dependent biological functions via binding to KfuA and leading to heightened antimicrobial efficacy. Significantly, humans lack equivalent ABC transporters like SBP KfuA, underscoring the potential of KfuA as an attractive target for therapeutic intervention.
Collapse
Affiliation(s)
- Qi Zhao
- College of Life Sciences, Nankai University, Tianjin, 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Jiaqi Yan
- College of Life Sciences, Nankai University, Tianjin, 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Jingjing Wang
- College of Life Sciences, Nankai University, Tianjin, 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Ruihua Liu
- College of Life Sciences, Nankai University, Tianjin, 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.
| | - Mark Bartlam
- College of Life Sciences, Nankai University, Tianjin, 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China; Nankai International Advanced Research Institute (Shenzhen Futian), Nankai University, Tianjin, 300071, China.
| |
Collapse
|
13
|
Waters JE, Stevens-Cullinane L, Siebenmann L, Hess J. Recent advances in the development of metal complexes as antibacterial agents with metal-specific modes of action. Curr Opin Microbiol 2023; 75:102347. [PMID: 37467616 DOI: 10.1016/j.mib.2023.102347] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 07/21/2023]
Abstract
The mounting burden of antimicrobial resistance (AMR) is one of the most concerning threats to public health worldwide. With low economic incentives and a dwindling supply of new drugs in clinical pipelines, more innovative approaches to novel drug design and development are desperately required. Metal-based compounds are rapidly emerging as an alternative to organic drugs, as they have the ability to kill pathogens via metal-specific modes of action. We herein review recent advances in metal-based antibacterial agents, including metal complexes, metal ions and catalytic metallodrugs. The review concludes with a perspective on the rational design of metal-based antibiotics, and how we can exploit their unique properties to tackle AMR.
Collapse
Affiliation(s)
- Jessica E Waters
- Biological Inorganic Chemistry Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom; Department of Chemistry, King's College London, Britannia House, 7 Trinity Street, London SE1 1DB, United Kingdom
| | - Lars Stevens-Cullinane
- Biological Inorganic Chemistry Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom; Department of Chemistry, King's College London, Britannia House, 7 Trinity Street, London SE1 1DB, United Kingdom
| | - Lukas Siebenmann
- Biological Inorganic Chemistry Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom; Department of Chemistry, King's College London, Britannia House, 7 Trinity Street, London SE1 1DB, United Kingdom
| | - Jeannine Hess
- Biological Inorganic Chemistry Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom; Department of Chemistry, King's College London, Britannia House, 7 Trinity Street, London SE1 1DB, United Kingdom.
| |
Collapse
|
14
|
Guo M, Tian P, Li Q, Meng B, Ding Y, Liu Y, Li Y, Yu L, Li J. Gallium Nitrate Enhances Antimicrobial Activity of Colistin against Klebsiella pneumoniae by Inducing Reactive Oxygen Species Accumulation. Microbiol Spectr 2023; 11:e0033423. [PMID: 37272820 PMCID: PMC10434156 DOI: 10.1128/spectrum.00334-23] [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: 01/23/2023] [Accepted: 05/08/2023] [Indexed: 06/06/2023] Open
Abstract
Klebsiella pneumoniae, a pathogen of critical clinical concern, urgently demands effective therapeutic options owing to its drug resistance. Polymyxins are increasingly regarded as a last-line therapeutic option for the treatment of multidrug-resistant (MDR) Gram-negative bacterial infections. However, polymyxin resistance in K. pneumoniae is an emerging issue. Here, we report that gallium nitrate (GaNt), an antimicrobial candidate, exhibits a potentiating effect on colistin against MDR K. pneumoniae clinical isolates. To further confirm this, we investigated the efficacy of combined GaNt and colistin in vitro using spot dilution and rapid time-kill assays and growth curve inhibition tests and in vivo using a murine lung infection model. The results showed that GaNt significantly increased the antimicrobial activity of colistin, especially in the iron-limiting media. Mechanistic studies demonstrated that bacterial antioxidant activity was repressed by GaNt, as revealed by RNA sequencing (RNA-seq), leading to intracellular accumulation of reactive oxygen species (ROS) in K. pneumoniae, which was enhanced in the presence of colistin. Therefore, oxidative stress induced by GaNt and colistin augments the colistin-mediated killing of wild-type cells, which can be abolished by dimethyl sulfoxide (DMSO), an effective ROS scavenger. Collectively, our study indicates that GaNt has a notable impact on the antimicrobial activity of colistin against K. pneumoniae, revealing the potential of GaNt as a novel colistin adjuvant to improve the treatment outcomes of bacterial infections. IMPORTANCE This study aimed to determine the antimicrobial activity of GaNt combined with colistin against Klebsiella pneumoniae in vitro and in vivo. Our results suggest that by combining GaNt with colistin, antioxidant activity was suppressed and reactive oxygen species accumulation was induced in bacterial cells, enhancing antimicrobial activity against K. pneumoniae. We found that GaNt functioned as an antibiotic adjuvant when combined with colistin by inhibiting the growth of multidrug-resistant K. pneumoniae. Our study provides insight into the use of an adjuvant to boost the antibiotic potential of colistin for treating infections caused by multidrug-resistant K. pneumoniae.
Collapse
Affiliation(s)
- Mingjuan Guo
- Department of Infectious Disease, The Chaohu Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ping Tian
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qingqing Li
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bao Meng
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuting Ding
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yanyan Liu
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Center for Surveillance of Bacterial Resistance, Hefei, China
- Institute of Bacterial Resistance, Anhui Medical University, Hefei, Anhui, China
| | - Yasheng Li
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Center for Surveillance of Bacterial Resistance, Hefei, China
- Institute of Bacterial Resistance, Anhui Medical University, Hefei, Anhui, China
| | - Liang Yu
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Center for Surveillance of Bacterial Resistance, Hefei, China
- Institute of Bacterial Resistance, Anhui Medical University, Hefei, Anhui, China
| | - Jiabin Li
- Department of Infectious Disease, The Chaohu Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Center for Surveillance of Bacterial Resistance, Hefei, China
- Institute of Bacterial Resistance, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
15
|
Choi SR, Talmon GA, Hearne K, Woo J, Truong VL, Britigan BE, Narayanasamy P. Combination Therapy with Gallium Protoporphyrin and Gallium Nitrate Exhibits Enhanced Antimicrobial Activity In Vitro and In Vivo against Methicillin-Resistant Staphylococcus aureus. Mol Pharm 2023; 20:4058-4070. [PMID: 37471668 DOI: 10.1021/acs.molpharmaceut.3c00223] [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] [Indexed: 07/22/2023]
Abstract
There is a major need for the development of new therapeutics to combat antibiotic-resistant Staphylococcus aureus. Recently, gallium (Ga)-based complexes have shown promising antimicrobial effects against various bacteria, including multidrug-resistant organisms, by targeting multiple heme/iron-dependent metabolic pathways. Among these, Ga protoporphyrin (GaPP) inhibits bacterial growth by targeting heme pathways, including aerobic respiration. Ga(NO3)3, an iron mimetic, disrupts elemental iron pathways. Here, we demonstrate the enhanced antimicrobial activity of the combination of GaPP and Ga(NO3)3 against methicillin-resistant S. aureus (MRSA) under iron-limited conditions, including small colony variants (SCV). This therapy demonstrated significant antimicrobial activity without inducing slow-growing SCV. We also observed that the combination of GaPP and Ga(NO3)3 inhibited the MRSA catalase but not above that seen with Ga(NO3)3 alone. Neither GaPP nor Ga(NO3)3 alone or their combination inhibited the dominant superoxide dismutase expressed (SodA) under the iron-limited conditions examined. Intranasal administration of the combination of the two compounds improved drug biodistribution in the lungs compared to intraperitoneal administration. In a murine MRSA lung infection model, we observed a significant increase in survival and decrease in MRSA lung CFUs in mice that received combination therapy with intranasal GaPP and Ga(NO3)3 compared to untreated control or mice receiving GaPP or Ga(NO3)3 alone. No drug-related toxicity was observed as assessed histologically in the spleen, lung, nasal cavity, and kidney for both single and repeated doses of 10 mg Ga /Kg of mice over 13 days. Our results strongly suggest that GaPP and Ga(NO3)3 in combination have excellent synergism and potential to be developed as a novel therapy for infections with S. aureus.
Collapse
Affiliation(s)
- Seoung-Ryoung Choi
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Geoffrey A Talmon
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Kenneth Hearne
- Aridis Pharmaceuticals, Los Gatos, California 95032, United States
| | - Jennifer Woo
- Aridis Pharmaceuticals, Los Gatos, California 95032, United States
| | - Vu L Truong
- Aridis Pharmaceuticals, Los Gatos, California 95032, United States
| | - Bradley E Britigan
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Department of Internal Medicine and Research Service, Veterans Affairs Medical Center-Nebraska Western Iowa, Omaha, Nebraska 68105, United States
| | - Prabagaran Narayanasamy
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
16
|
Vaillancourt M, Galdino ACM, Limsuwannarot SP, Celedonio D, Dimitrova E, Broerman M, Bresee C, Doi Y, Lee JS, Parks WC, Jorth P. A compensatory RNase E variation increases Iron Piracy and Virulence in multidrug-resistant Pseudomonas aeruginosa during Macrophage infection. PLoS Pathog 2023; 19:e1010942. [PMID: 37027441 PMCID: PMC10115287 DOI: 10.1371/journal.ppat.1010942] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 04/19/2023] [Accepted: 03/24/2023] [Indexed: 04/08/2023] Open
Abstract
During chronic cystic fibrosis (CF) infections, evolved Pseudomonas aeruginosa antibiotic resistance is linked to increased pulmonary exacerbations, decreased lung function, and hospitalizations. However, the virulence mechanisms underlying worse outcomes caused by antibiotic resistant infections are poorly understood. Here, we investigated evolved aztreonam resistant P. aeruginosa virulence mechanisms. Using a macrophage infection model combined with genomic and transcriptomic analyses, we show that a compensatory mutation in the rne gene, encoding RNase E, increased pyoverdine and pyochelin siderophore gene expression, causing macrophage ferroptosis and lysis. We show that iron-bound pyochelin was sufficient to cause macrophage ferroptosis and lysis, however, apo-pyochelin, iron-bound pyoverdine, or apo-pyoverdine were insufficient to kill macrophages. Macrophage killing could be eliminated by treatment with the iron mimetic gallium. RNase E variants were abundant in clinical isolates, and CF sputum gene expression data show that clinical isolates phenocopied RNase E variant functions during macrophage infection. Together these data show how P. aeruginosa RNase E variants can cause host damage via increased siderophore production and host cell ferroptosis but may also be targets for gallium precision therapy.
Collapse
Affiliation(s)
- Mylene Vaillancourt
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Anna Clara Milesi Galdino
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Sam P. Limsuwannarot
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Diana Celedonio
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Elizabeth Dimitrova
- Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Matthew Broerman
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Catherine Bresee
- Biostatistics Core, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Yohei Doi
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Janet S. Lee
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - William C. Parks
- Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Peter Jorth
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| |
Collapse
|
17
|
Stelitano G, Cocorullo M, Mori M, Villa S, Meneghetti F, Chiarelli LR. Iron Acquisition and Metabolism as a Promising Target for Antimicrobials (Bottlenecks and Opportunities): Where Do We Stand? Int J Mol Sci 2023; 24:ijms24076181. [PMID: 37047161 PMCID: PMC10094389 DOI: 10.3390/ijms24076181] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
The emergence of multidrug-resistant (MDR) and extensively drug-resistant (XDR) infections is one of the most crucial challenges currently faced by the scientific community. Developments in the fundamental understanding of their underlying mechanisms may open new perspectives in drug discovery. In this review, we conducted a systematic literature search in PubMed, Web of Science, and Scopus, to collect information on innovative strategies to hinder iron acquisition in bacteria. In detail, we discussed the most interesting targets from iron uptake and metabolism pathways, and examined the main chemical entities that exhibit anti-infective activities by interfering with their function. The mechanism of action of each drug candidate was also reviewed, together with its pharmacodynamic, pharmacokinetic, and toxicological properties. The comprehensive knowledge of such an impactful area of research will hopefully reflect in the discovery of newer antibiotics able to effectively tackle the antimicrobial resistance issue.
Collapse
|
18
|
Qiao Y, Li Y, Ye Y, Yu Y, Wang W, Yao K, Zhou M. Gallium-Based Nanoplatform for Combating Multidrug-Resistant Pseudomonas aeruginosa and Postoperative Inflammation in Endophthalmitis Secondary to Cataract Surgery. ACS APPLIED MATERIALS & INTERFACES 2022; 14:51763-51775. [PMID: 36373472 DOI: 10.1021/acsami.2c15834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Postcataract endophthalmitis (PCE), a devastating complication following cataract surgeries, is one of the most crucial diseases causing irreversible eye blindness. Pseudomonas aeruginosa (PA), a multiple-drug-resistance (MDR) pathogen, always leads to uncontrolled infection and severe inflammation in PCE that can be difficult to treat by antibiotics. Therefore, it is urgent to develop new feasible strategies composed of both antibacterial and anti-inflammatory capabilities. Here, we report a multifunctional non-antibiotic nanoplatform (Ga-mSiO2-BFN) comprised of clinically approved gallium, mesoporous silica, and bromfenac (BFN) as a co-modified release system to simultaneously eradicate MDR-PA infection and cure inflammation for PCE. The released gallium ions can disrupt bacterial iron metabolism. Meanwhile, the simultaneously released BFN can suppresses the inflammation both postoperation and postinfection of PCE. In the PCE rabbit model, the slit-lamp dispersion and retro-illumination micrograph, ophthalmic clinical grading, and etiological histopathology analysis demonstrated that Ga-mSiO2-BFN could eradicate the MDR infection and alleviate the secondary inflammation from MDR-PA infection. Moreover, both cellular biocompatibility and in vivo animal model application verified the biocompatibility. A potential antibacterial mechanism implicated in the antibacterial action was demonstrated by comprehensive assays of iron antagonism evolutionary curve, colony autofluorescence, polymerase chain reaction, and electron microscopy, showing a repressing siderophore peptide pyoverdine, pyoverdine synthetase D, and interfering with bacterial DNA synthesis. All composites of our nanoplatform were FDA approved, making the Ga-mSiO2-BFN as a potentially promising therapeutic approach for treating MDR-PA in PCE accompanying satisfactory prognosis and prospects for clinical translations.
Collapse
Affiliation(s)
- Yue Qiao
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou 310009, China
| | - Yangyang Li
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
- Key Laboratory of Women's Reproductive Health Research of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yang Ye
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou 310009, China
| | - Yinhui Yu
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou 310009, China
| | - Wei Wang
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou 310009, China
| | - Ke Yao
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou 310009, China
| | - Min Zhou
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
- Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou 310009, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou 310053, China
| |
Collapse
|
19
|
Scott ZW, Choi SR, Talmon GA, Britigan BE, Narayanasamy P. Combining Gallium Protoporphyrin and Gallium Nitrate Enhances In Vitro and In Vivo Efficacy against Pseudomonas aeruginosa: Role of Inhibition of Bacterial Antioxidant Enzymes and Resultant Increase in Cytotoxic Reactive Oxygen Species. ACS Infect Dis 2022; 8:2096-2105. [PMID: 36049087 DOI: 10.1021/acsinfecdis.2c00196] [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: 01/29/2023]
Abstract
Pseudomonas aeruginosa is a highly antibiotic-resistant opportunistic pathogenic bacteria that is responsible for thousands of deaths each year. Infections with P. aeruginosa disproportionately impact individuals with compromised immune systems as well as cystic fibrosis patients, where P. aeruginosa lung infection is a leading cause of morbidity and mortality. In previous work, we showed that a combination of gallium (Ga) nitrate and Ga protoporphyrin worked well in several bacterial infection models but its mechanism of action (MOA) is unknown. In the current work, we have investigated the MOA of Ga combination therapy in P. aeruginosa and its analysis in the in vivo model. In P. aeruginosa treated with Ga combination therapy, we saw a decrease in catalase and superoxide dismutase (SOD) activity, key antioxidant enzymes, which could correlate with a higher potential for oxidative stress. Consistent with this hypothesis, we found that, following combination therapy, P. aeruginosa demonstrated higher levels of reactive oxygen species, as measured using the redox-sensitive fluorescent probe, H2DCFDA. We also saw that the Ga combination therapy killed phagocytosed bacteria inside macrophages in vitro. The therapy with low dose was able to fully prevent mortality in a murine model of P. aeruginosa lung infection and also significantly reduced lung damage. These results support our previous data that Ga combination therapy acts synergistically to kill P. aeruginosa, and we now show that this may occur through increasing the organism's susceptibility to oxidative stress. Ga combination therapy also showed itself to be effective at treating infection in a murine pulmonary-infection model.
Collapse
Affiliation(s)
- Zachary W Scott
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Seoung-Ryoung Choi
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Geoffrey A Talmon
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Bradley E Britigan
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States.,Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States.,Department of Internal Medicine and Research Service, Veterans Affairs Medical Center-Nebraska Western Iowa, Omaha, Nebraska 68105, United States
| | - Prabagaran Narayanasamy
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
20
|
Awad M, Barnes TJ, Thomas N, Joyce P, Prestidge CA. Gallium Protoporphyrin Liquid Crystalline Lipid Nanoparticles: A Third-Generation Photosensitizer against Pseudomonas aeruginosa Biofilms. Pharmaceutics 2022; 14:pharmaceutics14102124. [PMID: 36297559 PMCID: PMC9610264 DOI: 10.3390/pharmaceutics14102124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 11/07/2022] Open
Abstract
The looming antimicrobial resistance pandemic has encouraged the investigation of antimicrobial photodynamic therapy (aPDT) as a promising technology to combat recalcitrant bacterial infections caused by antibiotic resistant strains. Here, we report on the optimization and effective application of gallium protoporphyrin liquid crystalline lipid nanoparticles (GaPP-LCNP) as a photosensitizer for aPDT against the Gram-negative bacteria P. aeruginosa in both planktonic and biofilm modes of growth. LCNP significantly enhanced the performance of GaPP as photosensitizer by two-fold, which was correlated with higher antibacterial activity, reducing the viability of planktonic P. aeruginosa by 7 log10 using 0.8 µM GaPP-LCNP and a light dose of 17 J.cm−2. Importantly, GaPP-LCNP also reduced the viability of biofilms by 6 log10 at relatively low light dose of 34.2 J.cm−2 using only 3 µM GaPP-LCNP. The high antibiofilm activity of GaPP-LCNP at low GaPP-LCNP dose indicated the high efficiency and safety profile of GaPP-LCNP as a promising platform for photodynamic inactivation of recalcitrant infections.
Collapse
Affiliation(s)
- Muhammed Awad
- Centre for Pharmaceutical Innovation, University of South Australia, Clinical and Health Sciences, Adelaide 5000, Australia
- Basil Hetzel Institute for Translational Health Research, Woodville 5011, Australia
| | - Timothy J. Barnes
- Centre for Pharmaceutical Innovation, University of South Australia, Clinical and Health Sciences, Adelaide 5000, Australia
| | - Nicky Thomas
- Centre for Pharmaceutical Innovation, University of South Australia, Clinical and Health Sciences, Adelaide 5000, Australia
| | - Paul Joyce
- Centre for Pharmaceutical Innovation, University of South Australia, Clinical and Health Sciences, Adelaide 5000, Australia
| | - Clive A. Prestidge
- Centre for Pharmaceutical Innovation, University of South Australia, Clinical and Health Sciences, Adelaide 5000, Australia
- Correspondence:
| |
Collapse
|
21
|
Qu CC, Liang YT, Wang XQ, Gao S, He ZZ, Sun XY. Gallium-Based Liquid Metal Materials for Antimicrobial Applications. Bioengineering (Basel) 2022; 9:416. [PMID: 36134962 PMCID: PMC9495447 DOI: 10.3390/bioengineering9090416] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
The hazards caused by drug-resistant bacteria are rocketing along with the indiscriminate use of antibiotics. The development of new non-antibiotic antibacterial drugs is urgent. The excellent biocompatibility and diverse multifunctionalities of liquid metal have stimulated the studies of antibacterial application. Several gallium-based antimicrobial agents have been developed based on the mechanism that gallium (a type of liquid metal) ions disorder the normal metabolism of iron ions. Other emerging strategies, such as physical sterilization by directly using LM microparticles to destroy the biofilm of bacteria or thermal destruction via infrared laser irradiation, are gaining increasing attention. Different from traditional antibacterial agents of gallium compounds, the pronounced property of gallium-based liquid metal materials would bring innovation to the antibacterial field. Here, LM-based antimicrobial mechanisms, including iron metabolism disorder, production of reactive oxygen species, thermal injury, and mechanical destruction, are highlighted. Antimicrobial applications of LM-based materials are summarized and divided into five categories, including liquid metal motors, antibacterial fabrics, magnetic field-responsive microparticles, liquid metal films, and liquid metal polymer composites. In addition, future opportunities and challenges towards the development and application of LM-based antimicrobial materials are presented.
Collapse
Affiliation(s)
- Chun-Chun Qu
- College of Engineering, China Agricultural University, Beijing 100083, China
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- Center for Crop Functional Genomics and Molecular Breeding, China Agricultural University, Beijing 100083, China
- Hainan Institute of China Agricultural University, China Agricultural University, Sanya 572000, China
| | - Yu-Tong Liang
- College of Engineering, China Agricultural University, Beijing 100083, China
| | - Xi-Qing Wang
- Center for Crop Functional Genomics and Molecular Breeding, China Agricultural University, Beijing 100083, China
| | - Shang Gao
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Zhi-Zhu He
- College of Engineering, China Agricultural University, Beijing 100083, China
| | - Xu-Yang Sun
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| |
Collapse
|
22
|
Curcumin-Based β-Diketo Ligands for Ga3+: Thermodynamic Investigation of Potential Metal-Based Drugs. Pharmaceuticals (Basel) 2022; 15:ph15070854. [PMID: 35890151 PMCID: PMC9321647 DOI: 10.3390/ph15070854] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 02/06/2023] Open
Abstract
Curcumin is known for its therapeutic properties; among these, antioxidant, anti-inflammatory and anti-cancer ones stand out. Besides, curcumin metal complexes have shown widespread application in medicine and can be exploited as lead structures for developing metal-based drugs. Unfortunately, curcumin is poorly bioavailable, mainly due to its instability in physiological conditions; this weakness is tightly connected to the presence of the β-diketo moiety undergoing tautomeric equilibrium. Stability and metal-chelating ability can be tuned by modulating the electronic effects and steric hindrance close to the β-diketo moiety; in addition, formation of a metal complex shifts the tautomeric equilibrium towards the β-keto–enol form and increases stability in biological media. Among the metals used in clinical therapy, gallium nitrate has shown to have significant antitumor activity against non-Hodgkin lymphoma and bladder cancer, thus indicating that gallium-based drugs have potential for further development as antineoplastic agents with improved therapeutic activity. Curcuminoids have demonstrated high affinity for gallium(III), allowing the formation of stable positively charged M:L 1:2 β-diketonate complexes that benefit from the therapeutic activity of both the metal and the ligand. Seven new curcumin derivatives were synthesized and completely characterized. The new derivatives retain the solvent-dependent keto–enol tautomerism, with the prevalence of the diketo form in aqueous solution. Enhanced stability in simulated physiological conditions was observed in comparison to the lead compound curcumin. The presence of Ga3+ anticipates the dissociation of the enolic proton, allowing chelate complex formation, and simultaneously it shifts the tautomeric equilibrium towards the keto–enol form. A complete 1H/13C NMR and UV–Vis study was performed to define the metal-to-ligand stoichiometry ratio and the overall stability constants. In addition, we demonstrated that some of the derivatives have increased antiproliferative activity on colon cancer cells compared to curcumin and antioxidant properties. On the whole, the synthesized curcumin-based molecules may act as new gallium(III) chelators with improved stability with respect to curcumin and could open interesting perspectives for the development of novel therapeutic agents for cancer.
Collapse
|
23
|
Evolution of gallium applications in medicine and microbiology: a timeline. Biometals 2022; 35:675-688. [PMID: 35729414 DOI: 10.1007/s10534-022-00406-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 05/26/2022] [Indexed: 11/02/2022]
Abstract
Characterized as a semi-metal, gallium is a chemical element not found freely in the environment, but extracted as a by-product from other minerals. Despite of this, there are several gallium compounds with various applications, such as in the production of semiconductors, light emitting diodes; commercially as a potential cost reducer; pharmacology as cancer-related hypercalcemia, non-Hodgkin' lymphoma, breast and bladder cancer mainly and antimicrobial treatments. The latter will be emphasized in this work due to the contemporary emergence of the development of compounds with antimicrobial potential as a result of the spread of multidrug-resistant bacteria. So, this article discusses the main works, from the discovery of gallium to those that culminated in the current research in microbiology of the last two decades. The antimicrobial activity of gallium can be confirmed through the experimental data and be a promising mean to other investigations, especially due to its iron mimicry ability and the capacity to disrupt microorganisms' metabolism.
Collapse
|
24
|
Choi SR, Britigan BE, Narayanasamy P. Synthesis and in vitro analysis of novel gallium tetrakis(4-methoxyphenyl)porphyrin and its long-acting nanoparticle as a potent antimycobacterial agent. Bioorg Med Chem Lett 2022; 62:128645. [PMID: 35219822 DOI: 10.1016/j.bmcl.2022.128645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/21/2022] [Accepted: 02/21/2022] [Indexed: 11/24/2022]
Abstract
Bacterial heme uptake pathways offer a novel target for antimicrobial drug discovery. Recently, gallium (Ga) porphyrin complexes were found to be effective against mycobacterial heme uptake pathways. The goal of the current study is to build on this foundation and develop a new Ga(III) porphyrin and its nanoparticles, formulated by a single emulsion-evaporation technique to inhibit the growth of Mycobacterium avium complex (MAC) with enhanced properties. Gallium 5,10,15,20-tetrakis(4-methoxyphenyl)porphyrin chloride (GaMeOTP) was synthesized from 5,10,15,20-tetrakis(4-methoxyphenyl)porphyrin and GaCl3. GaMeOTP showed enhanced antimicrobial activity against MAC104 and some clinical M. avium isolates. The synthesized Ga(III) porphyrin antimicrobial activity resulted in the overproduction of reactive oxygen species. Our study also demonstrated that F127 nanoparticles encapsulating GaMeOTP exhibited a smaller size than GaTP nanoparticles and a better duration of activity in MAC-infected macrophages compared to the free GaMeOTP. The nanoparticles were trafficked to endosomal compartments within MAC-infected macrophages, likely contributing to the antimicrobial activity of the drug.
Collapse
Affiliation(s)
- Seoung-Ryoung Choi
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bradley E Britigan
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Department of Internal Medicine and Research Service, Veterans Affairs Medical Center-Nebraska Western Iowa, Omaha, NE, USA
| | - Prabagaran Narayanasamy
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
25
|
Ratia C, Cepas V, Soengas R, Navarro Y, Velasco-de Andrés M, Iglesias MJ, Lozano F, López-Ortiz F, Soto SM. A C ∧S-Cyclometallated Gold(III) Complex as a Novel Antibacterial Candidate Against Drug-Resistant Bacteria. Front Microbiol 2022; 13:815622. [PMID: 35308343 PMCID: PMC8928146 DOI: 10.3389/fmicb.2022.815622] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/13/2022] [Indexed: 12/14/2022] Open
Abstract
The worldwide emergence and spread of infections caused by multidrug-resistant bacteria endangers the efficacy of current antibiotics in the clinical setting. The lack of new antibiotics in the pipeline points to the need of developing new strategies. Recently, gold-based drugs are being repurposed for antibacterial applications. Among them, gold(III) complexes have received increasing attention as metal-based anticancer agents. However, reports on their antibacterial activity are scarce due to stability issues. The present work demonstrates the antibacterial activity of the gold(III) complex 2 stabilized as C∧S-cycloaurated containing a diphenylphosphinothioic amide moiety, showing minimum inhibitory concentration (MIC) values that ranged from 4 to 8 and from 16 to 32 mg/L among Gram-positive and Gram-negative multidrug-resistant (MDR) pathogens, respectively. Complex 2 has a biofilm inhibitory activity of only two to four times than its MIC. We also describe for the first time a potent antibacterial synergistic effect of a gold(III) complex combined with colistin, showing a bactericidal effect in less than 2 h; confirming the role of the outer membrane as a permeability barrier. Complex 2 shows a low rate of internalization in Staphylococcus aureus and Acinetobacter baumannii; it does not interact with replication enzymes or efflux pumps, causes ultrastructural damages in both membrane and cytoplasmic levels, and permeabilizes the bacterial membrane. Unlike control antibiotics, complex 2 did not generate resistant mutants in 30-day sequential cultures. We detected lower cytotoxicity in a non-tumoral THLE-2 cell line (IC50 = 25.5 μM) and no acute toxicity signs in vivo after an i.v. 1-mg/kg dose. The characterization presented here reassures the potential of complex 2 as a new chemical class of antimicrobial agents.
Collapse
Affiliation(s)
- Carlos Ratia
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Virginio Cepas
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Raquel Soengas
- Área de Química Orgánica, Centro de Investigación CIAIMBITAL, Universidad de Almería, Almería, Spain
| | - Yolanda Navarro
- Área de Química Orgánica, Centro de Investigación CIAIMBITAL, Universidad de Almería, Almería, Spain
| | - María Velasco-de Andrés
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - María José Iglesias
- Área de Química Orgánica, Centro de Investigación CIAIMBITAL, Universidad de Almería, Almería, Spain
| | - Francisco Lozano
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Servei d’Immunologia, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Barcelona, Spain
- Departament de Biomedicina, Universitat de Barcelona, Barcelona, Spain
| | - Fernando López-Ortiz
- Área de Química Orgánica, Centro de Investigación CIAIMBITAL, Universidad de Almería, Almería, Spain
| | - Sara M. Soto
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
26
|
Li F, Liu F, Huang K, Yang S. Advancement of Gallium and Gallium-Based Compounds as Antimicrobial Agents. Front Bioeng Biotechnol 2022; 10:827960. [PMID: 35186906 PMCID: PMC8855063 DOI: 10.3389/fbioe.2022.827960] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/13/2022] [Indexed: 12/30/2022] Open
Abstract
With the abuse and misuse of antibiotics, antimicrobial resistance has become a challenging issue in the medical system. Iatrogenic and non-iatrogenic infections caused by multidrug-resistant (MDR) pathogens pose serious threats to global human life and health because the efficacy of traditional antibiotics has been greatly reduced and the resulting socio-economic burden has increased. It is important to find and develop non-antibiotic-dependent antibacterial strategies because the development of new antibiotics can hardly keep pace with the emergence of resistant bacteria. Gallium (III) is a multi-target antibacterial agent that has an excellent antibacterial activity, especially against MDR pathogens; thus, a gallium (III)-based treatment is expected to become a new antibacterial strategy. However, some limitations of gallium ions as antimicrobials still exist, including low bioavailability and explosive release. In recent years, with the development of nanomaterials and clathrates, the progress of manufacturing technology, and the emergence of synergistic antibacterial strategies, the antibacterial activities of gallium have greatly improved, and the scope of application in medical systems has expanded. This review summarizes the advancement of current optimization for these key factors. This review will enrich the knowledge about the efficiency and mechanism of various gallium-based antibacterial agents and provide strategies for the improvement of the antibacterial activity of gallium-based compounds.
Collapse
Affiliation(s)
| | - Fengxiang Liu
- *Correspondence: Fengxiang Liu, ; Kai Huang, ; Shengbing Yang,
| | - Kai Huang
- *Correspondence: Fengxiang Liu, ; Kai Huang, ; Shengbing Yang,
| | - Shengbing Yang
- *Correspondence: Fengxiang Liu, ; Kai Huang, ; Shengbing Yang,
| |
Collapse
|
27
|
Visaggio D, Frangipani E, Hijazi S, Pirolo M, Leoni L, Rampioni G, Imperi F, Bernstein L, Sorrentino R, Ungaro F, Visca P. Variable Susceptibility to Gallium Compounds of Major Cystic Fibrosis Pathogens. ACS Infect Dis 2022; 8:78-85. [PMID: 34965085 PMCID: PMC8762661 DOI: 10.1021/acsinfecdis.1c00409] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
![]()
The decreasing efficacy
of existing antibiotics against pulmonary
pathogens that affect cystic fibrosis (CF) patients calls for the
development of novel antimicrobials. Iron uptake and metabolism are
vital processes for bacteria, hence potential therapeutic targets.
Gallium [Ga(III)] is a ferric iron-mimetic that inhibits bacterial
growth by disrupting iron uptake and metabolism. In this work we evaluate
the efficacy of three Ga(III) compounds, namely, Ga(NO3)3, (GaN), Ga(III)-maltolate (GaM), and Ga(III)-protoporphyrin
IX (GaPPIX), against a collection of CF pathogens using both reference
media and media mimicking biological fluids. All CF pathogens, except Streptococcus pneumoniae, were susceptible to at
least one Ga(III) compound. Notably, Mycobacterium
abscessus and Stenotrophomonas maltophilia were susceptible to all Ga(III) compounds. Achromobacter
xylosoxidans, Burkholderia cepacia complex, and Pseudomonas aeruginosa were more susceptible to GaN and GaM, whereas Staphylococcus
aureus and Haemophilus influenzae were more sensitive to GaPPIX. The results of this study support
the development of Ga(III)-based therapy as a broad-spectrum strategy
to treat CF lung infections.
Collapse
Affiliation(s)
- Daniela Visaggio
- Department of Science, Roma Tre University, 00146 Rome, Italy
- Santa Lucia Fundation IRCCS, 00179 Rome, Italy
| | - Emanuela Frangipani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Sarah Hijazi
- Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Mattia Pirolo
- Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Livia Leoni
- Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Giordano Rampioni
- Department of Science, Roma Tre University, 00146 Rome, Italy
- Santa Lucia Fundation IRCCS, 00179 Rome, Italy
| | - Francesco Imperi
- Department of Science, Roma Tre University, 00146 Rome, Italy
- Santa Lucia Fundation IRCCS, 00179 Rome, Italy
| | | | - Raffaella Sorrentino
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80138 Naples, Italy
| | - Francesca Ungaro
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Paolo Visca
- Department of Science, Roma Tre University, 00146 Rome, Italy
- Santa Lucia Fundation IRCCS, 00179 Rome, Italy
| |
Collapse
|
28
|
Oliveira F, Rohde H, Vilanova M, Cerca N. Fighting Staphylococcus epidermidis Biofilm-Associated Infections: Can Iron Be the Key to Success? Front Cell Infect Microbiol 2021; 11:798563. [PMID: 34917520 PMCID: PMC8670311 DOI: 10.3389/fcimb.2021.798563] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 11/11/2021] [Indexed: 12/29/2022] Open
Abstract
Staphylococcus epidermidis is one of the most important commensal microorganisms of human skin and mucosae. However, this bacterial species is also the cause of severe infections in immunocompromised patients, specially associated with the utilization of indwelling medical devices, that often serve as a scaffold for biofilm formation. S. epidermidis strains are often multidrug resistant and its association with biofilm formation makes these infections hard to treat. Their remarkable ability to form biofilms is widely regarded as its major pathogenic determinant. Although a significant amount of knowledge on its biofilm formation mechanisms has been achieved, we still do not understand how the species survives when exposed to the host harsh environment during invasion. A previous RNA-seq study highlighted that iron-metabolism associated genes were the most up-regulated bacterial genes upon contact with human blood, which suggested that iron acquisition plays an important role in S. epidermidis biofilm development and escape from the host innate immune system. In this perspective article, we review the available literature on the role of iron metabolism on S. epidermidis pathogenesis and propose that exploiting its dependence on iron could be pursued as a viable therapeutic alternative.
Collapse
Affiliation(s)
- Fernando Oliveira
- Centre of Biological Engineering, Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), University of Minho, Braga, Portugal
| | - Holger Rohde
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel Vilanova
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto (ICBAS-UP), Porto, Portugal
| | - Nuno Cerca
- Centre of Biological Engineering, Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), University of Minho, Braga, Portugal
| |
Collapse
|
29
|
Rossato L, Arantes JP, Ribeiro SM, Simionatto S. Antibacterial activity of gallium nitrate against polymyxin-resistant Klebsiella pneumoniae strains. Diagn Microbiol Infect Dis 2021; 102:115569. [PMID: 34775292 DOI: 10.1016/j.diagmicrobio.2021.115569] [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: 01/23/2021] [Revised: 07/19/2021] [Accepted: 10/02/2021] [Indexed: 11/03/2022]
Abstract
Iron uptake and metabolism have become attractive targets for the development of new antibacterial drugs. In this scenario, the FDA-approved iron mimetic metal gallium [Ga (III)] has been successfully researched as an antimicrobial drug. Ga (III) inhibits microbial growth by disrupting ferric iron-dependent metabolic pathways. In this study, we revealed that gallium nitrate III (GaN) inhibits the growth of a collection of twenty polymyxin-resistant Klebsiella pneumoniae strains at concentrations ranging from 2 to 16µg/mL, using a medium, on which the low iron content and the presence of human serum better mimic the in vivo environment. GaN was also successful in protecting Caenorhabditis elegans from polymyxin-resistant K. pneumoniae strains lethal infection, with survival rates of >75%. GaN also exhibited synergism with polymyxin B, suggesting that a polymyxin B-GaN combination holds promise like as one alternative therapy for infections caused by resistant polymyxin B K. pneumoniae strains.
Collapse
Affiliation(s)
- Luana Rossato
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados - UFGD, Dourados, Mato Grosso do Sul, Brazil
| | - Julia Pimentel Arantes
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados - UFGD, Dourados, Mato Grosso do Sul, Brazil
| | - Suzana Meira Ribeiro
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados - UFGD, Dourados, Mato Grosso do Sul, Brazil
| | - Simone Simionatto
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados - UFGD, Dourados, Mato Grosso do Sul, Brazil.
| |
Collapse
|
30
|
Xia W, Li N, Shan H, Lin Y, Yin F, Yu X, Zhou Z. Gallium Porphyrin and Gallium Nitrate Reduce the High Vancomycin Tolerance of MRSA Biofilms by Promoting Extracellular DNA-Dependent Biofilm Dispersion. ACS Infect Dis 2021; 7:2565-2582. [PMID: 34346692 DOI: 10.1021/acsinfecdis.1c00280] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Biofilms, structured communities of bacterial cells embedded in a self-produced extracellular matrix (ECM) which consists of proteins, polysaccharide intercellular adhesins (PIAs), and extracellular DNA (eDNA), play a key role in clinical infections and are associated with an increased morbidity and mortality by protecting the embedded bacteria against drug and immune response. The high levels of antibiotic tolerance render classical antibiotic therapies impractical for biofilm-related infections. Thus, novel drugs and strategies are required to reduce biofilm tolerance and eliminate biofilm-protected bacteria. Here, we showed that gallium, an iron mimetic metal, can lead to nutritional iron starvation and act as dispersal agent triggering the reconstruction and dispersion of mature methicillin-resistant Staphylococcus aureus (MRSA) biofilms in an eDNA-dependent manner. The extracellular matrix, along with the integral bacteria themselves, establishes the integrated three-dimensional structure of the mature biofilm. The structures and compositions of gallium-treated mature biofilms differed from those of natural or antibiotic-survived mature biofilms but were similar to those of immature biofilms. Similar to immature biofilms, gallium-treated biofilms had lower levels of antibiotic tolerance, and our in vitro tests showed that treatment with gallium agents reduced the antibiotic tolerance of mature MRSA biofilms. Thus, the sequential administration of gallium agents (gallium porphyrin and gallium nitrate) and relatively low concentrations of vancomycin (16 mg/L) effectively eliminated mature MRSA biofilms and eradicated biofilm-enclosed bacteria within 1 week. Our results suggested that gallium agents may represent a potential treatment for refractory biofilm-related infections, such as prosthetic joint infections (PJI) and osteomyelitis, and provide a novel basis for future biofilm treatments based on the disruption of normal biofilm-development processes.
Collapse
Affiliation(s)
- Wenyang Xia
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Niya Li
- Department of Laboratory, Shanghai Sixth People’s Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai 200233, China
| | - Haojie Shan
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Yiwei Lin
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Fuli Yin
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Xiaowei Yu
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Zubin Zhou
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| |
Collapse
|
31
|
Choi SR, Talmon GA, Britigan BE, Narayanasamy P. Nanoparticulate β-Cyclodextrin with Gallium Tetraphenylporphyrin Demonstrates in Vitro and in Vivo Antimicrobial Efficacy against Mycobacteroides abscessus and Mycobacterium avium. ACS Infect Dis 2021; 7:2299-2309. [PMID: 34314150 DOI: 10.1021/acsinfecdis.0c00896] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The emergence of drug-resistant pathogens causes the greatest challenge for drug development research. Recently, gallium(III)-based compounds have received great attention as novel antimicrobial agents against drug-resistant pathogens. Here, we synthesized a new β-cyclodextrin Ga nanoparticle (CDGaTP) using Ga tetraphenylporphyrin (GaTP, a hemin analogue) and β-cyclodextrin. The newly synthesized nanoparticle was nontoxic and efficient at a single dose, showing sustained drug release for 15 days in vitro. CDGaTP's activity with transferrin or lactoferrin was tested, and synergism in activity was observed against nontuberculosis mycobacteria (NTM), Mycobacterium avium (M. avium), and Mycobacteroides abscessus. Human serum albumin (HSA) decreased the efficacy of both GaTP and CDGaTP in a concentration-dependent manner. The NTMs incubated with GaTP or CDGaTP significantly produced reactive oxygen species (ROS), indicating potential inhibition of antioxidant enzymes, such as catalase. The single-dose CDGaTP displayed a prolonged intracellular inhibitory activity in an in vitro macrophage infection model against both NTMs. In addition, CDGaTP, not GaTP, was effective in a murine lung M. avium infection model when delivered via intranasal administration. These results suggest that CDGaTP provides new opportunities for the development of gallium-porphyrin based antibiotics.
Collapse
Affiliation(s)
- Seoung-Ryoung Choi
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Geoffrey A. Talmon
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Bradley E. Britigan
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Department of Internal Medicine and Research Service, Veterans Affairs Medical Center−Nebraska Western Iowa, Omaha, Nebraska 68105, United States
| | - Prabagaran Narayanasamy
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
32
|
Qian Y, Xu C, Xiong W, Jiang N, Zheng Y, He X, Ding F, Lu X, Shen J. Dual cross-linked organic-inorganic hybrid hydrogels accelerate diabetic skin wound healing. CHEMICAL ENGINEERING JOURNAL 2021; 417:129335. [DOI: 10.1016/j.cej.2021.129335] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
33
|
Li K, Tian H, Guo A, Jin L, Chen W, Tao B. Gallium (Ga)-strontium (Sr) layered double hydroxide composite coating on titanium substrates for enhanced osteogenic and antibacterial abilities. J Biomed Mater Res A 2021; 110:273-286. [PMID: 34323363 DOI: 10.1002/jbm.a.37284] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 07/17/2021] [Accepted: 07/21/2021] [Indexed: 01/25/2023]
Abstract
Bacterial infection and poor osteogenic capacity can result in the loosing or failure of titanium (Ti)-based implants in the clinic. Therefore, it is urgent to design an effective approach to enhance the osteogenic property and restrict bacterial activity. In this study, a layered double hydroxide (LDH) composed of Ga and Sr ions on Ti substrates by a hydrothermal method, then calcined in 250°C and denoted as LDH250. The scanning electron microscopy (SEM), X-ray diffraction (XRD), and X-ray photoelectron spectroscopy (XPS) were confirmed that the LDH films were successfully formed on the Ti substrates. Importantly, the obtained LDH films can induce an alkaline microenvironment around the Ti surface and regulate the behaviors of osteogenic cells and bacteria. In vitro cellular experiments, the LDH250 can enhance the differentiation of both MC3T3-E1 cells and osteoblasts, stimulate alkaline phosphatase activity (ALP), collagen secretion, and mineralization levels. Meanwhile, antimicrobial assay against Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus) demonstrated that the LDH250 samples had strong antibacterial abilities, which attributed to the release profile of Ga3+ could act as a "Trojan horse" to destroy the bacterial iron metabolism, inducing of local alkaline environment, and producing reactive oxygen species. Hence, this study provides an effective method for reducing antibacterial infection and enhancing the bone integrative capacity of Ti-based implants for orthopedic applications.
Collapse
Affiliation(s)
- Kai Li
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongchuan Tian
- Department of Orthopedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ai Guo
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liguo Jin
- College of Chemistry, Nanchang University, Nanchang, Jiangxi, China
| | - Weizhen Chen
- Department of Laboratory Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, Hangzhou, China
| | - Bailong Tao
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
34
|
Kumaravel V, Nair KM, Mathew S, Bartlett J, Kennedy JE, Manning HG, Whelan BJ, Leyland NS, Pillai SC. Antimicrobial TiO 2 nanocomposite coatings for surfaces, dental and orthopaedic implants. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2021; 416:129071. [PMID: 33642937 PMCID: PMC7899925 DOI: 10.1016/j.cej.2021.129071] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/13/2021] [Accepted: 02/16/2021] [Indexed: 05/03/2023]
Abstract
Engineering of self-disinfecting surfaces to constrain the spread of SARS-CoV-2 is a challenging task for the scientific community because the human coronavirus spreads through respiratory droplets. Titania (TiO2) nanocomposite antimicrobial coatings is one of the ideal remedies to disinfect pathogens (virus, bacteria, fungi) from common surfaces under light illumination. The photocatalytic disinfection efficiency of recent TiO2 nanocomposite antimicrobial coatings for surfaces, dental and orthopaedic implants are emphasized in this review. Mostly, inorganic metals (e.g. copper (Cu), silver (Ag), manganese (Mn), etc), non-metals (e.g. fluorine (F), calcium (Ca), phosphorus (P)) and two-dimensional materials (e.g. MXenes, MOF, graphdiyne) were incorporated with TiO2 to regulate the charge transfer mechanism, surface porosity, crystallinity, and the microbial disinfection efficiency. The antimicrobial activity of TiO2 coatings was evaluated against the most crucial pathogenic microbes such as Escherichia coli, methicillin-resistant Staphylococcus aureus, Pseudomonas aeruginosa, Bacillus subtilis, Legionella pneumophila, Staphylococcus aureus, Streptococcus mutans, T2 bacteriophage, H1N1, HCoV-NL63, vesicular stomatitis virus, bovine coronavirus. Silane functionalizing agents and polymers were used to coat the titanium (Ti) metal implants to introduce superhydrophobic features to avoid microbial adhesion. TiO2 nanocomposite coatings in dental and orthopaedic metal implants disclosed exceptional bio-corrosion resistance, durability, biocompatibility, bone-formation capability, and long-term antimicrobial efficiency. Moreover, the commercial trend, techno-economics, challenges, and prospects of antimicrobial nanocomposite coatings are also discussed briefly.
Collapse
Affiliation(s)
- Vignesh Kumaravel
- Nanotechnology and Bio-Engineering Research Group, Department of Environmental Science, School of Science, Institute of Technology Sligo, Ash Lane, Sligo, Ireland
- Centre for Precision Engineering, Materials and Manufacturing Research (PEM), Institute of Technology Sligo, Ash Lane, Sligo, Ireland
| | - Keerthi M Nair
- Nanotechnology and Bio-Engineering Research Group, Department of Environmental Science, School of Science, Institute of Technology Sligo, Ash Lane, Sligo, Ireland
- Centre for Precision Engineering, Materials and Manufacturing Research (PEM), Institute of Technology Sligo, Ash Lane, Sligo, Ireland
| | - Snehamol Mathew
- Nanotechnology and Bio-Engineering Research Group, Department of Environmental Science, School of Science, Institute of Technology Sligo, Ash Lane, Sligo, Ireland
- Centre for Precision Engineering, Materials and Manufacturing Research (PEM), Institute of Technology Sligo, Ash Lane, Sligo, Ireland
| | - John Bartlett
- Nanotechnology and Bio-Engineering Research Group, Department of Environmental Science, School of Science, Institute of Technology Sligo, Ash Lane, Sligo, Ireland
- Centre for Precision Engineering, Materials and Manufacturing Research (PEM), Institute of Technology Sligo, Ash Lane, Sligo, Ireland
| | | | | | | | | | - Suresh C Pillai
- Nanotechnology and Bio-Engineering Research Group, Department of Environmental Science, School of Science, Institute of Technology Sligo, Ash Lane, Sligo, Ireland
- Centre for Precision Engineering, Materials and Manufacturing Research (PEM), Institute of Technology Sligo, Ash Lane, Sligo, Ireland
| |
Collapse
|
35
|
Liang J, Sun D, Yang Y, Li M, Li H, Chen L. Discovery of metal-based complexes as promising antimicrobial agents. Eur J Med Chem 2021; 224:113696. [PMID: 34274828 DOI: 10.1016/j.ejmech.2021.113696] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/02/2021] [Accepted: 07/07/2021] [Indexed: 01/03/2023]
Abstract
The antimicrobial resistance (AMR) is an intractable problem for the world. Metal ions are essential for the cell process and biological function in microorganisms. Many metal-based complexes with the potential for releasing ions are more likely to be absorbed for their higher lipid solubility. Hence, this review highlights the clinical potential of organometallic compounds for the treatment of infections caused by bacteria or fungi in recent five years. The common scaffolds, including antimicrobial peptides, N-heterocyclic carbenes, Schiff bases, photosensitive-grand-cycle skeleton structures, aliphatic amines-based ligands, and special metal-based complexes are summarized here. We also discuss their therapeutic targets and the risks that should be paid attention to in the future studies, aiming to provide information for researchers on metal-based complexes as antimicrobial agents and inspire the design and synthesis of new antimicrobial drugs.
Collapse
Affiliation(s)
- Jing Liang
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Dejuan Sun
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yueying Yang
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Mingxue Li
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Hua Li
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China; Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Lixia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
36
|
Wang Z, Li J, Benin BM, Yu B, Bunge SD, Abeydeera N, Huang SD, Kim MH. Lipophilic Ga Complex with Broad-Spectrum Antimicrobial Activity and the Ability to Overcome Gallium Resistance in both Pseudomonas aeruginosa and Staphylococcus aureus. J Med Chem 2021; 64:9381-9388. [PMID: 34137262 DOI: 10.1021/acs.jmedchem.1c00656] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Antibiotic resistance (AR) necessitates the discovery of new antimicrobials with alternative mechanisms of action to those employed by conventional antibiotics. One such strategy utilizes Ga3+ to target iron metabolism, a critical process for survival. Still, Ga-based therapies are generally ineffective against Gram-positive bacteria and promote Ga resistance. In response to these drawbacks, we report a lipophilic Ga complex, [Ga2L3(bpy)2] (L = 2,2'-bis(3-hydroxy-1,4-naphthoquinone; bpy = 2,2'-bipyridine)), effective against drug-resistant Pseudomonas aeruginosa (DRPA; minimum inhibitory concentration, MIC = 10 μM = 14.8 μg/mL) and methicillin-resistant Staphylococcus aureus (MRSA, MIC = 100 μM = 148 μg/mL) without iron-limited conditions. Importantly, [Ga2L3(bpy)2] shows noticeably delayed and decreased resistance in both MRSA and DRPA, with only 8× MIC in DRPA and none in MRSA after 30 passages. This is likely due to the dual mode of action afforded by Ga (disruption of iron metabolism) and the ligand (reactive oxygen species production). Overall, [Ga2L3(bpy)2] demonstrates the utility of lipophilic metal complexes with multiple modes of action in combatting AR in Gram-positive and Gram-negative bacteria.
Collapse
Affiliation(s)
- Zhongxia Wang
- Department of Chemistry and Biochemistry, Kent State University, Kent, Ohio 44240, United States
| | - Junfeng Li
- Department of Chemistry and Biochemistry, Kent State University, Kent, Ohio 44240, United States
| | - Bogdan M Benin
- Department of Chemistry and Biochemistry, Kent State University, Kent, Ohio 44240, United States.,Lawsonex, LLC., Rootstown, Ohio 44272, United States
| | - Bing Yu
- Department of Biological Sciences, Kent State University, Kent, Ohio 44240, United States
| | - Scott D Bunge
- Department of Chemistry and Biochemistry, Kent State University, Kent, Ohio 44240, United States
| | - Nalin Abeydeera
- Department of Chemistry and Biochemistry, Kent State University, Kent, Ohio 44240, United States
| | - Songping D Huang
- Department of Chemistry and Biochemistry, Kent State University, Kent, Ohio 44240, United States
| | - Min-Ho Kim
- Department of Biological Sciences, Kent State University, Kent, Ohio 44240, United States
| |
Collapse
|
37
|
Choi SR, Narayanasamy P. Synthesis, optimization, in vitro and in vivo study of bicyclic substituted amine as MenA inhibitor. Bioorg Med Chem Lett 2021; 47:128203. [PMID: 34139327 DOI: 10.1016/j.bmcl.2021.128203] [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: 05/20/2021] [Accepted: 06/10/2021] [Indexed: 11/16/2022]
Abstract
Menaquinone (MK) plays essential role in the electron transport chain (ETC), suggesting MK biosynthesis enzymes as potential targets for drug development. Previously, we demonstrated that Methicillin-resistant Staphylococcus aureus (MRSA) is susceptible to naphthol-based compounds which were developed by mimicking demethylmenaquinone, a product of MenA enzymatic reaction. Here, a series of new MenA inhibitors (4-19) were synthesized and evaluated as MenA inhibitors in this study. The inhibitors were designed to improve growth inhibitory activity against MRSA. Among the MenA inhibitors, bicyclic substituted amine 3 showed MIC of 3 µg/mL, and alkenyl substituted amine 11 showed MIC of 8 µg/mL against USA300. Regrowth of MRSA was observed on addition of MK when exposed to 8 µg/mL of inhibitor 11, supporting inhibition of MK biosynthesis. However, inhibitor 11 did not show efficacy in treating USA300 infected C. elegans up to 25 µg/mL concentration. However, all infected C. elegans survived when exposed to a bicyclic substituted amine 3. Hence, a bicyclic substituted amine was tested in mice for tolerability and biodistribution and observed 100% tolerable and high level of compound accumulation in lungs.
Collapse
Affiliation(s)
- Seoung-Ryoung Choi
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Prabagaran Narayanasamy
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| |
Collapse
|
38
|
Vinuesa V, Cruces R, Nonnoi F, McConnell MJ. Inhibition of LpxC Increases the Activity of Iron Chelators and Gallium Nitrate in Multidrug-Resistant Acinetobacter baumannii. Antibiotics (Basel) 2021; 10:antibiotics10050609. [PMID: 34065605 PMCID: PMC8160660 DOI: 10.3390/antibiotics10050609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/18/2021] [Accepted: 05/18/2021] [Indexed: 11/17/2022] Open
Abstract
Infections caused by multidrug-resistant Acinetobacter baumannii would benefit from the development of novel treatment approaches. Compounds that interfere with bacterial iron metabolism, such as iron chelators and gallium nitrate, have previously been shown to have antimicrobial activity against A. baumannii. In this study, we characterize the effect of LpxC inhibitors on the antimicrobial activity of previously characterized iron chelators, 2,2′-bipyridyl (BIP) and deferiprone (DFP), and gallium nitrate (Ga(NO3)3) against A. baumannii reference strains and multidrug-resistant clinical isolates. The LpxC inhibitor LpxC-2 was synergistic with BIP for 30% of strains tested (FICI values: 0.38–1.02), whereas inhibition with LpxC-4 was synergistic with BIP for 60% of strains tested (FICI values: 0.09–0.75). In time–kill assays, combinations of BIP with both LpxC inhibitors demonstrated synergistic activity, with a more than 3 log10 reduction in bacterial counts compared to BIP alone. LpxC-2 was synergistic with Ga(NO3)3 for 50% of strains tested (FICI values: 0.27–1.0), whereas LpxC-4 was synergistic with Ga(NO3)3 for all strains tested (FICI values: 0.08–≤0.50). In time–kill assays, combinations of Ga(NO3)3 with LpxC-2 and LpxC-4 decreased the growth of both strains compared to each compound separately; however, only the combination with LpxC-4 met the defined criteria for synergy. These results identify a novel synergy between two antimicrobial classes against A. baumannii strains.
Collapse
Affiliation(s)
- Víctor Vinuesa
- Intrahospital Infections Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III, 28220 Madrid, Spain; (V.V.); (R.C.); (F.N.)
| | - Raquel Cruces
- Intrahospital Infections Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III, 28220 Madrid, Spain; (V.V.); (R.C.); (F.N.)
| | - Francesca Nonnoi
- Intrahospital Infections Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III, 28220 Madrid, Spain; (V.V.); (R.C.); (F.N.)
| | - Michael J. McConnell
- Vaxdyn S.L., Avenida Manuel Siurot s/n., 41010 Seville, Spain
- Correspondence: ; Tel.: +34-918-223-869
| |
Collapse
|
39
|
Vinuesa V, McConnell MJ. Recent Advances in Iron Chelation and Gallium-Based Therapies for Antibiotic Resistant Bacterial Infections. Int J Mol Sci 2021; 22:2876. [PMID: 33809032 PMCID: PMC8000330 DOI: 10.3390/ijms22062876] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/04/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
Iron is essential for multiple bacterial processes and is thus required for host colonization and infection. The antimicrobial activity of multiple iron chelators and gallium-based therapies against different bacterial species has been characterized in preclinical studies. In this review, we provide a synthesis of studies characterizing the antimicrobial activity of the major classes of iron chelators (hydroxamates, aminocarboxylates and hydroxypyridinones) and gallium compounds. Special emphasis is placed on recent in-vitro and in-vivo studies with the novel iron chelator DIBI. Limitations associated with iron chelation and gallium-based therapies are presented, with emphasis on limitations of preclinical models, lack of understanding regarding mechanisms of action, and potential host toxicity. Collectively, these studies demonstrate potential for iron chelators and gallium to be used as antimicrobial agents, particularly in combination with existing antibiotics. Additional studies are needed in order to characterize the activity of these compounds under physiologic conditions and address potential limitations associated with their clinical use as antimicrobial agents.
Collapse
Affiliation(s)
| | - Michael J. McConnell
- Intrahospital Infections Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III, 28220 Madrid, Spain;
| |
Collapse
|
40
|
Chen JP, Battini N, Ansari MF, Zhou CH. Membrane active 7-thiazoxime quinolones as novel DNA binding agents to decrease the genes expression and exert potent anti-methicillin-resistant Staphylococcus aureus activity. Eur J Med Chem 2021; 217:113340. [PMID: 33725630 DOI: 10.1016/j.ejmech.2021.113340] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/30/2021] [Accepted: 02/25/2021] [Indexed: 12/16/2022]
Abstract
A novel class of 7-thiazoxime quinolones was developed as potential antimicrobial agents for the sake of bypassing resistance of quinolones. Biological assays revealed that some constructed 7-thiazoxime quinolones possessed effective antibacterial efficiency. Methyl acetate oxime derivative 6l exhibited 32-fold more active than ciprofloxacin against MRSA, which also possessed rapidly bactericidal ability and low toxicity towards mammalian cells. The combination use of 7-thiazoxime quinolone 6l and ciprofloxacin was able to improve antibacterial potency and effectively alleviate bacterial resistance. The preliminarily mechanism exploration revealed that compound 6l could destroy the cell membrane and insert into MRSA DNA to bind with DNA gyrase, then decrease the expression of gyrB and femB genes. The above results strongly suggested that methyl acetate oxime derivative 6l held a promise for combating MRSA infection.
Collapse
Affiliation(s)
- Jin-Ping Chen
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Narsaiah Battini
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Mohammad Fawad Ansari
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Cheng-He Zhou
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China.
| |
Collapse
|
41
|
Lu S, Pan X, Chen D, Xie X, Wu Y, Shang W, Jiang X, Sun Y, Fan S, He J. Broad-spectrum antivirals of protoporphyrins inhibit the entry of highly pathogenic emerging viruses. Bioorg Chem 2021; 107:104619. [PMID: 33450541 PMCID: PMC7784547 DOI: 10.1016/j.bioorg.2020.104619] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/18/2020] [Accepted: 12/27/2020] [Indexed: 12/19/2022]
Abstract
PPIX possesses broad antiviral activities in vitro against a panel of enveloped viruses. PPIX interacts with the lipids of enveloped virions, thereby inhibiting the entry of enveloped viruses into host cells. PPIX shows the antiviral effect in vivo by testing mice infected with the influenza A/Puerto Rico/8/34 (H1N1) virus.
Severe emerging and re-emerging viral infections such as Lassa fever, Avian influenza (AI), and COVID-19 caused by SARS-CoV-2 urgently call for new strategies for the development of broad-spectrum antivirals targeting conserved components in the virus life cycle. Viral lipids are essential components, and viral-cell membrane fusion is the required entry step for most unrelated enveloped viruses. In this paper, we identified a porphyrin derivative of protoporphyrin IX (PPIX) that showed broad antiviral activities in vitro against a panel of enveloped pathogenic viruses including Lassa virus (LASV), Machupo virus (MACV), and SARS-CoV-2 as well as various subtypes of influenza A viral strains with IC50 values ranging from 0.91 ± 0.25 μM to 1.88 ± 0.34 μM. A mechanistic study using influenza A/Puerto Rico/8/34 (H1N1) as a testing strain showed that PPIX inhibits the infection in the early stage of virus entry through biophysically interacting with the hydrophobic lipids of enveloped virions, thereby inhibiting the entry of enveloped viruses into host cells. In addition, the preliminary antiviral activities of PPIX were further assessed by testing mice infected with the influenza A/Puerto Rico/8/34 (H1N1) virus. The results showed that compared with the control group without drug treatment, the survival rate and mean survival time of the mice treated with PPIX were apparently prolonged. These data encourage us to conduct further investigations using PPIX as a lead compound for the rational design of lipid-targeting antivirals for the treatment of infection with enveloped viruses.
Collapse
Affiliation(s)
- Shengsheng Lu
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Xiaoyan Pan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Daiwei Chen
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Xi Xie
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Yan Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Weijuan Shang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Xiaming Jiang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Yuan Sun
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Sheng Fan
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Jian He
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China.
| |
Collapse
|
42
|
Best MG, Cunha-Reis C, Ganin AY, Sousa A, Johnston J, Oliveira AL, Smith DGE, Yiu HHP, Cooper IR. Antimicrobial Properties of Gallium(III)- and Iron(III)-Loaded Polysaccharides Affecting the Growth of Escherichia coli, Staphylococcus aureus, and Pseudomonas aeruginosa, In Vitro. ACS APPLIED BIO MATERIALS 2020; 3:7589-7597. [PMID: 35019499 DOI: 10.1021/acsabm.0c00811] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Antimicrobial resistance (AMR) has become a global concern as many bacterial species have developed resistance to commonly prescribed antibiotics, making them ineffective to treatments. One type of antibiotics, gallium(III) compounds, stands out as possible candidates due to their unique "Trojan horse" mechanism to tackle bacterial growth, by substituting iron(III) in the metabolic cycles of bacteria. In this study, we tested three polysaccharides (carboxymethyl cellulose (CMC), alginate, and pectin) as the binding and delivery agent for gallium on three bacteria (Pseudomonas aeruginosa, Escherichia coli, and Staphylococcus aureus) with a potential bioresponsive delivery mode. Two types of analysis on bacterial growth (minimum inhibitory concentrations (MIC) and minimum bactericidal concentrations (MBC)) were carried out while iron(III)-loaded polysaccharide samples were also tested for comparison. The results suggested that gallium showed an improved inhibitory activity on bacterial growth, in particular gallium(III)-loaded carboxymethyl cellulose (Ga-CMC) sample showing an inhibiting effect on growth for all three tested bacteria. At the MIC for all three bacteria, Ga-CMC showed no cytotoxicity effect on human dermal neonatal fibroblasts (HDNF). Therefore, these bioresponsive gallium(III) polysaccharide compounds show significant potential to be developed as the next-generation antibacterial agents with controlled release capability.
Collapse
Affiliation(s)
- Mark G Best
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton BN2 4GJ, U.K
| | - Cassilda Cunha-Reis
- CBQF-Centro de Biotecnologia e Química Fina Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, R. de Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Alexey Y Ganin
- School of Chemistry, Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, U.K
| | - Aureliana Sousa
- 1i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Jenna Johnston
- Chemical Engineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, U.K
| | - Ana L Oliveira
- CBQF-Centro de Biotecnologia e Química Fina Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, R. de Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - David G E Smith
- The Institute of Biological Chemistry, Biophysics and. Bioengineering (IB3), School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, U.K
| | - Humphrey H P Yiu
- Chemical Engineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, U.K
| | - Ian R Cooper
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton BN2 4GJ, U.K
| |
Collapse
|
43
|
Neill CJ, Harris S, Goldstone RJ, Lau ECHT, Henry TB, Yiu HHP, Smith DGE. Antibacterial Activities of Ga(III) against E. coli Are Substantially Impacted by Fe(III) Uptake Systems and Multidrug Resistance in Combination with Oxygen Levels. ACS Infect Dis 2020; 6:2959-2969. [PMID: 32960047 DOI: 10.1021/acsinfecdis.0c00425] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The continued emergence and spread of antimicrobial resistance (AMR), particularly multidrug resistant (MDR) bacteria, are increasing threats driving the search for additional and alternative antimicrobial agents. The World Health Organization (WHO) has categorized bacterial risk levels and includes Escherichia coli among the highest priority, making this both a convenient model bacterium and a clinically highly relevant species on which to base investigations of antimicrobials. Among many compounds examined for use as antimicrobials, Ga(III) complexes have shown promise. Nonetheless, the spectrum of activities, susceptibility of bacterial species, mechanisms of antimicrobial action, and bacterial characteristics influencing antibacterial actions are far from being completely understood; these are important considerations for any implementation of an effective antibacterial agent. In this investigation, we show that an alteration in growth conditions to physiologically relevant lowered oxygen (anaerobic) conditions substantially increases the minimum inhibitory concentrations (MICs) of Ga(III) required to inhibit growth for 46 wild-type E. coli strains. Several studies have implicated a Trojan horse hypothesis wherein bacterial Fe uptake systems have been linked to the promotion of Ga(III) uptake and result in enhanced antibacterial activity. Our studies show that, conversely, the carriage of accessory Fe uptake systems (Fe_acc) significantly increased the concentrations of Ga(III) required for antibacterial action. Similarly, it is shown that MDR strains are more resistant to Ga(III). The increased tolerance of Fe_acc/MDR strains was apparent under anaerobic conditions. This phenomenon of heightened tolerance has not previously been shown although the mechanisms remain to be defined. Nonetheless, this further highlights the significant contributions of bacterial metabolism, fitness, and AMR characteristics and their implications in evaluating novel antimicrobials.
Collapse
Affiliation(s)
- Christopher J. Neill
- The Institute of Biological Chemistry, Biophysics and Bioengineering (IB3), School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, United Kingdom
| | - Susan Harris
- The Institute of Biological Chemistry, Biophysics and Bioengineering (IB3), School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, United Kingdom
| | - Robert J. Goldstone
- The Institute of Biological Chemistry, Biophysics and Bioengineering (IB3), School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, United Kingdom
| | - Elizabeth C. H. T. Lau
- Chemical Engineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, United Kingdom
| | - Theodore B. Henry
- The Institute of Life and Earth Sciences (ILES), School of Energy, Geoscience, Infrastructure and Society, Heriot-Watt University, Edinburgh EH14 4AS, United Kingdom
| | - Humphrey H. P. Yiu
- Chemical Engineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, United Kingdom
| | - David G. E. Smith
- The Institute of Biological Chemistry, Biophysics and Bioengineering (IB3), School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, United Kingdom
| |
Collapse
|
44
|
Expanding the family of tetrahalide iron complexes: Synthesis, structure and biological applications. Polyhedron 2020. [DOI: 10.1016/j.poly.2020.114755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
45
|
Quantitative proteomic reveals gallium maltolate induces an iron-limited stress response and reduced quorum-sensing in Pseudomonas aeruginosa. J Biol Inorg Chem 2020; 25:1153-1165. [DOI: 10.1007/s00775-020-01831-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/21/2020] [Indexed: 01/12/2023]
|
46
|
Abstract
Traditional organic antimicrobials mainly act on specific biochemical processes such as replication, transcription and translation. However, the emergence and wide spread of microbial resistance is a growing threat for human beings. Therefore, it is highly necessary to design strategies for the development of new drugs in order to target multiple cellular processes that should improve their efficiency against several microorganisms, including bacteria, viruses or fungi. The present review is focused on recent advances and findings of new antimicrobial strategies based on metal complexes. Recent studies indicate that some metal ions cause different types of damages to microbial cells as a result of membrane degradation, protein dysfunction and oxidative stress. These unique modes of action, combined with the wide range of three-dimensional geometries that metal complexes can adopt, make them suitable for the development of new antimicrobial drugs.
Collapse
|
47
|
Choi SR, Switzer B, Britigan BE, Narayanasamy P. Gallium Porphyrin and Gallium Nitrate Synergistically Inhibit Mycobacterial Species by Targeting Different Aspects of Iron/Heme Metabolism. ACS Infect Dis 2020; 6:2582-2591. [PMID: 32845117 DOI: 10.1021/acsinfecdis.0c00113] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
There is an urgent need for new effective and safe antibiotics active against pathogenic mycobacterial species. Gallium (Ga) nitrate (Ga(NO3)3) and Ga porphyrin (GaPP) have each been shown to inhibit the growth of a variety of mycobacterial species. The Ga(III) ion derived from Ga(NO3)3 has the potential to disrupt the mycobacterial Fe(III) uptake mechanisms and utilization, including replacing iron (Fe) in the active site of enzymes, resulting in the disruption of function. Similarly, noniron metalloporphyrins such as heme mimetics, which can be transported across the bacterial membrane via heme-uptake pathways, would potentially block the acquisition of iron-containing heme and bind to heme-utilizing proteins, making them nonfunctional. Given that they likely act on different aspects of mycobacterial Fe metabolism, the efficacy of combining Ga(NO3)3 and GaPP was studied in vitro against Mycobacterium avium, Mycobacterium abscessus, and Mycobacterium tuberculosis (M. tb). The combination was then assessed in vivo in a murine pulmonary infection model of M. abscessus. We observed that Ga(NO3)3 in combination with GaPP exhibited synergistic inhibitory activity against the growth of M. avium, M. tb, and M. abscessus, being most active against M. abscessus. Activity assays indicated that Ga(NO3)3 and GaPP inhibited both catalase and aconitase at high concentrations. However, the combination showed a synergistic effect on the aconitase activity of M. abscessus. The Ga(NO3)3/GaPP combination via intranasal administration showed significant antimicrobial activity in mice infected with M. abscessus. M. abscessus CFU from the lungs of the Ga(NO3)3/GaPP-treated mice was significantly less compared to that of nontreated or single Ga(III)-treated mice. These findings suggest that combinations of different Ga(III) compounds can synergistically target multiple iron/heme-utilizing mycobacterial enzymes. The results support the potential of combination Ga therapy for development against mycobacterial pathogens.
Collapse
Affiliation(s)
| | | | - Bradley E. Britigan
- Department of Internal Medicine and Research Service, Veterans Affairs Medical Center−Nebraska Western Iowa, Omaha, Nebraska 68105, United States
| | | |
Collapse
|
48
|
Frei A, Zuegg J, Elliott AG, Baker M, Braese S, Brown C, Chen F, G Dowson C, Dujardin G, Jung N, King AP, Mansour AM, Massi M, Moat J, Mohamed HA, Renfrew AK, Rutledge PJ, Sadler PJ, Todd MH, Willans CE, Wilson JJ, Cooper MA, Blaskovich MAT. Metal complexes as a promising source for new antibiotics. Chem Sci 2020; 11:2627-2639. [PMID: 32206266 PMCID: PMC7069370 DOI: 10.1039/c9sc06460e] [Citation(s) in RCA: 283] [Impact Index Per Article: 56.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/10/2020] [Indexed: 12/16/2022] Open
Abstract
There is a dire need for new antimicrobial compounds to combat the growing threat of widespread antibiotic resistance. With a currently very scarce drug pipeline, consisting mostly of derivatives of known antibiotics, new classes of antibiotics are urgently required. Metal complexes are currently in clinical development for the treatment of cancer, malaria and neurodegenerative diseases. However, only little attention has been paid to their application as potential antimicrobial compounds. We report the evaluation of 906 metal-containing compounds that have been screened by the Community for Open Antimicrobial Drug Discovery (CO-ADD) for antimicrobial activity. Metal-bearing compounds display a significantly higher hit-rate (9.9%) when compared to the purely organic molecules (0.87%) in the CO-ADD database. Out of 906 compounds, 88 show activity against at least one of the tested strains, including fungi, while not displaying any cytotoxicity against mammalian cell lines or haemolytic properties. Herein, we highlight the structures of the 30 compounds with activity against Gram-positive and/or Gram-negative bacteria containing Mn, Co, Zn, Ru, Ag, Eu, Ir and Pt, with activities down to the nanomolar range against methicillin resistant S. aureus (MRSA). 23 of these complexes have not been reported for their antimicrobial properties before. This work reveals the vast diversity that metal-containing compounds can bring to antimicrobial research. It is important to raise awareness of these types of compounds for the design of truly novel antibiotics with potential for combatting antimicrobial resistance.
Collapse
Affiliation(s)
- Angelo Frei
- Centre for Superbug Solutions , Institute for Molecular Bioscience , The University of Queensland , St. Lucia , Queensland 4072 , Australia . ;
| | - Johannes Zuegg
- Centre for Superbug Solutions , Institute for Molecular Bioscience , The University of Queensland , St. Lucia , Queensland 4072 , Australia . ;
| | - Alysha G Elliott
- Centre for Superbug Solutions , Institute for Molecular Bioscience , The University of Queensland , St. Lucia , Queensland 4072 , Australia . ;
| | - Murray Baker
- School of Molecular Sciences , The University of Western Australia , Stirling Highway , 6009 Perth , Australia
| | - Stefan Braese
- Institute of Organic Chemistry , Karlsruhe Institute of Technology (KIT) , Fritz-Haber-Weg 6 , 76131 Karlsruhe , Germany
- Institute of Biological and Chemical Systems - Functional Molecular Systems (IBCS-FMS) , Karlsruhe Institute of Technology (KIT) , Hermann-von-Helmholtz-Platz 1 , D-76344 Eggenstein-Leopoldshafen , Germany
| | - Christopher Brown
- School of Medical Sciences (Discipline of Pharmacology) , University of Sydney , Australia
| | - Feng Chen
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry CV4 7AL , UK
| | - Christopher G Dowson
- Antimicrobial Screening Facility , School of Life Sciences , University of Warwick , Gibbet Hill Road , Coventry CV4 7AL , UK
| | - Gilles Dujardin
- Institute of Molecules and Matter of Le Mans (IMMM) , UMR 6283 CNRS , Le Mans Université , France
| | - Nicole Jung
- Institute of Organic Chemistry , Karlsruhe Institute of Technology (KIT) , Fritz-Haber-Weg 6 , 76131 Karlsruhe , Germany
- Institute of Biological and Chemical Systems - Functional Molecular Systems (IBCS-FMS) , Karlsruhe Institute of Technology (KIT) , Hermann-von-Helmholtz-Platz 1 , D-76344 Eggenstein-Leopoldshafen , Germany
| | - A Paden King
- Department of Chemistry and Chemical Biology , Cornell University , Ithaca , NY 14853 , USA
| | - Ahmed M Mansour
- Chemistry Department , Faculty of Science , Cairo University , Egypt
| | - Massimiliano Massi
- School of Molecular and Life Sciences - Curtin Institute for Functional Materials and Interfaces , Curtin University , Kent Street , 6102 Bentley WA , Australia
| | - John Moat
- Antimicrobial Screening Facility , School of Life Sciences , University of Warwick , Gibbet Hill Road , Coventry CV4 7AL , UK
| | - Heba A Mohamed
- School of Chemistry , University of Leeds , Woodhouse Lane , Leeds LS2 9JT , UK
| | - Anna K Renfrew
- School of Chemistry , The University of Sydney , Sydney , NSW 2006 , Australia
| | - Peter J Rutledge
- School of Medical Sciences (Discipline of Pharmacology) , University of Sydney , Australia
| | - Peter J Sadler
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry CV4 7AL , UK
| | - Matthew H Todd
- School of Chemistry , The University of Sydney , Sydney , NSW 2006 , Australia
- School of Pharmacy , University College London , London , WC1N 1AX , UK
| | - Charlotte E Willans
- School of Chemistry , University of Leeds , Woodhouse Lane , Leeds LS2 9JT , UK
| | - Justin J Wilson
- Department of Chemistry and Chemical Biology , Cornell University , Ithaca , NY 14853 , USA
| | - Matthew A Cooper
- Centre for Superbug Solutions , Institute for Molecular Bioscience , The University of Queensland , St. Lucia , Queensland 4072 , Australia . ;
| | - Mark A T Blaskovich
- Centre for Superbug Solutions , Institute for Molecular Bioscience , The University of Queensland , St. Lucia , Queensland 4072 , Australia . ;
| |
Collapse
|
49
|
Frei A. Metal Complexes, an Untapped Source of Antibiotic Potential? Antibiotics (Basel) 2020; 9:E90. [PMID: 32085590 PMCID: PMC7168053 DOI: 10.3390/antibiotics9020090] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/07/2020] [Accepted: 02/12/2020] [Indexed: 01/08/2023] Open
Abstract
With the widespread rise of antimicrobial resistance, most traditional sources for new drug compounds have been explored intensively for new classes of antibiotics. Meanwhile, metal complexes have long had only a niche presence in the medicinal chemistry landscape, despite some compounds, such as the anticancer drug cisplatin, having had a profound impact and still being used extensively in cancer treatments today. Indeed, metal complexes have been largely ignored for antibiotic development. This is surprising as metal compounds have access to unique modes of action and exist in a wider range of three-dimensional geometries than purely organic compounds. These properties make them interesting starting points for the development of new drugs. In this perspective article, , the encouraging work that has been done on antimicrobial metal complexes, mainly over the last decade, is highlighted. Promising metal complexes, their activity profiles, and possible modes of action are discussed and issues that remain to be addressed are emphasized.
Collapse
Affiliation(s)
- Angelo Frei
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia 4072, Australia
| |
Collapse
|