1
|
Gao Q, Yang H, Sheiber J, Bartolomeu Halicki PC, Liu K, Blanco D, Milhous S, Jin S, Rohde KH, Fleeman RM, Huigens Iii RW. Identification of 6,8-ditrifluoromethyl halogenated phenazine as a potent bacterial biofilm-eradicating agent. Org Biomol Chem 2025; 23:3342-3357. [PMID: 39841058 PMCID: PMC11753200 DOI: 10.1039/d4ob02011a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 01/05/2025] [Indexed: 01/23/2025]
Abstract
Bacterial biofilms are surface-attached communities consisting of non-replicating persister cells encased within an extracellular matrix of biomolecules. Unlike bacteria that have acquired resistance to antibiotics, persister cells enable biofilms to demonstrate innate tolerance toward all classes of conventional antibiotic therapies. It is estimated that 50-80% of bacterial infections are biofilm associated, which is considered the underlying cause of chronic and recurring infections. Herein, we report a modular three-step synthetic route to new halogenated phenazine (HP) analogues from diverse aniline and nitroarene building blocks. The HPs were evaluated for antibacterial and biofilm-killing properties against a panel of lab strains and multidrug-resistant clinical isolates. Several HPs demonstrated potent antibacterial (MIC ≤ 0.39 μM) and biofilm-eradicating activities (MBEC < 10 μM) with 6,8-ditrifluoromethyl-HP 15 demonstrated remarkable biofilm-killing potencies (MBEC = 0.15-1.17 μM) against Gram-positive pathogens, including methicillin-resistant Staphylococcus aureus clinical isolates. Confocal microscopy showed HP 15 induced significant losses in the polysaccharide matrix in MRSA biofilms. In addition, HP 15 showed increased antibacterial activities against dormant Mycobacterium tuberculosis (Mtb, MIC = 1.35 μM) when compared to replicating Mtb (MIC = 3.69 μM). Overall, this new modular route has enabled rapid access to an interesting series of potent halogenated phenazine analogues to explore their unique antibacterial and biofilm-killing properties.
Collapse
Affiliation(s)
- Qiwen Gao
- Department of Pharmaceutical & Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602, USA.
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, USA.
| | - Hongfen Yang
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, USA.
| | - Jeremy Sheiber
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida 32827, USA.
| | - Priscila Cristina Bartolomeu Halicki
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida 32827, USA.
| | - Ke Liu
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, USA.
| | - David Blanco
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, USA.
| | - Sadie Milhous
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, USA.
| | - Shouguang Jin
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, Florida 32610, USA.
| | - Kyle H Rohde
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida 32827, USA.
| | - Renee M Fleeman
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida 32827, USA.
| | - Robert W Huigens Iii
- Department of Pharmaceutical & Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602, USA.
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, USA.
- Department of Chemistry, Franklin College of Arts and Sciences, University of Georgia, Athens, Georgia 30602, USA
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia 30602, USA
| |
Collapse
|
2
|
Risoen KR, Shaw CA, Weimer BC. Nutritional Stress Leads to Persistence and Persister-like Growth in Staphylococcus aureus. Pathogens 2025; 14:251. [PMID: 40137735 PMCID: PMC11944742 DOI: 10.3390/pathogens14030251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/29/2025] Open
Abstract
Staphylococcus aureus is a versatile zoonotic pathogen capable of causing a wide range of infections. Due to the organism's ability to persist, recalcitrant and recurring infections are a major concern for public and animal health. This study investigated the establishment of persistence using two S. aureus strains-ATCC 29740, a bovine mastitis isolate, and USA300, a human clinical isolate-under substrate depletion. This nutritional stress established a persistence phenotype where the strains remained persistent for >120 days at notable concentrations [>2 log10 CFU/mL] and developed persister-like growth, including small colony variant formations. With RT-qPCR, we found the cell density was higher than represented by the plate count while the intracellular ATP remained constant during the persistence phase. These findings indicate that S. aureus has complex survival strategies to support its persistent state, providing a host-specific perspective when addressing recurrent infections in human and animal infectious diseases.
Collapse
Affiliation(s)
| | | | - Bart C. Weimer
- 100K Pathogen Genome Project, Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| |
Collapse
|
3
|
Ali S, Karaynir A, Salih Doğan H, Nachimuthu R, Badu K, Bozdoğan B. Degradation of Preformed Gram-Positive and Gram-Negative Bacterial Biofilms Using Disintegrated and Intact Phages. PHAGE (NEW ROCHELLE, N.Y.) 2025; 6:20-31. [PMID: 40351404 PMCID: PMC12059619 DOI: 10.1089/phage.2024.0029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Introduction Biofilm is a major challenge across several sectors and contributes to serious risks to public health. This study aimed to evaluate the antibiofilm efficacy of disintegrated phages, whose lytic activities have been eliminated, against bacterial biofilms. Methods A total of seven lytic phages were disintegrated by sonication and confirmed to have completely lost their lytic activities by the spot test. Subsequently, both the disintegrated and intact phages were tested on the biofilms produced by five different biofilm-producing bacteria. The effects of heat and proteinase K on the ability of disintegrated phages to disrupt biofilms were determined. Moreover, the structural proteins released after the disintegration of phages were screened for their presence of lipase, amylase, protease, and DNase activities. Genome analysis of all the seven phages were screened for the presence of genes encoding proteins with enzymatic activities. Results The disintegrated phages showed more effectiveness in degrading the bacterial biofilm when compared with intact phages. The amylase test results were positive for all the seven disintegrated phages tested, confirming the presence of starch-degrading enzymes. Genomic analysis of five phages revealed the presence of genes encoding transglycosylases, amidases, and glycosaminidases, which could contribute to biofilm degradation. However, only four of these proteins were also structural proteins of phages. Conclusions Our study demonstrated that disintegrated phages without lytic effects can still possess biofilm degrading ability, probably associated with the enzymatic activities of their structural proteins. This study showed that phages may have activities beyond lytic phage activities exhibited by their structural enzymes.
Collapse
Affiliation(s)
- Sahd Ali
- Recombinant DNA and Protein Research Center (REDPROM), Aydin Adnan Menderes University, Aydin, Türkiye
| | - Abdulkerim Karaynir
- Recombinant DNA and Protein Research Center (REDPROM), Aydin Adnan Menderes University, Aydin, Türkiye
| | - Hanife Salih Doğan
- Recombinant DNA and Protein Research Center (REDPROM), Aydin Adnan Menderes University, Aydin, Türkiye
| | - Ramesh Nachimuthu
- Antibiotic Resistance and Phage Therapy Laboratory, SBST, Vellore Institute of Technology, Vellore, India
| | - Kingsley Badu
- Department of Theoretical and Applied Biology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Bülent Bozdoğan
- Recombinant DNA and Protein Research Center (REDPROM), Aydin Adnan Menderes University, Aydin, Türkiye
- Department of Medical Microbiology, Medical Faculty, Aydin Adnan Menderes University, Aydin, Türkiye
| |
Collapse
|
4
|
Gilmore AL, Vu H, Martinez T, Peniata L, Kawaguchi B, Armbruster DA, Ashton NN, Williams DL. In vitro antibiofilm efficacy of ertapenem, tobramycin, and moxifloxacin against biofilms grown in a glass bead or CDC Biofilm Reactor®. PLoS One 2025; 20:e0318487. [PMID: 39928650 PMCID: PMC11809895 DOI: 10.1371/journal.pone.0318487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/16/2025] [Indexed: 02/12/2025] Open
Abstract
Laboratory grown biofilms are used to simulate bacterial growth in diverse environmental conditions and screen the effectiveness of anti-biofilm therapies. Recently, we developed a glass bead biofilm reactor that utilizes low broth volume to provide high-throughput biofilm growth for testing and translation across the research continuum (e.g., benchtop assays to preclinical models). Bioburden per mm2 surface area of Staphylococcus aureus and Pseudomonas aeruginosa biofilms were comparable on beads and CDC Biofilm Reactor® coupons. In this study, we hypothesized that biofilms grown on beads would be more susceptible to ertapenem, moxifloxacin, and tobramycin than those grown on coupons. Results indicated a significant reduction in S. aureus bioburden on glass beads compared to glass coupons following treatment with ertapenem (p = 0.005) and tobramycin (p = 0.014). P. aeruginosa biofilms had smaller differences in antibiotic response between the two systems. There was a significantly greater reduction in bead P. aeruginosa biofilm than coupon when treated with tobramycin (p = 0.035). This work offered insight into how the bead biofilm reactor could be used as a tool for antibiotic screening and translation across the continuum of in vitro to in vivo systems that support development of antimicrobial technology.
Collapse
Affiliation(s)
- Annika L. Gilmore
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States of America
- Department of Orthopaedics, University of Utah, Salt Lake City, Utah, United States of America
- Bone and Biofilm Research Lab, University of Utah, Salt Lake City, Utah, United States of America
| | - Helena Vu
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States of America
- Department of Orthopaedics, University of Utah, Salt Lake City, Utah, United States of America
- Bone and Biofilm Research Lab, University of Utah, Salt Lake City, Utah, United States of America
| | - Travis Martinez
- Department of Orthopaedics, University of Utah, Salt Lake City, Utah, United States of America
- Bone and Biofilm Research Lab, University of Utah, Salt Lake City, Utah, United States of America
| | - Lousili Peniata
- Department of Orthopaedics, University of Utah, Salt Lake City, Utah, United States of America
- Bone and Biofilm Research Lab, University of Utah, Salt Lake City, Utah, United States of America
| | - Brooke Kawaguchi
- Department of Orthopaedics, University of Utah, Salt Lake City, Utah, United States of America
- Bone and Biofilm Research Lab, University of Utah, Salt Lake City, Utah, United States of America
| | | | - Nicholas N. Ashton
- Department of Orthopaedics, University of Utah, Salt Lake City, Utah, United States of America
- Bone and Biofilm Research Lab, University of Utah, Salt Lake City, Utah, United States of America
| | - Dustin L. Williams
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States of America
- Department of Orthopaedics, University of Utah, Salt Lake City, Utah, United States of America
- Bone and Biofilm Research Lab, University of Utah, Salt Lake City, Utah, United States of America
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
- Department of Physical Medicine and Rehabilitation, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| |
Collapse
|
5
|
Takada K, Yoshioka Y, Morikawa K, Ariyoshi W, Yamasaki R. Glucose Supplementation Enhances the Bactericidal Effect of Penicillin and Gentamicin on Streptococcus sanguinis Persisters. Antibiotics (Basel) 2025; 14:36. [PMID: 39858322 PMCID: PMC11762801 DOI: 10.3390/antibiotics14010036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/20/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Streptococcus sanguinis is a leading cause of infective endocarditis (IE), which causes diverse clinical symptoms and even death. Recurrence after treatment is a crucial problem in IE, possibly caused by the presence of "persister" cells, a small bacterial population that can survive antimicrobials. In this study, the residual risk for penicillin G (PCG) and gentamicin (GM), used for treating IE, to induce Streptococcus sanguinis persisters, was investigated. Methods: The bactericidal effects of PCG and GM on S. sanguinis were evaluated. Furthermore, we confirmed whether the S. sanguinis that survived following combination treatment with PCG and GM were persisters. The bactericidal effect of the combination of PCG and GM against S. sanguinis was measured after the addition of glucose or arginine. Results: Following 48 h of treatment with PCG (1600 μg/mL) and GM (64 μg/mL), S. sanguinis survived, albeit with a low bacterial count, indicating the presence of persisters. The addition of glucose or arginine to PCG and GM increased the bactericidal effect on residual persister cells and reduced the number of persister cells. Moreover, the addition of glucose at concentrations of 10 mg/mL or higher was substantially effective in achieving sterilization. Conclusions: Our findings demonstrate that persisters of S. sanguinis that survive antimicrobial treatment may make the treatment of IE challenging, and that combining antimicrobial treatment with glucose is effective for eliminating persisters of S. sanguinis. Taken together, these findings may facilitate the development of novel therapeutic strategies against IE caused by oral streptococcal infection.
Collapse
Affiliation(s)
- Kazuya Takada
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu 803-8580, Fukuoka, Japan; (K.T.); (Y.Y.); (W.A.)
- Division of Oral Functional Development, Department of Health Promotion, Kyushu Dental University, Kitakyushu 803-8580, Fukuoka, Japan;
| | - Yoshie Yoshioka
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu 803-8580, Fukuoka, Japan; (K.T.); (Y.Y.); (W.A.)
| | - Kazumasa Morikawa
- Division of Oral Functional Development, Department of Health Promotion, Kyushu Dental University, Kitakyushu 803-8580, Fukuoka, Japan;
| | - Wataru Ariyoshi
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu 803-8580, Fukuoka, Japan; (K.T.); (Y.Y.); (W.A.)
| | - Ryota Yamasaki
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu 803-8580, Fukuoka, Japan; (K.T.); (Y.Y.); (W.A.)
- Collaborative Research Centre for Green Materials on Environmental Technology, Kyushu Institute of Technology, 1-1 Sensui-chou, Tobata-ku, Kitakyushu 803-8580, Fukuoka, Japan
| |
Collapse
|
6
|
Xiao Y, Wang J, Sun P, Ding T, Li J, Deng Y. Formation and resuscitation of viable but non-culturable (VBNC) yeast in the food industry: A review. Int J Food Microbiol 2025; 426:110901. [PMID: 39243533 DOI: 10.1016/j.ijfoodmicro.2024.110901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
The viable but non-culturable (VBNC) state is a survival strategy adopted by microorganisms in response to unfavorable conditions in the environment. VBNC cells are unable to form colonies but still maintain a low level of activity, posing a potential threat to food safety and public health. Therefore, the development of effective strategies to prevent the formation and resuscitation of VBNC cells of microorganisms is a key challenge in food science and microbiology research. However, current research on VBNC cells has primarily focused on bacteria, with relatively limited reports on fungi. This paper provides a comprehensive and systematic review of yeast in the VBNC state, discussing various factors that induce and facilitate resuscitation, along with detection methods and formation and recovery mechanisms. A comprehensive understanding of the induction and resuscitation of yeast in the VBNC state and exploration of its molecular mechanism hold significant implications for food safety and public health. It is imperative to enhance our comprehension of the underlying mechanisms and contributory factors pertaining to VBNC yeast, thereby facilitating the efficient management of the food fermentation process and ensuring the integrity of food quality and safety.
Collapse
Affiliation(s)
- Yang Xiao
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; School of Food Engineering, Qingdao Institute of Technology, Qingdao 266300, China
| | - Jiayang Wang
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; Key Laboratory of Special Food Processing (Co-construction by Ministry and Province), Ministry of Agriculture Rural Affairs, Qingdao Agricultural University, Qingdao 266109, China; Shandong Technology Innovation Center of Special Food, Qingdao 266109, China
| | - Pengdong Sun
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; Key Laboratory of Special Food Processing (Co-construction by Ministry and Province), Ministry of Agriculture Rural Affairs, Qingdao Agricultural University, Qingdao 266109, China; Shandong Technology Innovation Center of Special Food, Qingdao 266109, China; Qingdao Special Food Research Institute, Qingdao 266109, China
| | - Ting Ding
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; Key Laboratory of Special Food Processing (Co-construction by Ministry and Province), Ministry of Agriculture Rural Affairs, Qingdao Agricultural University, Qingdao 266109, China; Shandong Technology Innovation Center of Special Food, Qingdao 266109, China; Qingdao Special Food Research Institute, Qingdao 266109, China
| | - Jingyuan Li
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; Key Laboratory of Special Food Processing (Co-construction by Ministry and Province), Ministry of Agriculture Rural Affairs, Qingdao Agricultural University, Qingdao 266109, China; Shandong Technology Innovation Center of Special Food, Qingdao 266109, China; Qingdao Special Food Research Institute, Qingdao 266109, China
| | - Yang Deng
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; Key Laboratory of Special Food Processing (Co-construction by Ministry and Province), Ministry of Agriculture Rural Affairs, Qingdao Agricultural University, Qingdao 266109, China; Shandong Technology Innovation Center of Special Food, Qingdao 266109, China; Qingdao Special Food Research Institute, Qingdao 266109, China.
| |
Collapse
|
7
|
Rahman KMT, Amaratunga R, Butzin XY, Singh A, Hossain T, Butzin NC. Rethinking dormancy: Antibiotic persisters are metabolically active, non-growing cells. Int J Antimicrob Agents 2025; 65:107386. [PMID: 39551274 DOI: 10.1016/j.ijantimicag.2024.107386] [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: 02/19/2024] [Revised: 10/23/2024] [Accepted: 11/11/2024] [Indexed: 11/19/2024]
Abstract
OBJECTIVES Bacterial persisters are a subpopulation of multidrug-tolerant cells capable of surviving and resuming activity after exposure to bactericidal antibiotic concentrations, contributing to relapsing infections and the development of antibiotic resistance. In this study, we challenge the conventional view that persisters are metabolically dormant by providing compelling evidence that an isogenic population of Escherichia coli remains metabolically active in persistence. METHODS Using transcriptomic analysis, we examined E. coli persisters at multiple time points following exposure to bactericidal concentrations of ampicillin (Amp). Some genes were consistently upregulated in Amp treated persisters compared to the untreated controls, a change that can only occur in metabolically active cells capable of increasing RNA levels. RESULTS Some of the identified genes have been previously linked to persister cells, while others have not been associated with them before. If persister cells were metabolically dormant, gene expression changes over time would be minimal during Amp treatment. However, network analysis revealed major shifts in gene network activity at various time points of antibiotic exposure. CONCLUSIONS These findings reveal that persisters are metabolically active, non-dividing cells, thereby challenging the traditional view that they are dormant.
Collapse
Affiliation(s)
- K M Taufiqur Rahman
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA
| | - Ruqayyah Amaratunga
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA
| | - Xuan Yi Butzin
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA
| | - Abhyudai Singh
- Electrical & Computer Engineering, University of Delaware, Newark, Delaware, USA
| | - Tahmina Hossain
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA.
| | - Nicholas C Butzin
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA; Department of Chemistry, Biochemistry and Physics, South Dakota State University, Brookings, South Dakota, USA.
| |
Collapse
|
8
|
Salina EG, Martini BA, Sorokin VV, Mulyukin AL. Fate of in vitro cultured Mycobacterium abscessus populations when exposed to moxifloxacin. Front Microbiol 2024; 15:1494147. [PMID: 39669783 PMCID: PMC11635960 DOI: 10.3389/fmicb.2024.1494147] [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: 09/10/2024] [Accepted: 11/11/2024] [Indexed: 12/14/2024] Open
Abstract
Given the current need for predictive persisting model for Mycobacterium abscessus, we adopted a classical assay to study drug-tolerant bacterial persisters, focusing on the behavior of a small antibiotic-insensitive subpopulation during prolonged exposure to moxifloxacin. Our study showed a wide-ranging response of M. abscessus, depending on antibiotic concentration, growth stage of mycobacterial cultures, and the availability of potassium ions in the medium. Mid-logarithmic cultures, initially grown in either balanced or K+-free medium, contained small sup-populations capable of prolonged and stable survival in the presence of moxifloxacin. The response of these mid-log cultures to antibiotic exposure involved initial killing, followed by regrowth at 1-2 MBCs of moxifloxacin or a substantial reduction of the antibiotic-insensitive subpopulation to fewer than 102 CFU/mL at 16 MBCs. In stationary-phase cultures grown in a complete medium, a consistent number of viable cells was observed when exposed to a high dose of moxifloxacin. In contrast, antibiotic-insensitive subpopulations in stationary-phase M. abscessus cultures under potassium-deficient conditions experienced gradual killing across a wide range of moxifloxacin concentrations (1-16 MBCs). Studies on electron microscopy demonstrated that singular cells were rapidly destroyed after relatively short-term exposure to moxifloxacin, while cells in aggregates or clumps persisted longer, explaining the delayed biocidal effect. The small subpopulation that survived under intense moxifloxacin pressure was notably heterogeneous in cell morphology and fine structure, consisting of ovoid forms and cell-wall-deficient cells with reduced size. These findings suggest that the same antibiotic dose may have varying effects on M. abscessus cells, depending on their physiological state and abundance within infected cells or tissues. Taken together, our study may contribute to the development of strategies to combat recalcitrant survivor subpopulations.
Collapse
Affiliation(s)
- Elena G. Salina
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russia
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Billy A. Martini
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russia
| | - Vladimir V. Sorokin
- Winogradsky Institute of Microbiology, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russia
| | - Andrey L. Mulyukin
- Winogradsky Institute of Microbiology, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
9
|
Gilmore A, Badham M, Rudisin W, Ashton N, Williams D. A Bead Biofilm Reactor for High-Throughput Growth and Translational Applications. Microorganisms 2024; 12:1588. [PMID: 39203430 PMCID: PMC11356137 DOI: 10.3390/microorganisms12081588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/19/2024] [Accepted: 08/01/2024] [Indexed: 09/03/2024] Open
Abstract
Bacteria in natural ecosystems such as soil, dirt, or debris preferentially reside in the biofilm phenotype. When a traumatic injury, such as an open fracture, occurs, these naturally dwelling biofilms and accompanying foreign material can contaminate the injury site. Given their high tolerance of systemic levels of antibiotics that may be administered prophylactically, biofilms may contribute to difficult-to-treat infections. In most animal models, planktonic bacteria are used as initial inocula to cause infection, and this might not accurately mimic clinically relevant contamination and infection scenarios. Further, few approaches and systems utilize the same biofilm and accompanying substrate throughout the experimental continuum. In this study, we designed a unique reactor to grow bacterial biofilms on up to 50 silica beads that modeled environmental wound contaminants. The data obtained indicated that the reactor system repeatably produced mature Staphylococcus aureus and Pseudomonas aeruginosa biofilms on the silica beads, with an average of 5.53 and 6.21 log10 colony-forming units per mm2, respectively. The bead substrates are easily manipulable for in vitro or in vivo applications, thus improving translatability. Taken together, the bead biofilm reactor presented herein may be a useful system for repeatably growing established biofilms on silica beads that could be used for susceptibility testing and as initial inocula in future animal models of trauma-related injuries.
Collapse
Affiliation(s)
- Annika Gilmore
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84108, USA (W.R.)
| | - Marissa Badham
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84108, USA (W.R.)
| | - Winston Rudisin
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84108, USA (W.R.)
| | - Nicholas Ashton
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84108, USA (W.R.)
| | - Dustin Williams
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84108, USA (W.R.)
- Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
- Department of Physical Medicine and Rehabilitation, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
10
|
Pham HN, Than TDN, Nguyen HA, Vu DH, Phung TH, Nguyen TK. Antibiotic Resistance, Biofilm Formation, and Persistent Phenotype of Klebsiella pneumoniae in a Vietnamese Tertiary Hospital: A Focus on Amikacin. Microb Drug Resist 2024; 30:203-209. [PMID: 38512170 DOI: 10.1089/mdr.2023.0267] [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: 03/22/2024] Open
Abstract
Klebsiella pneumoniae stands out as a major opportunistic pathogen responsible for both hospital- and community-acquired bacterial infections. This study comprehensively assesses the antibiotic resistance, amikacin persistent patterns, and biofilm-forming ability of 247 isolates of K. pneumoniae obtained from an intensive care unit of a tertiary hospital in Vietnam. Microdilution assays, conducted on a 96-well plate, determined the minimum inhibitory concentrations (MICs) of amikacin. Susceptibility data for other antibiotics were gathered from the antibiogram profile. Stationary-phase bacteria were exposed to 50 × MIC, and viable bacteria counts were measured to determine amikacin persistence. Biofilm forming capacity on 96-well polystyrene surfaces was assessed by biomass and viable bacteria. The prevalence of resistance was notably high across most antibiotics, with 64.8% classified as carbapenem-resistant K. pneumoniae and 81.4% as multidrug resistant. Amikacin, however, exhibited a relatively low rate of resistance. Of the isolates, 58.2% demonstrated a moderate to strong biofilm formation capacity, and these were found to be poorly responsive to amikacin. K. pneumoniae reveals a significant inclination for amikacin persistence, with ∼45% of isolates displaying an antibiotic antibiotic-survival ratio exceeding 10%. The study sheds light on challenges in treating of K. pneumoniae infection in Vietnam, encompassing a high prevalence of antibiotic resistance, a substantial ability to form biofilm, and a notable rate of antibiotic persistence.
Collapse
Affiliation(s)
- Hong Nhung Pham
- Department Microbiology, Hanoi Medical University, Hanoi, Vietnam
- Department of Microbiology, Bach Mai Hospital, Hanoi, Vietnam
| | | | - Hoang Anh Nguyen
- National Drug Information and Adverse Drug Reactions Monitoring Centre, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Dinh Hoa Vu
- National Drug Information and Adverse Drug Reactions Monitoring Centre, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Thanh Huong Phung
- Faculty of Biotechnology, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Tiep Khac Nguyen
- Faculty of Biotechnology, Hanoi University of Pharmacy, Hanoi, Vietnam
| |
Collapse
|
11
|
Petit M, Tessier J, Sahli C, Schmitzer AR. Confronting the Threat: Designing Highly Effective bis-Benzimidazolium Agents to Overcome Biofilm Persistence and Antimicrobial Resistance. ACS Infect Dis 2023; 9:2202-2214. [PMID: 37882623 DOI: 10.1021/acsinfecdis.3c00289] [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: 10/27/2023]
Abstract
The objective of this study is to take the initial steps toward developing novel antibiotics to counteract the escalating problem of antimicrobial and bacterial persistence, particularly in relation to biofilms. Our approach involves emulating the structural characteristics of cationic antimicrobial peptides. To circumvent resistance development, we have designed a library of bis-benzimidazolium salts that selectively target the microbial membranes in a nonspecific manner. To explore their structure-activity relationship, we conducted experiments using these compounds on various pathogens known for their resistance to conventional antibiotics, including Gram-positive methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant Enterococcus faecium (VRE), and Gram-negative Escherichia coli (E. coli). Notably, two bis-benzimidazolium salts exhibited robust antimicrobial activity while maintaining a high level of selectivity compared with mammalian cells. Our investigations revealed significant antibiofilm activity, as these compounds rapidly acted against established biofilms. In addition, bis-benzimidazolium compounds exhibited consistent results in resistance development and cross-resistance studies. Consequently, amphiphilic bis-benzimidazolium salts hold promise as potential candidates to combat resistance-associated infections.
Collapse
Affiliation(s)
- Maude Petit
- Département de Chimie, Faculté des Arts et des Sciences, Université de Montréal, Campus MIL, 1375, Ave. Thérèse Lavoie-Roux, Montréal, Québec H2 V 0B3, Canada
| | - Jérémie Tessier
- Département de Chimie, Faculté des Arts et des Sciences, Université de Montréal, Campus MIL, 1375, Ave. Thérèse Lavoie-Roux, Montréal, Québec H2 V 0B3, Canada
- Collège Bois-de-Boulogne, 10555 Ave. de Bois-de-Boulogne, Montréal H4N 1L4, Canada
| | - Célia Sahli
- Département de Chimie, Faculté des Arts et des Sciences, Université de Montréal, Campus MIL, 1375, Ave. Thérèse Lavoie-Roux, Montréal, Québec H2 V 0B3, Canada
- CNRS-UMR 7086, Interfaces, Traitements, Organisation et Dynamique des Systèmes (ITODYS), Université Paris Cité, Paris 75013 , France
| | - Andreea R Schmitzer
- Département de Chimie, Faculté des Arts et des Sciences, Université de Montréal, Campus MIL, 1375, Ave. Thérèse Lavoie-Roux, Montréal, Québec H2 V 0B3, Canada
| |
Collapse
|
12
|
Lapa IR, Dos Santos Siqueira F, Cordeiro CF, de Campos MMA, Bonfilio R, de Figueiredo Diniz L, Pereira GM, Hawkes JA, Franco LL, Carvalho DT. Combining eugenol and dihydroeugenol with a piperazine moiety to create new antimicrobial agents that are effective against resistant species. Microb Pathog 2023; 184:106369. [PMID: 37778705 DOI: 10.1016/j.micpath.2023.106369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/03/2023]
Abstract
Historically, the piperazine moiety has been demonstrated to possess pharmacophoric properties, and has subsequently been incorporated in many drugs that have antitumor, antimalarial, antiviral, antibacterial and antifungal properties. Derivatives of eugenol and dihydroeugenol have also been reported as being bioactive compounds. This study reports the synthesis of a range of eugenol/dihydroeugenol - piperazine derivatives which have been tested as antimicrobial compounds against Gram positive, Gram negative and rapid-growing mycobacteria (RGM). The rationale employed in the design of the structural pattern of these new derivatives, provides useful insights into the structure-activity relationships (SAR) of the series. Antimicrobial activity tests were extremely encouraging, with the majority of the synthesised compounds being more active than eugenol and dihydroeugenol starting materials. The antimicrobial potential was most notable against the Gram-negative species K. pneumoniae and P. aeruginosa, but there was also significant performance against the Gram-positive strains S. epidermidis and S. aureus and the Rapidly Growing Mycobacteria (RGM) strains tested. Tests using the synthesised compounds against multidrug-resistance clinical (MDR) isolates also showed high activity. The biofilm inhibition tests using M. fortuitum showed that all evaluated derivatives were able to inhibit biofilm formation even at low concentrations. In terms of structural-activity relationships; the results generated by this study demonstrate that the compounds with bulky substituents on the piperazine subunit were much more active than those with less bulky groups, or no groups. Importantly, the derivatives with a sulfonamide side chain were the most potent compounds. A further observation was that those compounds with a para-substituted benzenesulfonamide ring stand out, regardless of whether this substituent is a donor or an electron-withdrawing group.
Collapse
Affiliation(s)
- Igor Rodrigues Lapa
- Faculdade de Ciências Farmacêuticas, Departamento de Alimentos e Medicamentos, Universidade Federal de Alfenas, MG, 37130-001, Brazil
| | - Fallon Dos Santos Siqueira
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal de Santa Maria, RS, 9710590, Brazil
| | - Cleydson Finotti Cordeiro
- Faculdade de Ciências Farmacêuticas, Departamento de Alimentos e Medicamentos, Universidade Federal de Alfenas, MG, 37130-001, Brazil
| | | | - Rudy Bonfilio
- Faculdade de Ciências Farmacêuticas, Departamento de Alimentos e Medicamentos, Universidade Federal de Alfenas, MG, 37130-001, Brazil
| | - Lívia de Figueiredo Diniz
- Instituto de Ciências Biomédicas, Departamento de Patologia e Parasitologia, Universidade Federal de Alfenas, MG, 37130-001, Brazil
| | - Gabriella Martiniano Pereira
- Instituto de Ciências Biomédicas, Departamento de Patologia e Parasitologia, Universidade Federal de Alfenas, MG, 37130-001, Brazil
| | - Jamie Anthony Hawkes
- Faculdade de Ciências Farmacêuticas, Departamento de Alimentos e Medicamentos, Universidade Federal de Alfenas, MG, 37130-001, Brazil
| | - Lucas Lopardi Franco
- Faculdade de Ciências Farmacêuticas, Departamento de Alimentos e Medicamentos, Universidade Federal de Alfenas, MG, 37130-001, Brazil
| | - Diogo Teixeira Carvalho
- Faculdade de Ciências Farmacêuticas, Departamento de Alimentos e Medicamentos, Universidade Federal de Alfenas, MG, 37130-001, Brazil.
| |
Collapse
|
13
|
Mattiello SP, Barth VC, Scaria J, Ferreira CAS, Oliveira SD. Fluoroquinolone and beta-lactam antimicrobials induce different transcriptome profiles in Salmonella enterica persister cells. Sci Rep 2023; 13:18696. [PMID: 37907566 PMCID: PMC10618250 DOI: 10.1038/s41598-023-46142-8] [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: 08/16/2023] [Accepted: 10/27/2023] [Indexed: 11/02/2023] Open
Abstract
Here, we investigate the transcriptome profiles of two S. Enteritidis and one S. Schwarzengrund isolates that present different persister levels when exposed to ciprofloxacin or ceftazidime. It was possible to note a distinct transcript profile among isolates, time of exposure, and treatment. We could not find a commonly expressed transcript profile that plays a role in persister formation after S. enterica exposure to beta-lactam or fluoroquinolone, as only three DEGs presented the same behavior under the conditions and isolates tested. It appears that the formation of persisters in S. enterica after exposure to ciprofloxacin is linked to the overexpression of genes involved in the SOS response (recA), cell division inhibitor (sulA), iron-sulfur metabolism (hscA and iscS), and type I TA system (tisB). On the other hand, most genes differentially expressed in S. enterica after exposure to ceftazidime appeared to be downregulated and were part of the flagellar assembly apparatus, citrate cycle (TCA cycle), glycolysis/gluconeogenesis, carbon metabolism, bacterial secretion system, quorum sensing, pyruvate metabolism pathway, and biosynthesis of secondary metabolites. The different transcriptome profiles found in S. enterica persisters induced by ciprofloxacin and ceftazidime suggest that these cells modulate their response differently according to each stress.
Collapse
Affiliation(s)
- S P Mattiello
- Laboratório de Imunologia e Microbiologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, PUCRS, Av. Ipiranga, 6681, Porto Alegre, 90619-900, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, PUCRS, Porto Alegre, Brazil
- College of Mathematics and Science, The University of Tennessee Southern, UTS, Pulaski, TN, USA
- Department of Veterinary and Biomedical Sciences, South Dakota State University, SDSU, Brookings, SD, USA
| | - V C Barth
- Laboratório de Imunoterapia, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - J Scaria
- Department of Veterinary and Biomedical Sciences, South Dakota State University, SDSU, Brookings, SD, USA
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK, USA
| | - C A S Ferreira
- Laboratório de Imunologia e Microbiologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, PUCRS, Av. Ipiranga, 6681, Porto Alegre, 90619-900, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, PUCRS, Porto Alegre, Brazil
| | - S D Oliveira
- Laboratório de Imunologia e Microbiologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, PUCRS, Av. Ipiranga, 6681, Porto Alegre, 90619-900, Brazil.
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, PUCRS, Porto Alegre, Brazil.
| |
Collapse
|
14
|
Bicer M, Fidan O. Can mesenchymal stem/stromal cells and their secretomes combat bacterial persisters? World J Microbiol Biotechnol 2023; 39:276. [PMID: 37567959 DOI: 10.1007/s11274-023-03725-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 08/07/2023] [Indexed: 08/13/2023]
Abstract
The increasing number of life-threatening infections caused by persister bacteria is associated with various issues, including antimicrobial resistance and biofilm formation. Infections due to persister cells are often difficult to suppress without the use of last-resort antibiotics. Throughout the world, bacterial persistence and resistance create an unmet clinical demand for the exploration of newly introduced therapeutic approaches. Mesenchymal stem / stromal cells (MSCs) have an antimicrobial activity to protect against bacterial infections, including those caused by bacterial persisters. MSCs have substantial potential to secrete antimicrobial peptides (AMPs), including cathelicidin, beta-defensins, lipocalin-2, hepcidin, indoleamine 2,3-dioxygenase (IDO), cysteine proteases, and inducible nitric oxide synthases (iNOS). MSCs possess the potential to contribute to innate immunity by regulating the immune response. Recently, MSCs and their secreted components have been reported to improve antimicrobial activity. Bactericidal activity by MSCs and their secretomes has been shown to be mediated in part by the secretion of AMPs. Even though they were discovered more than 80 years ago, therapeutic options for persisters are restricted, and there is an urgent need for alternative treatment regimens. Hence, this review intends to critically assess the current literature on the effects of MSCs and their secretomes on persister bacteria. MSCs and their secretome-based therapies could be preferred as an up-and-coming approach to reinforce the antimicrobial efficiency in persister infections.
Collapse
Affiliation(s)
- Mesude Bicer
- Department of Bioengineering, Faculty of Life and Natural Sciences, Abdullah Gul University, Kayseri, 38080, Turkey.
| | - Ozkan Fidan
- Department of Bioengineering, Faculty of Life and Natural Sciences, Abdullah Gul University, Kayseri, 38080, Turkey
| |
Collapse
|
15
|
Das A, Patro S, Simnani FZ, Singh D, Sinha A, Kumari K, Rao PV, Singh S, Kaushik NK, Panda PK, Suar M, Verma SK. Biofilm modifiers: The disparity in paradigm of oral biofilm ecosystem. Biomed Pharmacother 2023; 164:114966. [PMID: 37269809 DOI: 10.1016/j.biopha.2023.114966] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/19/2023] [Accepted: 05/29/2023] [Indexed: 06/05/2023] Open
Abstract
A biofilm is a population of sessile microorganisms that has a distinct organized structure and characteristics like channels and projections. Good oral hygiene and reduction in the prevalence of periodontal diseases arise from minimal biofilm accumulation in the mouth, however, studies focusing on modifying the ecology of oral biofilms have not yet been consistently effective. The self-produced matrix of extracellular polymeric substances and greater antibiotic resistance make it difficult to target and eliminate biofilm infections, which lead to serious clinical consequences that are often lethal. Therefore, a better understanding is required to target and modify the ecology of biofilms in order to eradicate the infection, not only in instances of oral disorders but also in terms of nosocomial infections. The review focuses on several biofilm ecology modifiers to prevent biofilm infections, as well as the involvement of biofilm in antibiotic resistance, implants or in-dwelling device contamination, dental caries, and other periodontal disorders. It also discusses recent advances in nanotechnology that may lead to novel strategies for preventing and treating infections caused by biofilms as well as a novel outlook to infection control.
Collapse
Affiliation(s)
- Antarikshya Das
- KIIT School of Dental Sciences, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Swadheena Patro
- KIIT School of Dental Sciences, KIIT University, Bhubaneswar 751024, Odisha, India.
| | | | - Dibyangshee Singh
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Adrija Sinha
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Khushbu Kumari
- KIIT School of Dental Sciences, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Patnala Vedika Rao
- KIIT School of Medical Sciences, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Sarita Singh
- BVG Life Sciences Limited, Sagar Complex, Old Pune-Mumbai Road, Chinchwad, Pune 411034, India
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, Republic of Korea.
| | - Pritam Kumar Panda
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden.
| | - Mrutyunjay Suar
- KIIT School of Dental Sciences, KIIT University, Bhubaneswar 751024, Odisha, India.
| | - Suresh K Verma
- KIIT School of Dental Sciences, KIIT University, Bhubaneswar 751024, Odisha, India.
| |
Collapse
|
16
|
Nakamura Y, Watanabe K, Yoshioka Y, Ariyoshi W, Yamasaki R. Persister Cell Formation and Elevated lsrA and lsrC Gene Expression upon Hydrogen Peroxide Exposure in a Periodontal Pathogen Aggregatibacter actinomycetemcomitans. Microorganisms 2023; 11:1402. [PMID: 37374903 DOI: 10.3390/microorganisms11061402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
The effect of hydrogen peroxide, an antiseptic dental treatment, on Aggregatibacter actinomycetemcomitans, the main causative agent of localized invasive periodontitis, was investigated. Hydrogen peroxide treatment (0.06%, 4× minimum inhibitory concentration) resulted in the persistence and survival of approximately 0.5% of the bacterial population. The surviving bacteria did not genetically acquire hydrogen peroxide resistance but exhibited a known persister behavior. Sterilization with mitomycin C significantly reduced the number of A. actinomycetemcomitans persister survivors. RNA sequencing of hydrogen peroxide-treated A. actinomycetemcomitans showed elevated expression of Lsr family members, suggesting a strong involvement of autoinducer uptake. In this study, we found a risk of A. actinomycetemcomitans persister residual from hydrogen peroxide treatment and hypothesized associated genetic mechanisms of persister from RNA sequencing.
Collapse
Affiliation(s)
- Yohei Nakamura
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu 803-8580, Fukuoka, Japan
- Division of Developmental Stomatognathic Function Science, Department of Health Promotion, Kyushu Dental University, Kitakyushu 803-8580, Fukuoka, Japan
| | - Koji Watanabe
- Division of Developmental Stomatognathic Function Science, Department of Health Promotion, Kyushu Dental University, Kitakyushu 803-8580, Fukuoka, Japan
| | - Yoshie Yoshioka
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu 803-8580, Fukuoka, Japan
| | - Wataru Ariyoshi
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu 803-8580, Fukuoka, Japan
| | - Ryota Yamasaki
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu 803-8580, Fukuoka, Japan
- Collaborative Research Centre for Green Materials on Environmental Technology, Kyushu Institute of Technology, 1-1 Sensui-chou, Tobata-ku, Kitakyushu 804-8550, Fukuoka, Japan
| |
Collapse
|
17
|
Xiao T, Liu K, Gao Q, Chen M, Kim YS, Jin S, Ding Y, Huigens RW. Design, Synthesis, and Evaluation of Carbonate-Linked Halogenated Phenazine-Quinone Prodrugs with Improved Water-Solubility and Potent Antibacterial Profiles. ACS Infect Dis 2023; 9:899-915. [PMID: 36867688 PMCID: PMC10551733 DOI: 10.1021/acsinfecdis.2c00558] [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] [Indexed: 03/05/2023]
Abstract
Pathogenic bacteria have devastating impacts on human health as a result of acquired antibiotic resistance and innate tolerance. Every class of our current antibiotic arsenal was initially discovered as growth-inhibiting agents that target actively replicating (individual, free-floating) planktonic bacteria. Bacteria are notorious for utilizing a diversity of resistance mechanisms to overcome the action of conventional antibiotic therapies and forming surface-attached biofilm communities enriched in (non-replicating) persister cells. To address problems associated with pathogenic bacteria, our group is developing halogenated phenazine (HP) molecules that demonstrate potent antibacterial and biofilm-eradicating activities through a unique iron starvation mode of action. In this study, we designed, synthesized, and investigated a focused collection of carbonate-linked HP prodrugs bearing a quinone trigger to target the reductive cytoplasm of bacteria for bioactivation and subsequent HP release. The quinone moiety also contains a polyethylene glycol group, which dramatically enhances the water-solubility properties of the HP-quinone prodrugs reported herein. We found carbonate-linked HP-quinone prodrugs 11, 21-23 to demonstrate good linker stability, rapid release of the active HP warhead following dithiothreitol (reductive) treatment, and potent antibacterial activities against methicillin-resistant Staphylococcus aureus (MRSA), methicillin-resistant Staphylococcus epidermidis, and Enterococcus faecalis. In addition, HP-quinone prodrug 21 induced rapid iron starvation in MRSA and S. epidermidis biofilms, illustrating prodrug action within these surface-attached communities. Overall, we are highly encouraged by these findings and believe that HP prodrugs have the potential to address antibiotic resistant and tolerant bacterial infections.
Collapse
Affiliation(s)
- Tao Xiao
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Ke Liu
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Qiwen Gao
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Manyun Chen
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Young S Kim
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Shouguang Jin
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Yousong Ding
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Robert W Huigens
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
18
|
Iu HTV, Fong PM, Yam HCB, Gao P, Yan B, Lai PM, Tang VYM, Li KH, Ma CW, Ng KHK, Sze KH, Yang D, Davies J, Kao RYT. Identification of a Small Molecule Compound Active against Antibiotic-Tolerant Staphylococcus aureus by Boosting ATP Synthesis. Int J Mol Sci 2023; 24:ijms24076242. [PMID: 37047217 PMCID: PMC10094146 DOI: 10.3390/ijms24076242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/29/2023] Open
Abstract
Antibiotic tolerance poses a threat to current antimicrobial armamentarium. Bacteria at a tolerant state survive in the presence of antibiotic treatment and account for persistence, relapse and recalcitrance of infections. Antibiotic treatment failure may occur due to antibiotic tolerance. Persistent infections are difficult to treat and are often associated with poor prognosis, imposing an enormous burden on the healthcare system. Effective strategies targeting antibiotic-tolerant bacteria are therefore highly warranted. In this study, small molecule compound SA-558 was identified to be effective against Staphylococcus aureus that are tolerant to being killed by conventional antibiotics. SA-558 mediated electroneutral transport across the membrane and led to increased ATP and ROS generation, resulting in a reduction of the population of antibiotic-tolerant bacteria. In a murine chronic infection model, of which vancomycin treatment failed, we demonstrated that SA-558 alone and in combination with vancomycin caused significant reduction of MRSA abundance. Our results indicate that SA-558 monotherapy or combinatorial therapy with vancomycin is an option for managing persistent S. aureus bacteremia infection and corroborate that bacterial metabolism is an important target for counteracting antibiotic tolerance.
Collapse
|
19
|
Fiore C, Lekhan A, Bordignon S, Chierotti MR, Gobetto R, Grepioni F, Turner RJ, Braga D. Mechanochemical Preparation, Solid-State Characterization, and Antimicrobial Performance of Copper and Silver Nitrate Coordination Polymers with L- and DL-Arginine and Histidine. Int J Mol Sci 2023; 24:ijms24065180. [PMID: 36982258 PMCID: PMC10049651 DOI: 10.3390/ijms24065180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/15/2023] [Accepted: 03/01/2023] [Indexed: 03/11/2023] Open
Abstract
The antimicrobial activity of the novel coordination polymers obtained by co-crystallizing the amino acids arginine or histidine, as both enantiopure L and racemic DL forms, with the salts Cu(NO3)2 and AgNO3 has been investigated to explore the effect of chirality in the cases of enantiopure and racemic forms. The compounds [Cu·AA·(NO3)2]CPs and [Ag·AA·NO3]CPs (AA = L-Arg, DL-Arg, L-His, DL-His) were prepared by mechanochemical, slurry, and solution methods and characterized by X-ray single-crystal and powder diffraction in the cases of the copper coordination polymers, and by powder diffraction and by solid-state NMR spectroscopy in the cases of the silver compounds. The two pairs of coordination polymers, [Cu·L-Arg·(NO3)2·H2O]CP and [Cu·DL-Arg·(NO3)2·H2O]CP, and [Cu·L-Hys·(NO3)2·H2O]CP and [Cu·DL-His·(NO3)2·H2O]CP, have been shown to be isostructural in spite of the different chirality of the amino acid ligands. A similar structural analogy could be established for the silver complexes on the basis of SSNMR. The activity against the bacterial pathogens Pseudomonas aeruginosa, Escherichia coli, and Staphylococcus aureus was assessed by carrying out disk diffusion assays on lysogeny agar media showing that, while there is no significant effect arising from the use of enantiopure or chiral amino acids, the coordination polymers exert an appreciable antimicrobial activity comparable, when not superior, to that of the metal salts alone.
Collapse
Affiliation(s)
- Cecilia Fiore
- Dipartimento di Chimica “Giacomo Ciamician”, University of Bologna, Via Selmi, 2, 40126 Bologna, Italy
| | - Andrii Lekhan
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Simone Bordignon
- Dipartimento di Chimica and NIS Centre, University of Torino, Via P. Giuria, 7, 10125 Torino, Italy
| | - Michele R. Chierotti
- Dipartimento di Chimica and NIS Centre, University of Torino, Via P. Giuria, 7, 10125 Torino, Italy
| | - Roberto Gobetto
- Dipartimento di Chimica and NIS Centre, University of Torino, Via P. Giuria, 7, 10125 Torino, Italy
| | - Fabrizia Grepioni
- Dipartimento di Chimica “Giacomo Ciamician”, University of Bologna, Via Selmi, 2, 40126 Bologna, Italy
| | - Raymond J. Turner
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
- Correspondence: (R.J.T.); (D.B.)
| | - Dario Braga
- Dipartimento di Chimica “Giacomo Ciamician”, University of Bologna, Via Selmi, 2, 40126 Bologna, Italy
- Correspondence: (R.J.T.); (D.B.)
| |
Collapse
|
20
|
Siqueira FDS, Siqueira JD, Denardi LB, Moreira KS, Lima Burgo TA, de Lourenço Marques L, Machado AK, Davidson CB, Chaves OA, Anraku de Campos MM, Back DF. Antibacterial, antifungal, and anti-biofilm effects of sulfamethoxazole-complexes against pulmonary infection agents. Microb Pathog 2023; 175:105960. [PMID: 36587926 DOI: 10.1016/j.micpath.2022.105960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/22/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022]
Abstract
Antibiotic resistance associated with pulmonary infection agents has become a public health problem, being considered one of the main priorities for immediate resolution. Thus, to increase the therapeutic options in the fight against resistant microorganisms, the synthesis of molecules from pre-existing drugs has shown to be a promising alternative. In this sense, the present work reports the synthesis, characterization, and biological evaluation (against fungal and bacterial agents that cause lung infections) of potential metallodrugs based on sulfamethoxazole complexed with AuI, AgI, HgII, CdII, NiII, and CuII. The minimal inhibitory concentration (MIC) value was used to evaluate the antifungal and antibacterial properties of the compounds. In addition, it was also evaluated the antibiofilm capacity in Pseudomonas aeruginosa, through the quantification of its biomass and visualization using atomic force microscopy. For each case, molecular docking calculations were carried out to suggest the possible biological target of the assayed inorganic complexes. Our results indicated that the novel inorganic complexes are better antibacterial and antifungal than the commercial antibiotic sulfamethoxazole, highlighting the AgI-complex, which was able to inhibit the growth of microorganisms that cause lung diseases with concentrations in the 2-8 μg mL-1 range, probably at targeting dihydropteroate synthetase - a key enzyme involved in the folate synthesis. Furthermore, sulfamethoxazole complexes were able to inhibit the formation of bacterial biofilms at significantly lower concentrations than free sulfamethoxazole, probably mainly targeting the active site of LysR-type transcriptional regulator (PqsR). Overall, the present study reports preliminary results that demonstrate the derivatization of sulfamethoxazole with transition metal cations to obtain potential metallodrugs with applications as antimicrobial and antifungal against pulmonary infections, being an alternative for drug-resistant strains.
Collapse
Affiliation(s)
- Fallon Dos Santos Siqueira
- Mycobacteriology Laboratory, Graduate Program in Pharmaceutical Sciences, Department of Clinical and Toxicological Analysis, Federal University of Santa Maria, Roraima Avenue 1000, zip code:, 97105-900, Santa Maria, Brazil
| | - Josiéli Demetrio Siqueira
- Inorganic Materials Laboratory, Graduate Program in Chemistry, Department of Chemistry Federal University of Santa Maria, Roraima Avenue 1000, zip code:, 97105-900, Santa Maria, Brazil
| | - Laura Bedin Denardi
- Mycobacteriology Laboratory, Graduate Program in Pharmaceutical Sciences, Department of Clinical and Toxicological Analysis, Federal University of Santa Maria, Roraima Avenue 1000, zip code:, 97105-900, Santa Maria, Brazil
| | - Kelly Schneider Moreira
- Coulomb Electrostatic and Mechanochemical Laboratory, Graduate Program in Chemistry, Department of Chemistry, Federal University of Santa Maria, Roraima Avenue 1000, zip code:, 97105-900, Santa Maria, Brazil
| | - Thiago Augusto Lima Burgo
- Coulomb Electrostatic and Mechanochemical Laboratory, Graduate Program in Chemistry, Department of Chemistry, Federal University of Santa Maria, Roraima Avenue 1000, zip code:, 97105-900, Santa Maria, Brazil
| | - Lenice de Lourenço Marques
- Inorganic Materials Laboratory, Graduate Program in Chemistry, Department of Chemistry Federal University of Santa Maria, Roraima Avenue 1000, zip code:, 97105-900, Santa Maria, Brazil
| | - Alencar Kolinski Machado
- Laboratory of Genetics and Cell Culture, Graduate Program in Nanosciences, Franciscan University, Andradas' Street, 1614, zip code:, 97010-032, Santa Maria, Brazil
| | - Carolina Bordin Davidson
- Laboratory of Genetics and Cell Culture, Graduate Program in Nanosciences, Franciscan University, Andradas' Street, 1614, zip code:, 97010-032, Santa Maria, Brazil
| | - Otávio Augusto Chaves
- Coimbra Chemistry Center - Institute of Molecular Sciences (CQC-IMS), Faculty of Science and Technology, Department of Chemistry, University of Coimbra, Rua Larga, 3004-535, Coimbra, Portugal
| | - Marli Matiko Anraku de Campos
- Mycobacteriology Laboratory, Graduate Program in Pharmaceutical Sciences, Department of Clinical and Toxicological Analysis, Federal University of Santa Maria, Roraima Avenue 1000, zip code:, 97105-900, Santa Maria, Brazil.
| | - Davi Fernando Back
- Inorganic Materials Laboratory, Graduate Program in Chemistry, Department of Chemistry Federal University of Santa Maria, Roraima Avenue 1000, zip code:, 97105-900, Santa Maria, Brazil.
| |
Collapse
|
21
|
Pidhatika B, Widyaya VT, Nalam PC, Swasono YA, Ardhani R. Surface Modifications of High-Performance Polymer Polyetheretherketone (PEEK) to Improve Its Biological Performance in Dentistry. Polymers (Basel) 2022; 14:polym14245526. [PMID: 36559893 PMCID: PMC9787615 DOI: 10.3390/polym14245526] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/17/2022] [Accepted: 11/20/2022] [Indexed: 12/23/2022] Open
Abstract
This comprehensive review focuses on polyetheretherketone (PEEK), a synthetic thermoplastic polymer, for applications in dentistry. As a high-performance polymer, PEEK is intrinsically robust yet biocompatible, making it an ideal substitute for titanium-the current gold standard in dentistry. PEEK, however, is also inert due to its low surface energy and brings challenges when employed in dentistry. Inert PEEK often falls short of achieving a few critical requirements of clinical dental materials, such as adhesiveness, osseoconductivity, antibacterial properties, and resistance to tribocorrosion. This study aims to review these properties and explore the various surface modification strategies that enhance the performance of PEEK. Literatures searches were conducted on Google Scholar, Research Gate, and PubMed databases using PEEK, polyetheretherketone, osseointegration of PEEK, PEEK in dentistry, tribology of PEEK, surface modifications, dental applications, bonding strength, surface topography, adhesive in dentistry, and dental implant as keywords. Literature on the topics of surface modification to increase adhesiveness, tribology, and osseointegration of PEEK were included in the review. The unavailability of full texts was considered when excluding literature. Surface modifications via chemical strategies (such as sulfonation, plasma treatment, UV treatment, surface coating, surface polymerization, etc.) and/or physical approaches (such as sandblasting, laser treatment, accelerated neutral atom beam, layer-by-layer assembly, particle leaching, etc.) discussed in the literature are summarized and compared. Further, approaches such as the incorporation of bioactive materials, e.g., osteogenic agents, antibacterial agents, etc., to enhance the abovementioned desired properties are explored. This review presents surface modification as a critical and essential approach to enhance the biological performance of PEEK in dentistry by retaining its mechanical robustness.
Collapse
Affiliation(s)
- Bidhari Pidhatika
- Research Center for Polymer Technology, National Research and Innovation Agency, Republic of Indonesia PRTPL BRIN Indonesia, Serpong, Tangerang Selatan 15314, Indonesia
- Collaborative Research Center for Biomedical Scaffolds, National Research and Innovation Agency of the Republic Indonesia and Universitas Gadjah Mada, Jalan Denta No. 1, Sekip Utara, Yogyakarta 55281, Indonesia
| | - Vania Tanda Widyaya
- Research Center for Polymer Technology, National Research and Innovation Agency, Republic of Indonesia PRTPL BRIN Indonesia, Serpong, Tangerang Selatan 15314, Indonesia
| | - Prathima C. Nalam
- Department of Materials Design and Innovation, University at Buffalo, Buffalo, NY 14260-1900, USA
| | - Yogi Angga Swasono
- Research Center for Polymer Technology, National Research and Innovation Agency, Republic of Indonesia PRTPL BRIN Indonesia, Serpong, Tangerang Selatan 15314, Indonesia
| | - Retno Ardhani
- Department of Dental Biomedical Science, Faculty of Dentistry, Universitas Gadjah Mada, Jalan Denta No. 1, Sekip Utara, Yogyakarta 55281, Indonesia
- Correspondence:
| |
Collapse
|
22
|
Nemchenko UM, Sitnikova KO, Belkova NL, Grigorova EV, Voropaeva NM, Sukhоreva MV, Sukhareva ES, Savilov ED. Effects of аntimicrobials on Pseudomonas aeruginosa biofilm formation. Vavilovskii Zhurnal Genet Selektsii 2022; 26:495-501. [PMID: 36128574 PMCID: PMC9450032 DOI: 10.18699/vjgb-22-60] [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: 03/31/2022] [Revised: 06/30/2022] [Accepted: 06/30/2022] [Indexed: 11/19/2022] Open
Abstract
Pseudomonas aeruginosa is one of the most problematic pathogens in medical institutions, which may be due to the ability of this microorganism to exist in a biofilm, which increases its resistance to antimicrobials, as well as its prevalence and survival ability in the external environment. This work aimed to evaluate the antimicrobial susceptibility of P. aeruginosa strains in planktonic and biofilm forms. We studied 20 strains of P. aeruginosa collected during 2018-2021 by specialists from the Laboratory of Microbiome and Microecology of the Scientific Centre for Family Health and Human Reproduction Problems. The identification of strains was carried out using test systems for differentiating gram-negative non-fermenting bacteria (NEFERMtest 24 Erba Lachema s.r.o., Czech Republic), and confirmed by mass spectrometric analysis and 16S rRNA gene sequencing. Antimicrobial activity was assessed by the degree of inhibition of cell growth in planktonic and biofilm forms (on a flat-bottomed 96-well plastic immunological plate). All clinical isolates of P. aeruginosa were biofilm formers, 47.6 % of the isolates were weak biofilm formers, and 52.4 % of the isolates were moderate biofilm formers. Planktonic cells and the forming biofilm of the tested P. aeruginosa strains were carbapenems-resistant. Biofilm formation was suppressed in more than 90 % of cases by the agents of the cephalosporin and aminoglycoside groups. Antimicrobial susceptibility of P. aeruginosa strains in the formed biofilm was significantly lower (p < 0.05). Carbapenems and cephalosporins did not affect the mature biofilms of the tested P. aeruginosa strains in more than 60 % of cases. Only non-beta-lactam antibiotics (ciprofloxacin and amikacin) suppressed the growth of planktonic cells and destroyed the mature biofilm. The revealed differences in the effect of the tested antimicrobials on the P. aeruginosa strains biofilms correlate with resistance to a number of antibiotics. To prevent biofilm formation in the hospital strains of P. aeruginosa, the use of ceftazidime may be recommended, and antimicrobials such as ciprofloxacin and amikacin may be used to affect mature biofilms of P. aeruginosa.
Collapse
Affiliation(s)
- U M Nemchenko
- Scientific Сentre for Family Health and Human Reproduction Problems, Irkutsk, Russia
| | - K O Sitnikova
- Scientific Сentre for Family Health and Human Reproduction Problems, Irkutsk, Russia
| | - N L Belkova
- Scientific Сentre for Family Health and Human Reproduction Problems, Irkutsk, Russia
| | - E V Grigorova
- Scientific Сentre for Family Health and Human Reproduction Problems, Irkutsk, Russia
| | - N M Voropaeva
- Scientific Сentre for Family Health and Human Reproduction Problems, Irkutsk, Russia
| | - M V Sukhоreva
- City Ivano-Matreninskaya Children's Clinical Hospital, Irkutsk, Russia
| | - E S Sukhareva
- City Ivano-Matreninskaya Children's Clinical Hospital, Irkutsk, Russia
| | - E D Savilov
- Scientific Сentre for Family Health and Human Reproduction Problems, Irkutsk, Russia Irkutsk State Medical Academy of Postgraduate Education - Branch Campus of the Russian Medical Academy of Continuing Professional Education of the Ministry of Healthcare of the Russian Federation, Irkutsk, Russia
| |
Collapse
|
23
|
Antimicrobial tolerance and its role in the development of resistance: Lessons from enterococci. Adv Microb Physiol 2022; 81:25-65. [PMID: 36167442 DOI: 10.1016/bs.ampbs.2022.06.004] [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] [Indexed: 01/27/2023]
Abstract
Bacteria have developed resistance against every antimicrobial in clinical use at an alarming rate. There is a critical need for more effective use of antimicrobials to both extend their shelf life and prevent resistance from arising. Significantly, antimicrobial tolerance, i.e., the ability to survive but not proliferate during antimicrobial exposure, has been shown to precede the development of bona fide antimicrobial resistance (AMR), sparking a renewed and rapidly increasing interest in this field. As a consequence, problematic infections for the first time are now being investigated for antimicrobial tolerance, with increasing reports demonstrating in-host evolution of antimicrobial tolerance. Tolerance has been identified in a wide array of bacterial species to all bactericidal antimicrobials. Of particular interest are enterococci, which contain the opportunistic bacterial pathogens Enterococcus faecalis and Enterococcus faecium. Enterococci are one of the leading causes of hospital-acquired infection and possess intrinsic tolerance to a number of antimicrobial classes. Persistence of these infections in the clinic is of growing concern, particularly for the immunocompromised. Here, we review current known mechanisms of antimicrobial tolerance, and include an in-depth analysis of those identified in enterococci with implications for both the development and prevention of AMR.
Collapse
|
24
|
Abstract
Mechanisms of evolution and evolution of antibiotic resistance are both fundamental and world health problems. Stress-induced mutagenesis defines mechanisms of mutagenesis upregulated by stress responses, which drive adaptation when cells are maladapted to their environments—when stressed. Work in mutagenesis induced by antibiotics had produced tantalizing clues but not coherent mechanisms. We review recent advances in antibiotic-induced mutagenesis that integrate how reactive oxygen species (ROS), the SOS and general stress responses, and multichromosome cells orchestrate a stress response-induced switch from high-fidelity to mutagenic repair of DNA breaks. Moreover, while sibling cells stay stable, a mutable “gambler” cell subpopulation is induced by differentially generated ROS, which signal the general stress response. We discuss other evolvable subpopulations and consider diverse evolution-promoting molecules as potential targets for drugs to slow evolution of antibiotic resistance, cross-resistance, and immune evasion. An FDA-approved drug exemplifies “stealth” evolution-slowing drugs that avoid selecting resistance to themselves or antibiotics.
Collapse
|
25
|
Liu J, Wei Q, Wang Z, Sun X, He QY. Proteomic Study of the Adaptive Mechanism of Ciprofloxacin-Resistant Staphylococcus aureus to the Host Environment. J Proteome Res 2022; 21:1537-1547. [PMID: 35594371 DOI: 10.1021/acs.jproteome.2c00140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Antibiotic-resistant bacteria can escape host immune killing and settle in the host to form persistent infections. In this study we investigated the adaptive mechanism of resistant Staphylococcus aureus to the host environment by data-independent acquisition-based quantitative proteomics and functional validation. The growth curve and minimum inhibitory concentration (MIC) indicated that ciprofloxacin-resistant (Cip-R) S. aureus showed a survival advantage over sensitive strains. Cip-R also exhibited a stronger invasion and biofilm formation ability than sensitive bacteria. Cip-R stimulation resulted in the improved production of inflammatory factors of the host cells. Proteomics study combined with biochemical validations showed that Cip-R obtained adaptability to the host via upregulation of the tricarboxylic acid cycle (TCA cycle) and downregulation of ribosome metabolism and protein folding to maintain energy to support Cip-R's survival. Thus, this study will help us to further explain the growth strategy of resistant bacteria to adapt to the host environment, and provide important information for the development of new antibacterial drugs.
Collapse
Affiliation(s)
- Jiajia Liu
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Qiuxia Wei
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Zhen Wang
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xuesong Sun
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Qing-Yu He
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
26
|
Kaur H, Kaur A, Soni SK, Rishi P. Microbially-derived cocktail of carbohydrases as an anti-biofouling agents: a 'green approach'. BIOFOULING 2022; 38:455-481. [PMID: 35673761 DOI: 10.1080/08927014.2022.2085566] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 05/12/2022] [Accepted: 05/30/2022] [Indexed: 06/15/2023]
Abstract
Enzymes, also known as biocatalysts, display vital properties like high substrate specificity, an eco-friendly nature, low energy inputs, and cost-effectiveness. Among their numerous known applications, enzymes that can target biofilms or their components are increasingly being investigated for their anti-biofouling action, particularly in healthcare, food manufacturing units and environmental applications. Enzymes can target biofilms at different levels like during the attachment of microorganisms, formation of exopolymeric substances (EPS), and their disruption thereafter. In this regard, a consortium of carbohydrases that can target heterogeneous polysaccharides present in the EPS matrix may provide an effective alternative to conventional chemical anti-biofouling methods. Further, for complete annihilation of biofilms, enzymes can be used alone or in conjunction with other antimicrobial agents. Enzymes hold the promise to replace the conventional methods with greener, more economical, and more efficient alternatives. The present article explores the potential and future perspectives of using carbohydrases as effective anti-biofilm agents.
Collapse
Affiliation(s)
- Harmanpreet Kaur
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Arashdeep Kaur
- Department of Microbiology, Panjab University, Chandigarh, India
| | | | - Praveen Rishi
- Department of Microbiology, Panjab University, Chandigarh, India
| |
Collapse
|
27
|
Fleming D, Niese B, Redman W, Vanderpool E, Gordon V, Rumbaugh KP. Contribution of Pseudomonas aeruginosa Exopolysaccharides Pel and Psl to Wound Infections. Front Cell Infect Microbiol 2022; 12:835754. [PMID: 35463635 PMCID: PMC9021892 DOI: 10.3389/fcimb.2022.835754] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/01/2022] [Indexed: 11/24/2022] Open
Abstract
Biofilms are the cause of most chronic bacterial infections. Living within the biofilm matrix, which is made of extracellular substances, including polysaccharides, proteins, eDNA, lipids and other molecules, provides microorganisms protection from antimicrobials and the host immune response. Exopolysaccharides are major structural components of bacterial biofilms and are thought to be vital to numerous aspects of biofilm formation and persistence, including adherence to surfaces, coherence with other biofilm-associated cells, mechanical stability, protection against desiccation, binding of enzymes, and nutrient acquisition and storage, as well as protection against antimicrobials, host immune cells and molecules, and environmental stressors. However, the contribution of specific exopolysaccharide types to the pathogenesis of biofilm infection is not well understood. In this study we examined whether the absence of the two main exopolysaccharides produced by the biofilm former Pseudomonas aeruginosa would affect wound infection in a mouse model. Using P. aeruginosa mutants that do not produce the exopolysaccharides Pel and/or Psl we observed that the severity of wound infections was not grossly affected; both the bacterial load in the wounds and the wound closure rates were unchanged. However, the size and spatial distribution of biofilm aggregates in the wound tissue were significantly different when Pel and Psl were not produced, and the ability of the mutants to survive antibiotic treatment was also impaired. Taken together, our data suggest that while the production of Pel and Psl do not appear to affect P. aeruginosa pathogenesis in mouse wound infections, they may have an important implication for bacterial persistence in vivo.
Collapse
Affiliation(s)
- Derek Fleming
- Department of Surgery, Texas Tech University Health Sciences, Lubbock, TX, United States
| | - Brandon Niese
- Department of Physics, Center for Nonlinear Dynamics, The University of Texas at Austin, Austin TX, United States
| | - Whitni Redman
- Department of Surgery, Texas Tech University Health Sciences, Lubbock, TX, United States
| | - Emily Vanderpool
- Department of Surgery, Texas Tech University Health Sciences, Lubbock, TX, United States
| | - Vernita Gordon
- Department of Physics, Center for Nonlinear Dynamics, The University of Texas at Austin, Austin TX, United States
- Interdisciplinary Life Sciences Graduate Programs, LaMontagne Center for Infectious Disease, The University of Texas at Austin, Austin, TX, United States
| | - Kendra P. Rumbaugh
- Department of Surgery, Texas Tech University Health Sciences, Lubbock, TX, United States
- Burn Center for Research Excellence, Texas Tech University Health Sciences, Lubbock, TX, United States
| |
Collapse
|
28
|
Kay W, Hunt C, Nehring L, Barnum B, Ashton N, Williams D. Biofilm Growth on Simulated Fracture Fixation Plates Using a Customized CDC Biofilm Reactor for a Sheep Model of Biofilm-Related Infection. Microorganisms 2022; 10:microorganisms10040759. [PMID: 35456808 PMCID: PMC9031587 DOI: 10.3390/microorganisms10040759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 02/04/2023] Open
Abstract
Most animal models of infection utilize planktonic bacteria as initial inocula. However, this may not accurately mimic scenarios where bacteria in the biofilm phenotype contaminate a site at the point of injury. We developed a modified CDC biofilm reactor in which biofilms can be grown on the surface of simulated fracture fixation plates. Multiple reactor runs were performed and demonstrated that monomicrobial biofilms of a clinical strain of methicillin-resistant Staphylococcus aureus, S. aureus ATCC 6538, and Pseudomonas aeruginosa ATCC 27853 consistently developed on fixation plates. We also identified a method by which to successfully grow polymicrobial biofilms of S. aureus ATCC 6538 and P. aeruginosa ATCC 27853 on fixation plates. This customized reactor can be used to grow biofilms on simulated fracture fixation plates that can be inoculated in animal models of biofilm implant-related infection that, for example, mimic open fracture scenarios. The reactor provides a method for growing biofilms that can be used as initial inocula and potentially improve the testing and development of antibiofilm technologies.
Collapse
Affiliation(s)
- Walker Kay
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84108, USA; (W.K.); (C.H.); (L.N.); (N.A.)
| | - Connor Hunt
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84108, USA; (W.K.); (C.H.); (L.N.); (N.A.)
| | - Lisa Nehring
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84108, USA; (W.K.); (C.H.); (L.N.); (N.A.)
| | - Brian Barnum
- Purgo Scientific, LLC, South Jordan, UT 84095, USA;
| | - Nicholas Ashton
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84108, USA; (W.K.); (C.H.); (L.N.); (N.A.)
- Purgo Scientific, LLC, South Jordan, UT 84095, USA;
| | - Dustin Williams
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84108, USA; (W.K.); (C.H.); (L.N.); (N.A.)
- Purgo Scientific, LLC, South Jordan, UT 84095, USA;
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
- Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
- Department of Physical Medicine and Rehabilitation, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Correspondence:
| |
Collapse
|
29
|
Liu K, Brivio M, Xiao T, Norwood VM, Kim YS, Jin S, Papagni A, Vaghi L, Huigens RW. Modular Synthetic Routes to Fluorine-Containing Halogenated Phenazine and Acridine Agents That Induce Rapid Iron Starvation in Methicillin-Resistant Staphylococcus aureus Biofilms. ACS Infect Dis 2022; 8:280-295. [PMID: 35089005 PMCID: PMC9004446 DOI: 10.1021/acsinfecdis.1c00402] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
During infection, bacteria use an arsenal of resistance mechanisms to negate antibiotic therapies. In addition, pathogenic bacteria form surface-attached biofilms bearing enriched populations of metabolically dormant persister cells. Bacteria develop resistance in response to antibiotic insults; however, nonreplicating biofilms are innately tolerant to all classes of antibiotics. As such, molecules that can eradicate antibiotic-resistant and antibiotic-tolerant bacteria are of importance. Here, we report modular synthetic routes to fluorine-containing halogenated phenazine (HP) and halogenated acridine (HA) agents with potent antibacterial and biofilm-killing activities. Nine fluorinated phenazines were rapidly accessed through a synthetic strategy involving (1) oxidation of fluorinated anilines to azobenzene intermediates, (2) SNAr with 2-methoxyaniline, and (3) cyclization to phenazines upon treatment with trifluoroacetic acid. Five structurally related acridine heterocycles were synthesized using SNAr and Buchwald-Hartwig approaches. From this focused collection, phenazines 5g, 5h, 5i, and acridine 9c demonstrated potent antibacterial activities against Gram-positive pathogens (MIC = 0.04-0.78 μM). Additionally, 5g and 9c eradicated Staphylococcus aureus, Staphylococcus epidermidis and Enterococcus faecalis biofilms with excellent potency (5g, MBEC = 4.69-6.25 μM; 9c, MBEC = 4.69-50 μM). Using real-time quantitative polymerase chain reaction (RT-qPCR), 5g, 5h, 5i, and 9c rapidly induce the transcription of iron uptake biomarkers isdB and sbnC in methicillin-resistant S. aureus (MRSA) biofilms, and we conclude that these agents operate through iron starvation. Overall, fluorinated phenazine and acridine agents could lead to ground-breaking advances in the treatment of challenging bacterial infections.
Collapse
Affiliation(s)
- Ke Liu
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Massimiliano Brivio
- Department of Materials Science, University of Milano-Bicocca, 20125 Milano, Italy
| | - Tao Xiao
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Verrill M. Norwood
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Young S. Kim
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Shouguang Jin
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Antonio Papagni
- Department of Materials Science, University of Milano-Bicocca, 20125 Milano, Italy
| | - Luca Vaghi
- Department of Materials Science, University of Milano-Bicocca, 20125 Milano, Italy
| | - Robert W. Huigens
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
30
|
Schrank CL, Wilt IK, Monteagudo Ortiz C, Haney BA, Wuest WM. Using membrane perturbing small molecules to target chronic persistent infections. RSC Med Chem 2021; 12:1312-1324. [PMID: 34458737 PMCID: PMC8372208 DOI: 10.1039/d1md00151e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022] Open
Abstract
After antibiotic treatment, a subpopulation of bacteria often remains and can lead to recalcitrant infections. This subpopulation, referred to as persisters, evades antibiotic treatment through numerous mechanisms such as decreased uptake of small molecules and slowed growth. Membrane perturbing small molecules have been shown to eradicate persisters as well as render these populations susceptible to antibiotic treatment. Chemotype similarities have emerged suggesting amphiphilic heteroaromatic compounds possess ideal properties to increase membrane fluidity and such molecules warrant further investigation as effective agents or potentiators against persister cells.
Collapse
Affiliation(s)
| | - Ingrid K Wilt
- Department of Chemistry Emory University Atlanta GA 30322 USA
| | | | | | - William M Wuest
- Department of Chemistry Emory University Atlanta GA 30322 USA
- Emory Antibiotic Resistance Center, Emory University School of Medicine Atlanta GA 30322 USA
| |
Collapse
|
31
|
Huigens Iii RW, Yang H, Liu K, Kim YS, Jin S. An ether-linked halogenated phenazine-quinone prodrug model for antibacterial applications. Org Biomol Chem 2021; 19:6603-6608. [PMID: 34286808 PMCID: PMC8525319 DOI: 10.1039/d1ob01107c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Antibiotic-resistant infections present significant challenges to patients. As a result, there is considerable need for new antibacterial therapies that eradicate pathogenic bacteria through non-conventional mechanisms. Our group has identified a series of halogenated phenazine (HP) agents that induce rapid iron starvation that leads to potent killing of methicillin-resistant Staphylococcus aureus biofilms. Here, we report the design, chemical synthesis and microbiological assessment of a HP-quinone ether prodrug model aimed to (1) eliminate general (off-target) iron chelation, and (2) release an active HP agent through the bioreduction of a quinone trigger. Here, we demonstrate prodrug analogue HP-29-Q to have a stable ether linkage that enables HP release and moderate to good antibacterial activities against lab strains and multi-drug resistant clinical isolates.
Collapse
Affiliation(s)
- Robert W Huigens Iii
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, USA.
| | - Hongfen Yang
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, USA.
| | - Ke Liu
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, USA.
| | - Young S Kim
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - Shouguang Jin
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| |
Collapse
|
32
|
Leonov PS, Flores-Alsina X, Gernaey KV, Sternberg C. Microbial biofilms in biorefinery - Towards a sustainable production of low-value bulk chemicals and fuels. Biotechnol Adv 2021; 50:107766. [PMID: 33965529 DOI: 10.1016/j.biotechadv.2021.107766] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 04/11/2021] [Accepted: 05/04/2021] [Indexed: 12/14/2022]
Abstract
Harnessing the potential of biocatalytic conversion of renewable biomass into value-added products is still hampered by unfavorable process economics. This has promoted the use of biofilms as an alternative to overcome the limitations of traditional planktonic systems. In this paper, the benefits and challenges of biofilm fermentations are reviewed with a focus on the production of low-value bulk chemicals and fuels from waste biomass. Our study demonstrates that biofilm fermentations can potentially improve productivities and product yields by increasing biomass retention and allowing for continuous operation at high dilution rates. Furthermore, we show that biofilms can tolerate hazardous environments, which improve the conversion of crude biomass under substrate and product inhibitory conditions. Additionally, we present examples for the improved conversion of pure and crude substrates into bulk chemicals by mixed microbial biofilms, which can benefit from microenvironments in biofilms for synergistic multi-species reactions, and improved resistance to contaminants. Finally, we suggest the use of mathematical models as useful tools to supplement experimental insights related to the effects of physico-chemical and biological phenomena on the process. Major challenges for biofilm fermentations arise from inconsistent fermentation performance, slow reactor start-up, biofilm carrier costs and carrier clogging, insufficient biofilm monitoring and process control, challenges in reactor sterilization and scale-up, and issues in recovering dilute products. The key to a successful commercialization of the technology is likely going to be an interdisciplinary approach. Crucial research areas might include genetic engineering combined with the development of specialized biofilm reactors, biofilm carrier development, in-situ biofilm monitoring, model-based process control, mixed microbial biofilm technology, development of suitable biofilm reactor scale-up criteria, and in-situ product recovery.
Collapse
Affiliation(s)
- Pascal S Leonov
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Building 221, 2800 Kgs. Lyngby, Denmark; Department of Chemical and Biochemical Engineering, Technical University of Denmark, Søltofts Plads, Building 228 A, 2800 Kgs. Lyngby, Denmark
| | - Xavier Flores-Alsina
- Department of Chemical and Biochemical Engineering, Technical University of Denmark, Søltofts Plads, Building 228 A, 2800 Kgs. Lyngby, Denmark
| | - Krist V Gernaey
- Department of Chemical and Biochemical Engineering, Technical University of Denmark, Søltofts Plads, Building 228 A, 2800 Kgs. Lyngby, Denmark
| | - Claus Sternberg
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Building 221, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
33
|
Bhardwaj S, Bhatia S, Singh S, Franco Jr F. Growing emergence of drug-resistant Pseudomonas aeruginosa and attenuation of its virulence using quorum sensing inhibitors: A critical review. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:699-719. [PMID: 34630947 PMCID: PMC8487598 DOI: 10.22038/ijbms.2021.49151.11254] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 11/24/2020] [Indexed: 12/25/2022]
Abstract
A perilous increase in the number of bacterial infections has led to developing throngs of antibiotics for increasing the quality and expectancy of life. Pseudomonas aeruginosa is becoming resistant to all known conventional antimicrobial agents thereby posing a deadly threat to the human population. Nowadays, targeting virulence traits of infectious agents is an alternative approach to antimicrobials that is gaining much popularity to fight antimicrobial resistance. Quorum sensing (QS) involves interspecies communication via a chemical signaling pathway. Under this mechanism, cells work in a concerted manner, communicate with each other with the help of signaling molecules called auto-inducers (AI). The virulence of these strains is driven by genes, whose expression is regulated by AI, which in turn acts as transcriptional activators. Moreover, the problem of antibiotic-resistance in case of infections caused by P. aeruginosa becomes more alarming among immune-compromised patients, where the infectious agents easily take over the cellular machinery of the host while hidden in the QS mediated biofilms. Inhibition of the QS circuit of P. aeruginosa by targeting various signaling pathways such as LasR, RhlR, Pqs, and QScR transcriptional proteins will help in blocking downstream signal transducers which could result in reducing the bacterial virulence. The anti-virulence agent does not pose an immediate selective pressure on growing bacterium and thus reduces the pathogenicity without harming the target species. Here, we review exclusively, the growing emergence of multi-drug resistant (MDR) P. aeruginosa and the critical literature survey of QS inhibitors with their potential application of blocking P. aeruginosa infections.
Collapse
Affiliation(s)
- Snigdha Bhardwaj
- Department of Pharmaceutical Science, SHALOM Institute of Health and Allied Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences (SHUATS), Naini, Prayagraj, India
| | - Sonam Bhatia
- Department of Pharmaceutical Science, SHALOM Institute of Health and Allied Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences (SHUATS), Naini, Prayagraj, India
| | - Shaminder Singh
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad - 121 001, Haryana, India
| | - Francisco Franco Jr
- Department of Chemistry, De La Salle University, Manila, Metro Manila, Philippines
| |
Collapse
|
34
|
Hossain T, Deter HS, Peters EJ, Butzin NC. Antibiotic tolerance, persistence, and resistance of the evolved minimal cell, Mycoplasma mycoides JCVI-Syn3B. iScience 2021; 24:102391. [PMID: 33997676 PMCID: PMC8091054 DOI: 10.1016/j.isci.2021.102391] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 02/01/2021] [Accepted: 03/31/2021] [Indexed: 12/22/2022] Open
Abstract
Antibiotic resistance is a growing problem, but bacteria can evade antibiotic treatment via tolerance and persistence. Antibiotic persisters are a small subpopulation of bacteria that tolerate antibiotics due to a physiologically dormant state. Hence, persistence is considered a major contributor to the evolution of antibiotic-resistant and relapsing infections. Here, we used the synthetically developed minimal cell Mycoplasma mycoides JCVI-Syn3B to examine essential mechanisms of antibiotic survival. The minimal cell contains only 473 genes, and most genes are essential. Its reduced complexity helps to reveal hidden phenomenon and fundamental biological principles can be explored because of less redundancy and feedback between systems compared to natural cells. We found that Syn3B evolves antibiotic resistance to different types of antibiotics expeditiously. The minimal cell also tolerates and persists against multiple antibiotics. It contains a few already identified persister-related genes, although lacking many systems previously linked to persistence (e.g. toxin-antitoxin systems, ribosome hibernation genes).
Collapse
Affiliation(s)
- Tahmina Hossain
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD 57006, USA
| | - Heather S. Deter
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Eliza J. Peters
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD 57006, USA
| | - Nicholas C. Butzin
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD 57006, USA
| |
Collapse
|
35
|
Sang S, Guo G, Yu J, Zhang X. Antibacterial application of gentamicin-silk protein coating with smart release function on titanium, polyethylene, and Al 2O 3 materials. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 124:112069. [PMID: 33947562 DOI: 10.1016/j.msec.2021.112069] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/02/2021] [Accepted: 03/16/2021] [Indexed: 01/12/2023]
Abstract
Peri-implant infection after hip and knee arthroplasty is a common and serious complication. Titanium (Ti), polyethylene (PE), and Al2O3 materials used as joint prosthesis materials have good biocompatibility and mechanical strength but no antibacterial effect. This study aimed to provide a theoretical basis for the design and manufacture of joint prosthesis materials with antibacterial effect. We applied a coating of gentamicin-silk protein (GS-Silk) on the surface of these materials. We characterized the Ti, PE, and Al2O3 materials coated with GS-Silk (experimental group) and performed in vivo and in vitro experiments to test antibacterial activity. Scanning electron microscopy confirmed successful GS-Silk coating, and infrared spectroscopy confirmed successful loading of gentamicin onto the three materials. Nanoscratch test proved that the GS-Silk coating is relatively reliable on the surface of these three materials. The antibacterial effect of the coating in vitro and in vivo was verified by performing bacteriostatic ring test in vitro, bacterial adhesion test, and subendothelial implant infection test. We demonstrated that GS-Silk coating can effectively load gentamicin onto Ti, PE, and Al2O3 materials and change the gentamicin release rate with a change in the solution pH to achieve intelligent release. The GS-Silk coating is relatively reliable on the surface of these three materials. Ti, PE, and Al2O3 materials coated with GS-Silk have good antibacterial ability, both in vivo and in vitro.
Collapse
Affiliation(s)
- Shang Sang
- Department of Orthopaedics, Shanghai Jiao Tong University, Affiliated Sixth People's Hospital, Shanghai, China
| | - Geyong Guo
- Department of Orthopaedics, Shanghai Jiao Tong University, Affiliated Sixth People's Hospital, Shanghai, China
| | - Jinlong Yu
- Department of Orthopaedics, Shanghai Jiao Tong University, Affiliated Sixth People's Hospital, Shanghai, China
| | - Xianlong Zhang
- Department of Orthopaedics, Shanghai Jiao Tong University, Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
36
|
Abstract
Introduction: As a result of progress in medical care, a huge number of medical devices are used in the treatment of human diseases. In turn, biofilm-related infection has become a growing threat due to the tolerance of biofilms to antimicrobials, a problem magnified by the development of antimicrobial resistance worldwide. As a result, successful treatment of biofilm-disease using only antimicrobials is problematic.Areas covered: We summarize some alternative approaches to classic antimicrobials for the treatment of biofilm disease. This review is not intended to be exhaustive but to give a clinical picture of alternatives to antimicrobial agents to manage biofilm disease. We highlight those strategies that may be closer to application in clinical practice.Expert opinion: There are a number of outstanding challenges in the development of novel antibiofilm therapies. Screening for effective antibiofilm compounds requires models relevant to all clinical scenarios. Although in vitro research of anti-biofilm strategies has progressed significantly over the past decade, there is a lack of in vivo research. In addition, the complexity of biofilm biology makes it difficult to develop a compound that is likely to provide the single 'magic bullet'. The multifaceted nature of biofilms imposes the need for multi-targeted or combinatorial therapies.
Collapse
Affiliation(s)
- Jose L Del Pozo
- Infectious Diseases Division, Clínica Universidad De Navarra, Pamplona, Spain.,Department of Microbiology, Clínica Universidad De Navarra, Pamplona, Spain.,Laboratory of Microbial Biofilms, Clínica Universidad De Navarra, Pamplona, Spain
| |
Collapse
|
37
|
Yang H, Kundra S, Chojnacki M, Liu K, Fuse MA, Abouelhassan Y, Kallifidas D, Zhang P, Huang G, Jin S, Ding Y, Luesch H, Rohde KH, Dunman PM, Lemos JA, Huigens RW. A Modular Synthetic Route Involving N-Aryl-2-nitrosoaniline Intermediates Leads to a New Series of 3-Substituted Halogenated Phenazine Antibacterial Agents. J Med Chem 2021; 64:7275-7295. [PMID: 33881312 DOI: 10.1021/acs.jmedchem.1c00168] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Pathogenic bacteria demonstrate incredible abilities to evade conventional antibiotics through the development of resistance and formation of dormant, surface-attached biofilms. Therefore, agents that target and eradicate planktonic and biofilm bacteria are of significant interest. We explored a new series of halogenated phenazines (HP) through the use of N-aryl-2-nitrosoaniline synthetic intermediates that enabled functionalization of the 3-position of this scaffold. Several HPs demonstrated potent antibacterial and biofilm-killing activities (e.g., HP 29, against methicillin-resistant Staphylococcus aureus: MIC = 0.075 μM; MBEC = 2.35 μM), and transcriptional analysis revealed that HPs 3, 28, and 29 induce rapid iron starvation in MRSA biofilms. Several HPs demonstrated excellent activities against Mycobacterium tuberculosis (HP 34, MIC = 0.80 μM against CDC1551). This work established new SAR insights, and HP 29 demonstrated efficacy in dorsal wound infection models in mice. Encouraged by these findings, we believe that HPs could lead to significant advances in the treatment of challenging infections.
Collapse
Affiliation(s)
- Hongfen Yang
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Shivani Kundra
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida 32610, United States
| | - Michaelle Chojnacki
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York 14642, United States
| | - Ke Liu
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Marisa A Fuse
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida 32827, United States
| | - Yasmeen Abouelhassan
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Dimitris Kallifidas
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Peilan Zhang
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Guangtao Huang
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville Florida 32610, United States
| | - Shouguang Jin
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville Florida 32610, United States
| | - Yousong Ding
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Hendrik Luesch
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Kyle H Rohde
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida 32827, United States
| | - Paul M Dunman
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York 14642, United States
| | - José A Lemos
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida 32610, United States
| | - Robert W Huigens
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
38
|
Kligman S, Ren Z, Chung CH, Perillo MA, Chang YC, Koo H, Zheng Z, Li C. The Impact of Dental Implant Surface Modifications on Osseointegration and Biofilm Formation. J Clin Med 2021; 10:1641. [PMID: 33921531 PMCID: PMC8070594 DOI: 10.3390/jcm10081641] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/26/2021] [Accepted: 04/06/2021] [Indexed: 12/11/2022] Open
Abstract
Implant surface design has evolved to meet oral rehabilitation challenges in both healthy and compromised bone. For example, to conquer the most common dental implant-related complications, peri-implantitis, and subsequent implant loss, implant surfaces have been modified to introduce desired properties to a dental implant and thus increase the implant success rate and expand their indications. Until now, a diversity of implant surface modifications, including different physical, chemical, and biological techniques, have been applied to a broad range of materials, such as titanium, zirconia, and polyether ether ketone, to achieve these goals. Ideal modifications enhance the interaction between the implant's surface and its surrounding bone which will facilitate osseointegration while minimizing the bacterial colonization to reduce the risk of biofilm formation. This review article aims to comprehensively discuss currently available implant surface modifications commonly used in implantology in terms of their impact on osseointegration and biofilm formation, which is critical for clinicians to choose the most suitable materials to improve the success and survival of implantation.
Collapse
Affiliation(s)
- Stefanie Kligman
- School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Zhi Ren
- Biofilm Research Laboratories, Department of Orthodontics, Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (Z.R.); (H.K.)
| | - Chun-Hsi Chung
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (C.-H.C.); (M.A.P.)
| | - Michael Angelo Perillo
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (C.-H.C.); (M.A.P.)
| | - Yu-Cheng Chang
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Hyun Koo
- Biofilm Research Laboratories, Department of Orthodontics, Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (Z.R.); (H.K.)
- Center for Innovation & Precision Dentistry, School of Dental Medicine and School of Engineering & Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhong Zheng
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chenshuang Li
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (C.-H.C.); (M.A.P.)
| |
Collapse
|
39
|
Wenzel M, Dekker MP, Wang B, Burggraaf MJ, Bitter W, van Weering JRT, Hamoen LW. A flat embedding method for transmission electron microscopy reveals an unknown mechanism of tetracycline. Commun Biol 2021; 4:306. [PMID: 33686188 PMCID: PMC7940657 DOI: 10.1038/s42003-021-01809-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/05/2021] [Indexed: 12/20/2022] Open
Abstract
Transmission electron microscopy of cell sample sections is a popular technique in microbiology. Currently, ultrathin sectioning is done on resin-embedded cell pellets, which consumes milli- to deciliters of culture and results in sections of randomly orientated cells. This is problematic for rod-shaped bacteria and often precludes large-scale quantification of morphological phenotypes due to the lack of sufficient numbers of longitudinally cut cells. Here we report a flat embedding method that enables observation of thousands of longitudinally cut cells per single section and only requires microliter culture volumes. We successfully applied this technique to Bacillus subtilis, Escherichia coli, Mycobacterium bovis, and Acholeplasma laidlawii. To assess the potential of the technique to quantify morphological phenotypes, we monitored antibiotic-induced changes in B. subtilis cells. Surprisingly, we found that the ribosome inhibitor tetracycline causes membrane deformations. Further investigations showed that tetracycline disturbs membrane organization and localization of the peripheral membrane proteins MinD, MinC, and MreB. These observations are not the result of ribosome inhibition but constitute a secondary antibacterial activity of tetracycline that so far has defied discovery.
Collapse
Affiliation(s)
- Michaela Wenzel
- Bacterial Cell Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH, Amsterdam, The Netherlands.
- Department of Medical Microbiology and Infection Control, Amsterdam University Medical Centers - Location VUMC, 1081 HZ, Amsterdam, The Netherlands.
- Chemical Biology, Department for Biology and Biological Engineering, Chalmers University of Technology, 412 96, Gothenburg, Sweden.
| | - Marien P Dekker
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Neuroscience Campus Amsterdam, Amsterdam University Medical Centers - Location VUMC, 1081 HZ, Amsterdam, The Netherlands
| | - Biwen Wang
- Bacterial Cell Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH, Amsterdam, The Netherlands
| | - Maroeska J Burggraaf
- Department of Medical Microbiology and Infection Control, Amsterdam University Medical Centers - Location VUMC, 1081 HZ, Amsterdam, The Netherlands
| | - Wilbert Bitter
- Department of Medical Microbiology and Infection Control, Amsterdam University Medical Centers - Location VUMC, 1081 HZ, Amsterdam, The Netherlands
- Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines, and Systems, Faculty of Science, Vrije Universiteit Amsterdam, 1081 HZ, Amsterdam, The Netherlands
| | - Jan R T van Weering
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Neuroscience Campus Amsterdam, Amsterdam University Medical Centers - Location VUMC, 1081 HZ, Amsterdam, The Netherlands.
| | - Leendert W Hamoen
- Bacterial Cell Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH, Amsterdam, The Netherlands
| |
Collapse
|
40
|
Shin J, Choe D, Ransegnola B, Hong H, Onyekwere I, Cross T, Shi Q, Cho B, Westblade LF, Brito IL, Dörr T. A multifaceted cellular damage repair and prevention pathway promotes high-level tolerance to β-lactam antibiotics. EMBO Rep 2021; 22:e51790. [PMID: 33463026 PMCID: PMC7857431 DOI: 10.15252/embr.202051790] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/17/2020] [Accepted: 12/02/2020] [Indexed: 12/27/2022] Open
Abstract
Bactericidal antibiotics are powerful agents due to their ability to convert essential bacterial functions into lethal processes. However, many important bacterial pathogens are remarkably tolerant against bactericidal antibiotics due to inducible damage repair responses. The cell wall damage response two-component system VxrAB of the gastrointestinal pathogen Vibrio cholerae promotes high-level β-lactam tolerance and controls a gene network encoding highly diverse functions, including negative control over multiple iron uptake systems. How this system contributes to tolerance is poorly understood. Here, we show that β-lactam antibiotics cause an increase in intracellular free iron levels and collateral oxidative damage, which is exacerbated in the ∆vxrAB mutant. Mutating major iron uptake systems dramatically increases ∆vxrAB tolerance to β-lactams. We propose that VxrAB reduces antibiotic-induced toxic iron and concomitant metabolic perturbations by downregulating iron uptake transporters and show that iron sequestration enhances tolerance against β-lactam therapy in a mouse model of cholera infection. Our results suggest that a microorganism's ability to counteract diverse antibiotic-induced stresses promotes high-level antibiotic tolerance and highlights the complex secondary responses elicited by antibiotics.
Collapse
Affiliation(s)
- Jung‐Ho Shin
- Weill Institute for Cell and Molecular BiologyCornell, UniversityIthacaNYUSA
- Department of MicrobiologyCornell UniversityIthacaNYUSA
| | - Donghui Choe
- Department of Biological SciencesKorea Advanced Institute of Science and TechnologyDaejeonKorea
- KI for the BioCenturyKorea Advanced Institute of Science and TechnologyDaejeonKorea
| | - Brett Ransegnola
- Weill Institute for Cell and Molecular BiologyCornell, UniversityIthacaNYUSA
- Department of MicrobiologyCornell UniversityIthacaNYUSA
| | - Hye‐Rim Hong
- Weill Institute for Cell and Molecular BiologyCornell, UniversityIthacaNYUSA
- Department of MicrobiologyCornell UniversityIthacaNYUSA
| | - Ikenna Onyekwere
- Weill Institute for Cell and Molecular BiologyCornell, UniversityIthacaNYUSA
- Department of MicrobiologyCornell UniversityIthacaNYUSA
| | - Trevor Cross
- Weill Institute for Cell and Molecular BiologyCornell, UniversityIthacaNYUSA
- Department of MicrobiologyCornell UniversityIthacaNYUSA
| | - Qiaojuan Shi
- Meinig School of Biomedical EngineeringCornell UniversityIthacaNYUSA
| | - Byung‐Kwan Cho
- Department of Biological SciencesKorea Advanced Institute of Science and TechnologyDaejeonKorea
- KI for the BioCenturyKorea Advanced Institute of Science and TechnologyDaejeonKorea
- Intelligent Synthetic Biology CenterDaejeonKorea
| | - Lars F Westblade
- Department of Pathology and Laboratory MedicineWeill Cornell MedicineNew YorkNYUSA
- Division of Infectious DiseasesDepartment of MedicineWeill Cornell MedicineNew YorkNYUSA
| | - Ilana L Brito
- Meinig School of Biomedical EngineeringCornell UniversityIthacaNYUSA
| | - Tobias Dörr
- Weill Institute for Cell and Molecular BiologyCornell, UniversityIthacaNYUSA
- Department of MicrobiologyCornell UniversityIthacaNYUSA
- Cornell Institute of Host‐Microbe Interactions and DiseaseCornell UniversityIthacaNYUSA
| |
Collapse
|
41
|
Huemer M, Mairpady Shambat S, Brugger SD, Zinkernagel AS. Antibiotic resistance and persistence-Implications for human health and treatment perspectives. EMBO Rep 2020; 21:e51034. [PMID: 33400359 PMCID: PMC7726816 DOI: 10.15252/embr.202051034] [Citation(s) in RCA: 376] [Impact Index Per Article: 75.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/13/2020] [Accepted: 11/02/2020] [Indexed: 12/24/2022] Open
Abstract
Antimicrobial resistance (AMR) and persistence are associated with an elevated risk of treatment failure and relapsing infections. They are thus important drivers of increased morbidity and mortality rates resulting in growing healthcare costs. Antibiotic resistance is readily identifiable with standard microbiological assays, and the threat imposed by antibiotic resistance has been well recognized. Measures aiming to reduce resistance development and spreading of resistant bacteria are being enforced. However, the phenomenon of bacteria surviving antibiotic exposure despite being fully susceptible, so-called antibiotic persistence, is still largely underestimated. In contrast to antibiotic resistance, antibiotic persistence is difficult to measure and therefore often missed, potentially leading to treatment failures. In this review, we focus on bacterial mechanisms allowing evasion of antibiotic killing and discuss their implications on human health. We describe the relationship between antibiotic persistence and bacterial heterogeneity and discuss recent studies that link bacterial persistence and tolerance with the evolution of antibiotic resistance. Finally, we review persister detection methods, novel strategies aiming at eradicating bacterial persisters and the latest advances in the development of new antibiotics.
Collapse
Affiliation(s)
- Markus Huemer
- Department of Infectious Diseases and Hospital EpidemiologyUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| | - Srikanth Mairpady Shambat
- Department of Infectious Diseases and Hospital EpidemiologyUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| | - Silvio D Brugger
- Department of Infectious Diseases and Hospital EpidemiologyUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| | - Annelies S Zinkernagel
- Department of Infectious Diseases and Hospital EpidemiologyUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| |
Collapse
|
42
|
Pessione E. The Russian Doll Model: How Bacteria Shape Successful and Sustainable Inter-Kingdom Relationships. Front Microbiol 2020; 11:573759. [PMID: 33193180 PMCID: PMC7606975 DOI: 10.3389/fmicb.2020.573759] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/31/2020] [Indexed: 12/20/2022] Open
Abstract
Successful inter-kingdom relationships are based upon a dynamic balance between defense and cooperation. A certain degree of competition is necessary to guarantee life spread and development. On the other hand, cooperation is a powerful tool to ensure a long lasting adaptation to changing environmental conditions and to support evolution to a higher level of complexity. Bacteria can interact with their (true or potential) parasites (i.e., phages) and with their multicellular hosts. In these model interactions, bacteria learnt how to cope with their inner and outer host, transforming dangerous signals into opportunities and modulating responses in order to achieve an agreement that is beneficial for the overall participants, thus giving rise to a more complex "organism" or ecosystem. In this review, particular attention will be addressed to underline the minimal energy expenditure required for these successful interactions [e.g., moonlighting proteins, post-translational modifications (PTMs), and multitasking signals] and the systemic vision of these processes and ways of life in which the system proves to be more than the sum of the single components. Using an inside-out perspective, I will examine the possibility of multilevel interactions, in which viruses help bacteria to cope with the animal host and bacteria support the human immune system to counteract viral infection in a circular vision. In this sophisticated network, bacteria represent the precious link that insures system stability with relative low energy expenditure.
Collapse
Affiliation(s)
- Enrica Pessione
- Department of Life Sciences and Systems Biology, School of Nature Sciences, Università degli Studi di Torino, Turin, Italy
| |
Collapse
|
43
|
MazEF-rifampicin interaction suggests a mechanism for rifampicin induced inhibition of persisters. BMC Mol Cell Biol 2020; 21:73. [PMID: 33109090 PMCID: PMC7590665 DOI: 10.1186/s12860-020-00316-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/11/2020] [Indexed: 12/15/2022] Open
Abstract
Background Persistence is a natural phenomenon whereby a subset of a population of isogenic bacteria either grow slow or become dormant conferring them with the ability to withstand various stresses including antibiotics. In a clinical setting bacterial persistence often leads to the recalcitrance of various infections increasing the treatment time and cost. Additionally, some studies also indicate that persistence can also pave way for the emergence of resistant strains. In a laboratory setting this persistent phenotype is enriched in nutritionally deprived environments. Consequently, in a batch culture the late stationary phase is enriched with persistent bacteria. The mechanism of persister cell formation and its regulation is not well understood. Toxin-antitoxin (TA) systems have been implicated to be responsible for bacterial persistence and rifampicin is used to treat highly persistent bacterial strains. The current study tries to explore a possible interaction between rifampicin and the MazEF TA system that furthers the former’s success rate in treating persistent bacteria. Results In the current study we found that the population of bacteria in the death phase of a batch culture consists of metabolically inactive live cells resembling persisters, which showed higher membrane depolarization as compared to the log phase bacteria. We also observed an increase in the expression of the MazEF TA modules in this phase. Since rifampicin is used to kill the persisters, we assessed the interaction of rifampicin with MazEF complex. We showed that rifampicin moderately interacts with MazEF complex with 1:1 stoichiometry. Conclusion Our study suggests that the interaction of rifampicin with MazEF complex might play an important role in inhibition of persisters. Supplementary information The online version contains supplementary material available at 10.1186/s12860-020-00316-8.
Collapse
|
44
|
Kawano A, Yamasaki R, Sakakura T, Takatsuji Y, Haruyama T, Yoshioka Y, Ariyoshi W. Reactive Oxygen Species Penetrate Persister Cell Membranes of Escherichia coli for Effective Cell Killing. Front Cell Infect Microbiol 2020; 10:496. [PMID: 33042869 PMCID: PMC7530241 DOI: 10.3389/fcimb.2020.00496] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 08/10/2020] [Indexed: 02/05/2023] Open
Abstract
Persister cells are difficult to eliminate because they are tolerant to antibiotic stress. In the present study, using artificially induced Escherichia coli persister cells, we found that reactive oxygen species (ROS) have greater effects on persister cells than on exponential cells. Thus, we examined which types of ROS could effectively eliminate persister cells and determined the mechanisms underlying the effects of these ROS. Ultraviolet (UV) light irradiation can kill persister cells, and bacterial viability is markedly increased under UV shielding. UV induces the production of ROS, which kill bacteria by moving toward the shielded area. Electron spin resonance-based analysis confirmed that hydroxyl radicals are produced by UV irradiation, although singlet oxygen is not produced. These results clearly revealed that ROS sterilizes persister cells more effectively compared to the sterilization of exponential cells (**p < 0.01). These ROS do not injure the bacterial cell wall but rather invade the cell, followed by cell killing. Additionally, the sterilization effect on persister cells was increased by exposure to oxygen plasma during UV irradiation. However, vapor conditions decreased persister cell sterilization by reducing the levels of hydroxyl radicals. We also verified the effect of ROS against bacteria in biofilms that are more resistant than planktonic cells. Although UV alone could not completely sterilize the biofilm bacteria, UV with ROS achieved complete sterilization. Our results demonstrate that persister cells strongly resist the effects of antibiotics and starvation stress but are less able to withstand exposure to ROS. It was shown that ROS does not affect the cell membrane but penetrates it and acts internally to kill persister cells. In particular, it was clarified that the hydroxy radical is an effective sterilizer to kill persister cells.
Collapse
Affiliation(s)
- Aki Kawano
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Japan
| | - Ryota Yamasaki
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Japan
| | - Tatsuya Sakakura
- Division of Functional Interface Engineering, Department of Biological Systems and Engineering, Kyushu Institute of Technology, Kitakyushu, Japan
| | - Yoshiyuki Takatsuji
- Division of Functional Interface Engineering, Department of Biological Systems and Engineering, Kyushu Institute of Technology, Kitakyushu, Japan
| | - Tetsuya Haruyama
- Division of Functional Interface Engineering, Department of Biological Systems and Engineering, Kyushu Institute of Technology, Kitakyushu, Japan
| | - Yoshie Yoshioka
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Japan
| | - Wataru Ariyoshi
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Japan
| |
Collapse
|
45
|
Alvendal C, Mohanty S, Bohm-Starke N, Brauner A. Anti-biofilm activity of chlorhexidine digluconate against Candida albicans vaginal isolates. PLoS One 2020; 15:e0238428. [PMID: 32941438 PMCID: PMC7498037 DOI: 10.1371/journal.pone.0238428] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 08/17/2020] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES Recurrent vulvovaginal candidiasis (RVVC) causes significant morbidity. Candida albicans is the main pathogen associated with both sporadic and recurrent candidiasis. Due to unsatisfactory treatment effect, the impact of chlorhexidine digluconate and fluconazole alone or in combination on C. albicans and biofilm was investigated. METHODS Vaginal C. albicans isolates from 18 patients with recurrent candidiasis and commensals from 19 asymptomatic women were isolated by culture. Crystal violet, XTT and colony forming unit assay were used to analyze the effect of chlorhexidine digluconate and fluconazole on growth of C. albicans, formation of new and already established, mature, biofilm. RESULTS Fluconazole reduced the growth of planktonic C. albicans. However, in established biofilm, fluconazole had no effect on the candida cells and was not able to disperse and reduce the biofilm. By contrast, chlorhexidine digluconate had a direct killing effect on C. albicans grown both planktonically and in biofilm. Chlorhexidine digluconate also dispersed mature biofilm and inhibited formation of new biofilm. No major differences were observed between commensal isolates and candida causing recurrent vulvovaginitis with respect to biofilm or growth after chlorhexidine digluconate treatment. CONCLUSION Biofilm is a problem in patients with recurrent vulvovaginal candidiasis reducing the effect of antifungal treatment. Development of new treatment strategies are urgently needed to decrease the recurrences. In already established biofilm, chlorhexidine digluconate dispersed the biofilm and was more effective in eradicating candida compared to fluconazole. Future treatment strategy may thus be a combination of chlorhexidine digluconate and fluconazole and prophylactic use of chlorhexidine digluconate to prevent biofilm formation and restrict infections.
Collapse
Affiliation(s)
- Cathrin Alvendal
- Division of Obstetrics and Gynecology, Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, Sweden
- * E-mail:
| | - Soumitra Mohanty
- Division of Clinical Microbiology, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Nina Bohm-Starke
- Division of Obstetrics and Gynecology, Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, Sweden
| | - Annelie Brauner
- Division of Clinical Microbiology, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
46
|
Antibiotic resistance related to biofilm formation in Streptococcus suis. Appl Microbiol Biotechnol 2020; 104:8649-8660. [PMID: 32897417 DOI: 10.1007/s00253-020-10873-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/23/2020] [Accepted: 08/31/2020] [Indexed: 12/13/2022]
Abstract
Streptococcus suis (S. suis) is an important zoonotic agent, which seriously impacts the pig industry and human health in various countries. Biofilm formation is likely contributing to the virulence and drug resistance in S. suis. A better knowledge of biofilm formation as well as to biofilm-dependent drug resistance mechanisms in S. suis can be of great significance for the prevention and treatment of S. suis infections. This literature review updates the latest scientific data related to biofilm formation in S. suis and its impact on drug tolerance and resistance.Key points• Biofilm formation is the important reasons for drug resistance of SS infections.• The review includes the regulatory mechanism of SS biofilm formation.• The review includes the drug resistance mechanisms of SS biofilm.
Collapse
|
47
|
Snyder ER, Savitske BJ, Credille BC. Concordance of disk diffusion, broth microdilution, and whole-genome sequencing for determination of in vitro antimicrobial susceptibility of Mannheimia haemolytica. J Vet Intern Med 2020; 34:2158-2168. [PMID: 32893911 PMCID: PMC7517867 DOI: 10.1111/jvim.15883] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/13/2020] [Accepted: 08/19/2020] [Indexed: 12/25/2022] Open
Abstract
Background Extensive drug resistance (XDR) is an emerging concern with Mannheimia haemolytica, and a variety of testing methods are available for characterizing in vitro antimicrobial susceptibility. Objectives To compare the concordance among disk diffusion, broth microdilution, and whole genome sequencing (WGS) for susceptibility testing of M. haemolytica before and after mass treatment using tulathromycin. Animals Forty‐eight M. haemolytica isolates collected from high‐risk beef stocker calves before and after mass treatment (metaphylaxis) using tulathromycin (Draxxin, Zoetis, Parsippany, NJ) given at the label dosage of 2.5 mg/kg body weight SC in the neck. Methods In vitro antimicrobial susceptibility was determined for all 48 isolates using disk diffusion, broth microdilution, and WGS. Concordance was calculated between pairs of susceptibility testing methods as follows: number of isolates classified identically by the 2 testing methods for each timepoint, divided by the number of isolates tested at that timepoint. Discordance was calculated as follows: number of isolates classified differently by the 2 testing methods for each timepoint, divided by the number of isolates tested at that timepoint. Results Concordance between testing methods ranged from 42.3% to 100%, depending on antimicrobial evaluated, timing of sample collection, and testing method used. Very major errors were identified in up to 7.7% of classifications whereas minor errors were seen in up to 50% of classifications depending on antimicrobial evaluated, timing of sample collection, and testing method used. Conclusions and Clinical Importance Our results show that discrepancies in the results of different susceptibility testing methods occur and suggest a need for greater harmonization of susceptibility testing methods.
Collapse
Affiliation(s)
- Emily R Snyder
- Food Animal Health and Management Program, Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Bridget J Savitske
- Food Animal Health and Management Program, Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Brent C Credille
- Food Animal Health and Management Program, Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
48
|
Fleming D, Redman W, Welch GS, Mdluli NV, Rouchon CN, Frank KL, Rumbaugh KP. Utilizing glycoside hydrolases to improve the quantitation and visualization of biofilm bacteria. Biofilm 2020; 2:100037. [PMID: 33447822 PMCID: PMC7798457 DOI: 10.1016/j.bioflm.2020.100037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 01/06/2023] Open
Abstract
The complexity of microbial biofilms offers several challenges to the use of traditional means of microbial research. In particular, it can be difficult to calculate accurate numbers of biofilm bacteria, because even after thorough homogenization or sonication, small pieces of the biofilm remain, which contain numerous bacterial cells and result in inaccurately low colony forming units (CFU). In addition, imaging of infected tissue ex vivo often results in a disparity between the CFU and the number of bacterial cells observed under the microscope. We hypothesized that this phenomenon is due to the biofilm extracellular polymeric substance decreasing the accessibility of stains and antibodies to the embedded bacterial cells. In this study, we describe incorporating EPS-degrading glycoside hydrolases for CFU determination to obtain a more accurate estimation of the viable cells and for immunohistochemistry to disrupt the biofilm matrix and increase primary antibody binding to the bacterial cells.
Collapse
Affiliation(s)
- Derek Fleming
- Departments of Surgery, Immunology and Molecular Microbiology, and the Burn Center of Research Excellence, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Whitni Redman
- Departments of Surgery, Immunology and Molecular Microbiology, and the Burn Center of Research Excellence, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Garrett S Welch
- Departments of Surgery, Immunology and Molecular Microbiology, and the Burn Center of Research Excellence, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Nontokozo V Mdluli
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Candace N Rouchon
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Kristi L Frank
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Kendra P Rumbaugh
- Departments of Surgery, Immunology and Molecular Microbiology, and the Burn Center of Research Excellence, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
49
|
Doolin T, Amir HM, Duong L, Rosenzweig R, Urban LA, Bosch M, Pol A, Gross SP, Siryaporn A. Mammalian histones facilitate antimicrobial synergy by disrupting the bacterial proton gradient and chromosome organization. Nat Commun 2020; 11:3888. [PMID: 32753666 PMCID: PMC7403156 DOI: 10.1038/s41467-020-17699-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/07/2020] [Indexed: 12/30/2022] Open
Abstract
First proposed as antimicrobial agents, histones were later recognized for their role in condensing chromosomes. Histone antimicrobial activity has been reported in innate immune responses. However, how histones kill bacteria has remained elusive. The co-localization of histones with antimicrobial peptides (AMPs) in immune cells suggests that histones may be part of a larger antimicrobial mechanism in vivo. Here we report that histone H2A enters E. coli and S. aureus through membrane pores formed by the AMPs LL-37 and magainin-2. H2A enhances AMP-induced pores, depolarizes the bacterial membrane potential, and impairs membrane recovery. Inside the cytoplasm, H2A reorganizes bacterial chromosomal DNA and inhibits global transcription. Whereas bacteria recover from the pore-forming effects of LL-37, the concomitant effects of H2A and LL-37 are irrecoverable. Their combination constitutes a positive feedback loop that exponentially amplifies their antimicrobial activities, causing antimicrobial synergy. More generally, treatment with H2A and the pore-forming antibiotic polymyxin B completely eradicates bacterial growth.
Collapse
Affiliation(s)
- Tory Doolin
- Department of Developmental and Cell Biology, UC Irvine, Irvine, CA, 92697, USA
| | - Henry M Amir
- Department of Physics & Astronomy, UC Irvine, Irvine, CA, 92697, USA
| | - Leora Duong
- Department of Molecular Biology & Biochemistry, UC Irvine, Irvine, CA, 92697, USA
| | - Rachel Rosenzweig
- Department of Materials Sciences and Engineering, UC Irvine, Irvine, CA, 92697, USA
| | - Lauren A Urban
- Department of Microbiology and Molecular Genetics, UC Irvine, Irvine, CA, 92697, USA
| | - Marta Bosch
- Cell Compartments and Signaling Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, 08036, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, 08036, Spain
| | - Albert Pol
- Cell Compartments and Signaling Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, 08036, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, 08036, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, 08010, Spain
| | - Steven P Gross
- Department of Developmental and Cell Biology, UC Irvine, Irvine, CA, 92697, USA.
- Department of Physics & Astronomy, UC Irvine, Irvine, CA, 92697, USA.
| | - Albert Siryaporn
- Department of Physics & Astronomy, UC Irvine, Irvine, CA, 92697, USA.
- Department of Molecular Biology & Biochemistry, UC Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
50
|
Mangiaterra G, Cedraro N, Vaiasicca S, Citterio B, Galeazzi R, Laudadio E, Mobbili G, Minnelli C, Bizzaro D, Biavasco F. Role of Tobramycin in the Induction and Maintenance of Viable but Non-Culturable Pseudomonas aeruginosa in an In Vitro Biofilm Model. Antibiotics (Basel) 2020; 9:antibiotics9070399. [PMID: 32664334 PMCID: PMC7400124 DOI: 10.3390/antibiotics9070399] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/12/2020] [Accepted: 07/08/2020] [Indexed: 01/25/2023] Open
Abstract
The recurrence of Pseudomonas aeruginosa (PA) biofilm infections is a major issue in cystic fibrosis (CF) patients. A pivotal role is played by the presence of antibiotic-unresponsive persisters and/or viable but non-culturable (VBNC) forms, whose development might be favored by subinhibitory antibiotic concentrations. The involvement of tobramycin and ciprofloxacin, widely used to treat CF PA lung infections, in the abundance of VBNC cells was investigated in PA biofilms models. In vitro biofilms of the laboratory strain PAO1-N and the clinical strain C24 were developed and starved by subculture for 170 days in a non-nutrient (NN) broth, unsupplemented or supplemented with one-quarter minimal inhibitory concentration (MIC) of tobramycin or ciprofloxacin. VBNC cells abundance, estimated as the difference between total live (detected by qPCR and flow cytometry) and colony forming unit (CFU) counts, showed a strain- and drug-specific pattern. A greater and earlier abundance of VBNC PAO1-N cells was detected in all conditions. Exposure of the C24 strain to NN and NN + ciprofloxacin induced only a transient VBNC subpopulation, which was more abundant and stable until the end of the experiment in tobramycin-exposed biofilms. The same response to tobramycin was observed in the PAO1-N strain. These findings suggest that low tobramycin concentrations might contribute to PA infection recurrence by favoring the development of VBNC forms.
Collapse
Affiliation(s)
- Gianmarco Mangiaterra
- Department of Life and Environmental Sciences, Polytechnic University of Marche, via Brecce Bianche, 60131 Ancona, Italy; (N.C.); (S.V.); (R.G.); (G.M.); (C.M.); (D.B.); (F.B.)
- Correspondence: ; Tel.: +39-071-220-4622; Fax: +39-071-220-4316
| | - Nicholas Cedraro
- Department of Life and Environmental Sciences, Polytechnic University of Marche, via Brecce Bianche, 60131 Ancona, Italy; (N.C.); (S.V.); (R.G.); (G.M.); (C.M.); (D.B.); (F.B.)
| | - Salvatore Vaiasicca
- Department of Life and Environmental Sciences, Polytechnic University of Marche, via Brecce Bianche, 60131 Ancona, Italy; (N.C.); (S.V.); (R.G.); (G.M.); (C.M.); (D.B.); (F.B.)
| | - Barbara Citterio
- Department of Biomolecular Science, Biotechnology Section, University of Urbino “Carlo Bo”, via Arco d’Augusto 2, 61032 Fano, Italy;
| | - Roberta Galeazzi
- Department of Life and Environmental Sciences, Polytechnic University of Marche, via Brecce Bianche, 60131 Ancona, Italy; (N.C.); (S.V.); (R.G.); (G.M.); (C.M.); (D.B.); (F.B.)
| | - Emiliano Laudadio
- Department of Materials, Environmental Sciences and Urban Planning, Polytechnic University of Marche, via Brecce Bianche, 60131 Ancona, Italy;
| | - Giovanna Mobbili
- Department of Life and Environmental Sciences, Polytechnic University of Marche, via Brecce Bianche, 60131 Ancona, Italy; (N.C.); (S.V.); (R.G.); (G.M.); (C.M.); (D.B.); (F.B.)
| | - Cristina Minnelli
- Department of Life and Environmental Sciences, Polytechnic University of Marche, via Brecce Bianche, 60131 Ancona, Italy; (N.C.); (S.V.); (R.G.); (G.M.); (C.M.); (D.B.); (F.B.)
| | - Davide Bizzaro
- Department of Life and Environmental Sciences, Polytechnic University of Marche, via Brecce Bianche, 60131 Ancona, Italy; (N.C.); (S.V.); (R.G.); (G.M.); (C.M.); (D.B.); (F.B.)
| | - Francesca Biavasco
- Department of Life and Environmental Sciences, Polytechnic University of Marche, via Brecce Bianche, 60131 Ancona, Italy; (N.C.); (S.V.); (R.G.); (G.M.); (C.M.); (D.B.); (F.B.)
| |
Collapse
|