1
|
Patel M, Patel K. Emerging insights of Staphylococcus spp. in human mastitis. Microb Pathog 2025; 205:107685. [PMID: 40349998 DOI: 10.1016/j.micpath.2025.107685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 05/01/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Human mastitis represents a prevalent and intricate condition that significantly challenges breastfeeding women, often exacerbated by pathogenic bacteria such as Staphylococcus aureus. A deep understanding of the interplay between human mastitis, the breast milk microbiome, and causative agents is imperative. This understanding must focus on the bacterium's virulence and resistance genes, which critically influence the severity and persistence of mastitis. Current methods for detecting these genes, including Polymerase Chain Reaction (PCR), 16S rRNA gene sequencing, shotgun metagenomic sequencing, multiplex PCR, whole genome sequencing (WGS), loop-mediated isothermal amplification (LAMP), CRISPR-based assays, and microarray technology, are vital in elucidating bacterial pathogenicity and resistance profiles. However, advanced attention is required to refine diagnostic techniques, enabling earlier detection and more effective therapeutic approaches for human mastitis. The involvement of Staphylococcus aureus in human infection should be a prime focus, especially in women's health, which deals directly with neonates. Essential virulence genes in Staphylococcus species are instrumental in infection mechanisms and antibiotic resistance, serving as potential targets for personalized treatments. Thus, this review focuses on Staphylococcusaureus-induced mastitis, examining its virulence factors and detection techniques to advance diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Mansi Patel
- Department of Medical Laboratory Technology, Bapubhai Desaibhai Patel Institute of Paramedical Sciences (BDIPS), Charotar University of Science and Technology (CHARUSAT), Anand, Gujarat, 388421, India
| | - Khushal Patel
- Department of Medical Laboratory Technology, Bapubhai Desaibhai Patel Institute of Paramedical Sciences (BDIPS), Charotar University of Science and Technology (CHARUSAT), Anand, Gujarat, 388421, India.
| |
Collapse
|
2
|
Kim DY, Kim TJ. Metabolic Stress Induced by Quercetin Enhances Dormancy and Persistence in Staphylococcus aureus. Antibiotics (Basel) 2025; 14:424. [PMID: 40426490 DOI: 10.3390/antibiotics14050424] [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: 03/20/2025] [Revised: 04/14/2025] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: The persistence of Staphylococcus aureus poses a significant challenge in clinical treatments because of its ability to withstand antibiotic therapy. This study assessed the role of quercetin in promoting bacterial dormancy and persistence through ATP depletion and revealed its potential impact on antibiotic tolerance. Methods: To assess the effects of quercetin on bacterial metabolism and persistence, S. aureus cultures were treated with quercetin, and intracellular ATP levels were then measured. The effect of quercetin on persister cell formation was assessed using antibiotic exposure assays, including pre-treatment and post-treatment strategies. Results: Quercetin treatment significantly depleted intracellular ATP levels in a dose-dependent manner, suggesting the presence of metabolic stress. This ATP depletion correlated with increased persister cell formation across multiple antibiotic treatments, indicating that quercetin-induced dormancy enhances bacterial persistence. Notably, quercetin pre-treatment further increased persister cell counts, while delayed quercetin administration increased persister cell survival, highlighting the influence of the timing of metabolic stress on persistence outcomes. Conclusions: Quercetin promotes bacterial persistence by inducing ATP depletion and metabolic dormancy. Although quercetin's bactericidal properties may initially impair bacterial growth, its potential to enhance persistence underscores the complexity of its effects. Further research is necessary to determine optimal strategies for harnessing the antimicrobial properties of quercetin while minimizing its persistence-promoting effects.
Collapse
Affiliation(s)
- Dae-Youn Kim
- Department of Forest Products and Biotechnology, Kookmin University, Seoul 02707, Republic of Korea
| | - Tae-Jong Kim
- Department of Forest Products and Biotechnology, Kookmin University, Seoul 02707, Republic of Korea
| |
Collapse
|
3
|
Brandt TJ, Skaggs H, Hundley T, Yoder-Himes DR. Burkholderia cenocepacia-mediated inhibition of Staphylococcus aureus growth and biofilm formation. J Bacteriol 2025; 207:e0011623. [PMID: 40135855 PMCID: PMC12004965 DOI: 10.1128/jb.00116-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 09/01/2024] [Indexed: 03/27/2025] Open
Abstract
Staphylococcus aureus asymptomatically colonizes the nasal cavity and pharynx of up to 60% of the human population and, as an opportunistic pathogen, can breach its normal habitat, resulting in life-threatening infections. S. aureus infections are of additional concern for populations with impaired immune function such as those with cystic fibrosis (CF) or chronic granulomatous disease. Multi-drug resistance is increasingly common in S. aureus infections, creating an urgent need for new antimicrobials or compounds that improve efficacy of currently available antibiotics. S. aureus biofilms, such as those found in the lungs of people with CF and in soft tissue infections, are notoriously recalcitrant to antimicrobial treatment due to the characteristic metabolic differences associated with a sessile mode of growth. In this work, we show that another CF pathogen, Burkholderia cenocepacia, produces one or more secreted compounds that can prevent S. aureus biofilm formation and inhibit existing S. aureus biofilms. The B. cenocepacia-mediated antagonistic activity is restricted to S. aureus species and perhaps some other staphylococci; however, this inhibition does not necessarily extend to other Gram-positive species. This inhibitory activity is due to death of S. aureus through a contact-independent mechanism, potentially mediated through the siderophore pyochelin and perhaps additional compounds. This works paves the way to better understanding of interactions between these two bacterial pathogens.IMPORTANCEStaphylococcus aureus is a major nosocomial pathogen responsible for infecting thousands of people each year. Some strains are becoming increasingly resistant to antimicrobials, and consequently new treatments must be sought. This paper describes the characterization of one or more compounds capable of inhibiting S. aureus biofilm formation and may potentially lead to development of a new therapeutic.
Collapse
Affiliation(s)
- Tiffany J. Brandt
- Department of Biology, University of Louisville, Louisville, Kentucky, USA
| | - Hayden Skaggs
- Department of Biology, University of Louisville, Louisville, Kentucky, USA
| | - Thomas Hundley
- Department of Biology, University of Louisville, Louisville, Kentucky, USA
| | | |
Collapse
|
4
|
Lee B, Seo J, Jeong SW. Case Report: Long-term complications of subcutaneous ureteral bypass migration in an adult female Papillon. Front Vet Sci 2025; 12:1543299. [PMID: 40144524 PMCID: PMC11938841 DOI: 10.3389/fvets.2025.1543299] [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: 12/11/2024] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
A 6-year-old spayed female 4.4-kg Papillon with only the left kidney presented with vomiting. Imaging unveiled ureterolithiasis and hydronephrosis, while serum chemistry displayed elevated creatinine, blood urea nitrogen, and C-reactive protein. Urinalysis revealed bacteria and bacterial phagocytes. After subcutaneous ureteral bypass (SUB) placement, kidney panels were normalized. The nephrostomy and cystostomy catheters had migrated into the renal parenchyma and bladder wall on postoperative day (POD) 212 and 369, respectively. As the migration advanced, they entered the ureter and bladder on POD 369 and 796, respectively. The SUB, excluding the nephrostomy catheter, was removed on POD 930 due to migration, obstruction, and extrusion of the SUB shunting port. On POD 937, creatinine and BUN levels remained normal. By POD 1063, the ureteroliths had disappeared. This case highlights the need for vigilant monitoring of catheter migration as a potential complication following SUB placement. Early identification and timely intervention are essential for reducing morbidity and improving patient outcomes.
Collapse
Affiliation(s)
- Boram Lee
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Jeonghyun Seo
- Department of Veterinary Medicine, College of Veterinary Medicine, Jeju National University, Jeju, Republic of Korea
- Time Animal Medical Center, Daejeon, Republic of Korea
| | - Soon-Wuk Jeong
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
5
|
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
|
6
|
Meng M, Huo R, Li Z, Wang X, Qiu Y, Shen X, Chang G. Protective effect of curcumin-loaded zeolitic imidazolate framework-8-based pH-responsive drug delivery system against Staphylococcus aureus infection. Microb Pathog 2025; 200:107336. [PMID: 39864761 DOI: 10.1016/j.micpath.2025.107336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/08/2025] [Accepted: 01/23/2025] [Indexed: 01/28/2025]
Abstract
Mastitis, generally caused by pathogenic microorganisms, is a serious disease in dairy farming. Staphylococcus aureus (S. aureus) is one of the main pathogens that induces mastitis in dairy cows. It evades the innate and adaptive immune responses of dairy cows, causing recessive transmission and harming the health of the mammary glands. Antibiotics remain the primary treatment; however, their excessive use can lead to antimicrobial resistance. Therefore, it is necessary to develop new strategies to replace antibiotic therapies. The zeolitic imidazolate framework (ZIF-8) is a metal-organic skeleton material with applications in biology and drug delivery. This study aimed to construct a novel nanodrug delivery system for S. aureus infection by combining ZIF-8 with curcumin (ZIF-8@CCM), which exhibits antibacterial and anti-inflammatory properties. Bovine mammary epithelial cells (BMECs) and mice were used to evaluate the therapeutic efficacy and biotoxicity of the system, and to explore the protective mechanism of ZIF-8@CCM. The results showed that ZIF-8@CCM exhibited high drug loading capacity, stability, and pH responsiveness. Both in vitro and in vivo experiments revealed that ZIF-8@CCM effectively released encapsulated curcumin in response to the acidic microenvironment induced by bacterial infection, which in turn enhanced the bactericidal efficacy. It not only prevents biofilm formation, but also mitigates the toxic side effects associated with drug treatments, showing excellent bioavailability and biocompatibility. Furthermore, ZIF-8@CCM also attenuated S. aureus-induced inflammatory through suppressing the activation of TLR2-NF-κB pathway. Consequently, ZIF-8@CCM is an effective targeted antibacterial and anti-inflammatory drug, showing promise as a novel therapeutic agent for the clinical management of S. aureus-induced mastitis in dairy cows.
Collapse
Affiliation(s)
- Meijuan Meng
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, PR China
| | - Ran Huo
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, PR China
| | - Zhixin Li
- Animal Disease Prevention and Control Center of Ningxia Hui Autonomous Region, Yinchuan, 750001, Ningxia, PR China
| | - Xiaoliang Wang
- Animal Disease Prevention and Control Center of Ningxia Hui Autonomous Region, Yinchuan, 750001, Ningxia, PR China
| | - Yawei Qiu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, PR China
| | - Xiangzhen Shen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, PR China
| | - Guangjun Chang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, PR China; Animal Disease Prevention and Control Center of Ningxia Hui Autonomous Region, Yinchuan, 750001, Ningxia, PR China.
| |
Collapse
|
7
|
Zahid ALN, Wang KC, Thomsen LE, Meier S, Jensen PR. In-cell NMR reveals metabolic adaptations in central carbon pathways driving antibiotic tolerance in Salmonella Typhimurium. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2025; 17:1062-1073. [PMID: 39791466 DOI: 10.1039/d4ay02023e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Antibiotic tolerance presents a significant challenge in eradicating bacterial infections, as tolerant strains can survive antibiotic treatment, contributing to the recurrence of infections and the development of resistance. However, unlike antibiotic resistance, tolerance is not detectable by standard susceptibility assays such as minimal inhibitory concentration (MIC) tests. Consequently, antibiotic tolerance often goes unnoticed in clinical settings. Bacterial metabolism is closely linked to antibiotic efficacy, and thus presents as a potential target for novel diagnostic methods. Recent advancements in nuclear magnetic resonance (NMR) spectroscopy, including dynamic nuclear polarization (DNP-NMR), enable a non-invasive real-time approach to analyzing bacterial metabolism. In this study, we applied both 1H and in-cell 13C NMR spectroscopy to investigate metabolic adaptations in a tolerance-evolved Salmonella Typhimurium strain, C10, developed through ten cycles of ampicillin treatment. Our results demonstrated that despite similar MICs and growth rates, the C10 strain exhibited a 25-fold increase in tolerance compared to the wild-type, while exhibiting lower metabolic activity. Under ampicillin stress, however, the C10 strain maintained higher metabolic activity and demonstrated greater resilience in glucose consumption and metabolite production relative to the wild-type. Using DNP-NMR, rapid metabolic shifts in the C10 strain were identified within 10 minutes of exposure to high concentrations of ampicillin, characterized by accumulation of key metabolites such as pyruvate and acetate. Overall, our findings underscore the potential of real-time NMR-based analyses to provide deeper insights into antibiotic tolerance and distinguish between susceptible and tolerant bacterial strains.
Collapse
Affiliation(s)
- Alexandra L N Zahid
- Department of Health Technology, Technical University of Denmark, 2800 Kgs, Lyngby, Denmark.
| | - Ke-Chuan Wang
- Department of Health Technology, Technical University of Denmark, 2800 Kgs, Lyngby, Denmark.
| | - Line Elnif Thomsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark
| | - Sebastian Meier
- Department of Chemistry, Technical University of Denmark, 2800 Kgs, Lyngby, Denmark
| | - Pernille Rose Jensen
- Department of Health Technology, Technical University of Denmark, 2800 Kgs, Lyngby, Denmark.
| |
Collapse
|
8
|
Pengfei S, Yifan Y, Shasha L, Shaowei G, Guanqing H, Dan X, Yong W. Repurposing pinaverium bromide against Staphylococcus and its biofilms with new mechanisms. AMB Express 2024; 14:141. [PMID: 39718732 DOI: 10.1186/s13568-024-01809-x] [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: 11/11/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024] Open
Abstract
Antibiotic resistance by methicillin-resistant Staphylococcus aureus (MRSA) is an urgent threat to human health. The biofilm and persister cells formation ability of MRSA and Staphylococcus epidermidis often companied with extremely high antimicrobial resistance. Pinaverium bromide (PVB) is an antispasmodic compound mainly used for irritable bowel syndrome. Here we demonstrate that PVB could rapidly kill MRSA and S. epidermidis planktonic cells and persister cells avoiding resistance occurrence. Moreover, by crystal violet staining, viable cells counting and SYTO9/PI staining, PVB exhibited strong biofilm inhibition and eradication activities on the 96-well plates, glass surface or titanium discs. And the synergistic antimicrobial effects were observed between PVB and conventional antibiotics (ampicillin, oxacillin, and cefazolin). Mechanism study demonstrated the antimicrobial and antibiofilm effects by PVB were mainly mediated by proton motive force disrupting as well as reactive oxygen species inducing. Although, relatively poor pharmacokinetics were observed by systemic use, PVB could significantly reduce the viable bacterial cell loads and inflammatory infiltration in abscess in vivo caused by the biofilm forming strain ATCC 43,300. In all, our results indicated that PVB could be an alternative antimicrobial reagent for the treatment of MRSA, S. epidermidis and its biofilm related skin and soft tissue infections.
Collapse
Affiliation(s)
- She Pengfei
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Yang Yifan
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Liu Shasha
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Guo Shaowei
- Department of Laboratory Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine (The First Hospital of Changsha), Central South University, No. 311, Yingpan Road, Changsha, 410005, Hunan, China
| | - Huan Guanqing
- Department of Laboratory Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine (The First Hospital of Changsha), Central South University, No. 311, Yingpan Road, Changsha, 410005, Hunan, China
| | - Xiao Dan
- Department of Laboratory Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine (The First Hospital of Changsha), Central South University, No. 311, Yingpan Road, Changsha, 410005, Hunan, China
| | - Wu Yong
- Department of Laboratory Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine (The First Hospital of Changsha), Central South University, No. 311, Yingpan Road, Changsha, 410005, Hunan, China.
| |
Collapse
|
9
|
Batchelder JI, Taylor AJ, Mok WWK. Metabolites augment oxidative stress to sensitize antibiotic-tolerant Staphylococcus aureus to fluoroquinolones. mBio 2024; 15:e0271424. [PMID: 39475229 PMCID: PMC11633220 DOI: 10.1128/mbio.02714-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 10/08/2024] [Indexed: 12/12/2024] Open
Abstract
If left unchecked, infections involving antibiotic-refractory bacteria are expected to cause millions of deaths per year in the coming decades. Beyond genetically resistant bacteria, persisters, which are genetically susceptible cells that survive antibiotic doses that kill the rest of the clonal population, can potentially contribute to treatment failure and infection relapse. Stationary-phase bacterial cultures are enriched with persisters, and it has been shown that stimulating these populations with exogenous nutrients can reduce persistence to different classes of antibiotics, including topoisomerase-targeting fluoroquinolones (FQs). In this study, we show that adding glucose and amino acids to nutrient-starved Staphylococcus aureus cultures enhanced their sensitivity to FQs, including delafloxacin (Dela)-a drug that was recently approved for treating staphylococcal infections. We found that while the added nutrients increased nucleic acid synthesis, this increase was not required to sensitize S. aureus to FQs. We further demonstrate that addition of these nutrients increases membrane potential and the ability to generate harmful reactive oxygen species (ROS) during FQ treatment. Chelating iron, scavenging hydroxyl radicals, and limiting oxygenation during FQ treatment and during recovery following FQ treatment rescued nutrient-stimulated S. aureus. In all, our data suggest that while nutrient stimulation increases the activity of FQ targets in stationary-phase S. aureus, the resulting generation of ROS, presumably made possible through metabolic upregulation, is the primary driver of increased sensitivity to these drugs.IMPORTANCEStaphylococcus aureus causes many chronic and relapsing infections because of its ability to endure host immunity and antibiotic therapy. While several studies have focused on the nutrient requirements for the formation and maintenance of staphylococcal infections, the effects of the nutrient environment on bacterial responses to antibiotic treatment remain understudied. Here, we show that adding nutrients to starved S. aureus activates biosynthetic processes, including DNA synthesis, but it is the generation of harmful reactive oxidants that sensitizes S. aureus to DNA topoisomerase-targeting FQs. Our results suggest that the development of approaches aimed at perturbing metabolism and increasing oxidative stress can potentiate the bactericidal activity of FQs against antibiotic-tolerant S. aureus.
Collapse
Affiliation(s)
- Jonathan I. Batchelder
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, Connecticut, USA
| | - Andrew J. Taylor
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, Connecticut, USA
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
| | - Wendy W. K. Mok
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, Connecticut, USA
| |
Collapse
|
10
|
Lv C, Leng J, Qian M, Sun B, Ye H, Li M, Zhou N, Cheng Z, Chen Y, Guo X, Shang J, Zhang L, Zhu Y. Antimicrobial resistance in Escherichia coli and Staphylococcus aureus at human-animal interfaces on Chongming Island, Shanghai: A One Health perspective. One Health 2024; 19:100910. [PMID: 40206836 PMCID: PMC11980626 DOI: 10.1016/j.onehlt.2024.100910] [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: 08/24/2024] [Revised: 09/28/2024] [Accepted: 10/03/2024] [Indexed: 04/11/2025] Open
Abstract
Antimicrobial resistance (AMR) is a significant concern within the One Health framework due to its ability to spread across multiple interfaces. Phenotypic data remains the primary type for AMR surveillance, but exploring association across multiple interfaces poses certain challenges. In this study, AMR phenotypic data of clinical and food animal E. coli and S. aureus from Chongming Island over the past five years were analyzed to determine key characteristics of AMR and explore its association at the human-animal interface. The clinical E. coli isolates showed significant resistance to penicillins (83.92 %), cephems (63.05 %), fluoroquinolones (62.21 %), and tetracyclines (57.77 %), while S. aureus exhibited high resistance to penicillinase-labile penicillins (90.89 %), macrolides (51.51 %), penicillinase-stable penicillins (43.96 %), and lincosamides (43.55 %). Extended-spectrum β-lactamase (ESBL)-producing E. coli isolates accounted for 53.26 % (1398/2526), while methicillin-resistant Staphylococcus aureus (MRSA) prevalence was 43.81 % (435/993). Notably, there has been an increase in the proportion of E. coli isolates resistant to 8 to 12 antimicrobial classes, and in the proportion of S. aureus isolates resistant to 5 to 9 classes. Certain multi-drug resistance (MDR) phenotypes were first identified in food animal isolates and later emerged in clinical settings. Meanwhile, several MDR phenotypes were shared between the two interfaces, with 44 identified in E. coli and 12 in S. aureus. Further co-occurrence analysis in E. coli and S. aureus identified several co-occurrence phenotypic pairs or clusters, potentially mediated by a single plasmid or multiple plasmids within a bacterium, indicating potential associations at the human-animal interface. To summarize, a heightened prevalence of MDR in clinical E. coli and S. aureus has been observed, with some MDR profiles appearing in food animals before emerging in clinical settings. The co-occurrence of phenotypic pairs or clusters underscores the potential for AMR association and transmission between humans and food animals. Within the One Health framework, integrating genomic data into AMR monitoring is a crucial next step.
Collapse
Affiliation(s)
- Chao Lv
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), National Health Commission Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Shanghai, China
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Leng
- Department of Clinical Laboratory, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Minjian Qian
- Department of Clinical Laboratory, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Bingqing Sun
- Shanghai Center for Animal Diseases Prevention and Control, Shanghai Institute for Veterinary Drugs and Feeds Control, Shanghai, China
| | - HuiPing Ye
- Chongming District Center for Animal Disease Prevention and Control, Shanghai, China
| | - Min Li
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nan Zhou
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zile Cheng
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiwen Chen
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaokui Guo
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Shang
- Shanghai Center for Animal Diseases Prevention and Control, Shanghai Institute for Veterinary Drugs and Feeds Control, Shanghai, China
| | - Li Zhang
- Department of Clinical Laboratory, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Yongzhang Zhu
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Xu S, Zhu J, Li Y, Sun B. Co-transcribed genes SA1833-SA1832 promote persister formation by regulating the transcription of holin-like gene lrgA in methicillin-resistant Staphylococcus aureus strain N315. Int J Med Microbiol 2024; 317:151636. [PMID: 39426162 DOI: 10.1016/j.ijmm.2024.151636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 09/20/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024] Open
Abstract
Staphylococcus aureus, a facultative anaerobic gram-positive bacterial pathogen, has posed major threat to public health worldwide. Upon S. aureus infection, the host immune system is activated for clearance. However, intracellular S. aureus, which remains viable for an extended time, has evolved the ability to escape from immune response and extracellular antibiotics. One of possible strategies is the formation of persisters. Persistence is one of the major causes of S. aureus relapse infection but the underlying mechanisms remain obscure. Here, we identified two co-transcribed genes SA1833-SA1832 that are involved in persister formation in S. aureus. Dysfunction of SA1833 and/or SA1832 significantly reduces persister formation in the presence of ceftizoxime. Additionally, we found that the expression of SA1833 and SA1832 under the induction of oxidative stress and SOS response is strictly regulated by the LexA-RecA pathway. Interestingly, SA1833-SA1832 contributes to persister formation in an lrgA-dependent manner. Moreover, the mouse RAW264.7 macrophage infection model indicated that disrupting SA1833-SA1832 inhibits S. aureus from infecting macrophages and impairs its ability to survive in the intracellular environment.
Collapse
Affiliation(s)
- Shiwen Xu
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Jiade Zhu
- Department of Critical Care Medicine, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
| | - Yujie Li
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China.
| | - Baolin Sun
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China.
| |
Collapse
|
12
|
Liu HY, Prentice EL, Webber MA. Mechanisms of antimicrobial resistance in biofilms. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:27. [PMID: 39364333 PMCID: PMC11445061 DOI: 10.1038/s44259-024-00046-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 09/02/2024] [Indexed: 10/05/2024]
Abstract
Most bacteria in nature exist in aggregated communities known as biofilms, and cells within a biofilm demonstrate major physiological changes compared to their planktonic counterparts. Biofilms are associated with many different types of infections which can have severe impacts on patients. Infections involving a biofilm component are often chronic and highly recalcitrant to antibiotic therapy as a result of intrinsic physical factors including extracellular matrix production, low growth rates, altered antibiotic target production and efficient exchange of resistance genes. This review describes the biofilm lifecycle, phenotypic characteristics of a biofilm, and contribution of matrix and persister cells to biofilms intrinsic tolerance to antimicrobials. We also describe how biofilms can evolve antibiotic resistance and transfer resistance genes within biofilms. Multispecies biofilms and the impacts of various interactions, including cooperation and competition, between species on tolerance to antimicrobials in polymicrobial biofilm communities are also discussed.
Collapse
Affiliation(s)
- Ho Yu Liu
- Quadram Institute Biosciences, Norwich Research Park, Norwich, Norfolk NR4 7UQ UK
- Norwich Medical School, University of East Anglia, Norwich, Norfolk NR4 7TJ UK
- Centre for Microbial Interactions, Norwich Research Park, Norwich, Norfolk NR4 7UG UK
| | - Emma L Prentice
- Quadram Institute Biosciences, Norwich Research Park, Norwich, Norfolk NR4 7UQ UK
| | - Mark A Webber
- Quadram Institute Biosciences, Norwich Research Park, Norwich, Norfolk NR4 7UQ UK
- Norwich Medical School, University of East Anglia, Norwich, Norfolk NR4 7TJ UK
- Centre for Microbial Interactions, Norwich Research Park, Norwich, Norfolk NR4 7UG UK
| |
Collapse
|
13
|
Mishra A, Tabassum N, Aggarwal A, Kim YM, Khan F. Artificial Intelligence-Driven Analysis of Antimicrobial-Resistant and Biofilm-Forming Pathogens on Biotic and Abiotic Surfaces. Antibiotics (Basel) 2024; 13:788. [PMID: 39200087 PMCID: PMC11351874 DOI: 10.3390/antibiotics13080788] [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: 07/23/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024] Open
Abstract
The growing threat of antimicrobial-resistant (AMR) pathogens to human health worldwide emphasizes the need for more effective infection control strategies. Bacterial and fungal biofilms pose a major challenge in treating AMR pathogen infections. Biofilms are formed by pathogenic microbes encased in extracellular polymeric substances to confer protection from antimicrobials and the host immune system. Biofilms also promote the growth of antibiotic-resistant mutants and latent persister cells and thus complicate therapeutic approaches. Biofilms are ubiquitous and cause serious health risks due to their ability to colonize various surfaces, including human tissues, medical devices, and food-processing equipment. Detection and characterization of biofilms are crucial for prompt intervention and infection control. To this end, traditional approaches are often effective, yet they fail to identify the microbial species inside biofilms. Recent advances in artificial intelligence (AI) have provided new avenues to improve biofilm identification. Machine-learning algorithms and image-processing techniques have shown promise for the accurate and efficient detection of biofilm-forming microorganisms on biotic and abiotic surfaces. These advancements have the potential to transform biofilm research and clinical practice by allowing faster diagnosis and more tailored therapy. This comprehensive review focuses on the application of AI techniques for the identification of biofilm-forming pathogens in various industries, including healthcare, food safety, and agriculture. The review discusses the existing approaches, challenges, and potential applications of AI in biofilm research, with a particular focus on the role of AI in improving diagnostic capacities and guiding preventative actions. The synthesis of the current knowledge and future directions, as described in this review, will guide future research and development efforts in combating biofilm-associated infections.
Collapse
Affiliation(s)
- Akanksha Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144001, Punjab, India;
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; (N.T.); (Y.-M.K.)
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Ashish Aggarwal
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144001, Punjab, India;
| | - Young-Mog Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; (N.T.); (Y.-M.K.)
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; (N.T.); (Y.-M.K.)
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
- Institute of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea
- International Graduate Program of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea
| |
Collapse
|
14
|
Zhu L, Yang X, Fu X, Yang P, Lin X, Wang F, Shen Z, Wang J, Sun F, Qiu Z. Pheromone cCF10 inhibits the antibiotic persistence of Enterococcus faecalis by modulating energy metabolism. Front Microbiol 2024; 15:1408701. [PMID: 39040910 PMCID: PMC11260814 DOI: 10.3389/fmicb.2024.1408701] [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: 03/28/2024] [Accepted: 06/24/2024] [Indexed: 07/24/2024] Open
Abstract
Introduction Bacterial resistance presents a major challenge to both the ecological environment and human well-being, with persistence playing a key role. Multiple studies were recently undertaken to examine the factors influencing the formation of persisters and the underlying process, with a primary focus on Gram-negative bacteria and Staphylococcus aureus (Gram-positive bacteria). Enterococcus faecalis (E. faecalis) is capable of causing a variety of infectious diseases, but there have been few studies of E. faecalis persisters. Previous studies have shown that the sex pheromone cCF10 secreted by E. faecalis induces conjugative plasmid transfer. However, whether the pheromone cCF10 regulates the persistence of E. faecalis has not been investigated. Methods As a result, we investigated the effect and potential molecular mechanism of pheromone cCF10 in regulating the formation of persisters in E. faecalis OG1RF using a persistent bacteria model. Results and discussion The metabolically active E. faecalis OG1RF reached a persistence state and temporarily tolerated lethal antibiotic concentrations after 8 h of levofloxacin hydrochloride (20 mg/mL) exposure, exhibiting a persistence rate of 0.109 %. During the growth of E. faecalis OG1RF, biofilm formation was a critical factor contributing to antibiotic persistence, whereas 10 ng/mL cCF10 blocked persister cell formation. Notably, cCF10 mediated the antibiotic persistence of E. faecalis OG1RF via regulating metabolic activity rather than suppressing biofilm formation. The addition of cCF10 stimulated the Opp system and entered bacterial cells, inhibiting (p)ppGpp accumulation, thus maintaining the metabolically active state of bacteria and reducing persister cell generation. These findings offer valuable insights into the formation, as well as the control mechanism of E. faecalis persisters.
Collapse
Affiliation(s)
- Li Zhu
- School of Environmental and Chemical Engineering, Xi’an Polytechnic University, Xi’an, China
- Key Laboratory of Risk Assessment and Control for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Xiaobo Yang
- Key Laboratory of Risk Assessment and Control for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Xinyue Fu
- Key Laboratory of Risk Assessment and Control for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- College of Oceanography and Ecological Science, Shanghai Ocean University, Shanghai, China
| | - Panpan Yang
- Key Laboratory of Risk Assessment and Control for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Xiaoli Lin
- Key Laboratory of Risk Assessment and Control for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- Key Laboratory of Karst Geological Resources and Environment, Guizhou University, Guizhou, China
| | - Feng Wang
- Key Laboratory of Risk Assessment and Control for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- College of Oceanography and Ecological Science, Shanghai Ocean University, Shanghai, China
| | - Zhiqiang Shen
- Key Laboratory of Risk Assessment and Control for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Jingfeng Wang
- Key Laboratory of Risk Assessment and Control for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Feilong Sun
- School of Environmental and Chemical Engineering, Xi’an Polytechnic University, Xi’an, China
| | - Zhigang Qiu
- Key Laboratory of Risk Assessment and Control for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| |
Collapse
|
15
|
Petersen ME, Hansen LK, Mitkin AA, Kelly NM, Wood TK, Jørgensen NP, Østergaard LJ, Meyer RL. A high-throughput assay identifies molecules with antimicrobial activity against persister cells. J Med Microbiol 2024; 73:001856. [PMID: 38995832 PMCID: PMC11316564 DOI: 10.1099/jmm.0.001856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/13/2024] [Indexed: 07/14/2024] Open
Abstract
Introduction. Persister cells are transiently non-growing antibiotic-tolerant bacteria that cause infection relapse, and there is no effective antibiotic therapy to tackle these infections.Gap statement. High-throughput assays in drug discovery are biased towards detecting drugs that inhibit bacterial growth rather than killing non-growing bacteria. A new and simple assay to discover such drugs is needed.Aim. This study aims to develop a simple and high-throughput assay to identify compounds with antimicrobial activity against persister cells and use it to identify molecular motifs with such activity.Methodology. We quantified Staphylococcus aureus persister cells by enumeration of colony forming units after 24 h ciprofloxacin treatment. We first quantified how the cell concentration, antibiotic concentration, growth phase and presence/absence of nutrients during antibiotic exposure affected the fraction of persister cells in a population. After optimizing these parameters, we screened the antimicrobial activity of compound fragments to identify molecular structures that have activity against persister cells.Results. Exponential- and stationary-phase cultures transferred to nutrient-rich media displayed a bi-phasic time-kill curve and contained 0.001-0.07% persister cells. A short rifampicin treatment resulted in 100% persister cells for 7 h, after which cells resumed activity and became susceptible. Stationary-phase cultures displayed a low but constant death rate but ultimately resulted in similarly low survival rates as the exponential-phase cultures after 24 h ciprofloxacin treatment. The persister phenotype was only maintained in most of the population for 24 h if cells were transferred to a carbon-free minimal medium before exposure to ciprofloxacin. Keeping cells starved enabled the generation of high concentrations of S. aureus cells that tolerate 50× MIC ciprofloxacin, and we used this protocol for rapid screening for biocidal antibiotics. We identified seven compounds from four structural clusters with activity against antibiotic-tolerant S. aureus. Two compounds were moderately cytotoxic, and the rest were highly cytotoxic.Conclusion. Transferring a stationary-phase culture to a carbon-free minimal medium for antimicrobial testing is a simple strategy for high-throughput screening for new antibiotics that kill persister cells. We identified molecule fragments with such activity, but further screening is needed to identify motifs with lower general cytotoxicity.
Collapse
Affiliation(s)
| | - Liva Kjær Hansen
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, 8000 Aarhus C, Denmark
| | | | | | - Thomas Keith Wood
- Department of Chemical Engineering, Pennsylvania State University, University Park, USA
| | - Nis Pedersen Jørgensen
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus N, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Lars Jørgen Østergaard
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus N, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Rikke Louise Meyer
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, 8000 Aarhus C, Denmark
- Department of Biology, Aarhus University, 8000 Aarhus C, Denmark
| |
Collapse
|
16
|
Long DR, Holmes EA, Lo HY, Penewit K, Almazan J, Hodgson T, Berger NF, Bishop ZH, Lewis JD, Waalkes A, Wolter DJ, Salipante SJ. Clinical and in vitro models identify distinct adaptations enhancing Staphylococcus aureus pathogenesis in human macrophages. PLoS Pathog 2024; 20:e1012394. [PMID: 38991026 PMCID: PMC11265673 DOI: 10.1371/journal.ppat.1012394] [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: 02/16/2024] [Revised: 07/23/2024] [Accepted: 07/04/2024] [Indexed: 07/13/2024] Open
Abstract
Staphylococcus aureus is a facultative intracellular pathogen of human macrophages, which facilitates chronic infection. The genotypes, pathways, and mutations influencing that phenotype remain incompletely explored. Here, we used two distinct strategies to ascertain S. aureus gene mutations affecting pathogenesis in macrophages. First, we analyzed isolates collected serially from chronic cystic fibrosis (CF) respiratory infections. We found that S. aureus strains evolved greater macrophage invasion capacity during chronic human infection. Bacterial genome-wide association studies (GWAS) identified 127 candidate genes for which mutation was significantly associated with macrophage pathogenesis in vivo. In parallel, we passaged laboratory S. aureus strains in vitro to select for increased infection of human THP-1 derived macrophages, which identified 15 candidate genes by whole-genome sequencing. Functional validation of candidate genes using isogenic transposon mutant knockouts and CRISPR interference (CRISPRi) knockdowns confirmed virulence contributions from 37 of 39 tested genes (95%) implicated by in vivo studies and 7 of 10 genes (70%) ascertained from in vitro selection, with one gene in common to the two strategies. Validated genes included 17 known virulence factors (39%) and 27 newly identified by our study (61%), some encoding functions not previously associated with macrophage pathogenesis. Most genes (80%) positively impacted macrophage invasion when disrupted, consistent with the phenotype readily arising from loss-of-function mutations in vivo. This work reveals genes and mechanisms that contribute to S. aureus infection of macrophages, highlights differences in mutations underlying convergent phenotypes arising from in vivo and in vitro systems, and supports the relevance of S. aureus macrophage pathogenesis during chronic respiratory infection in CF. Additional studies will be needed to illuminate the exact mechanisms by which implicated mutations affect their phenotypes.
Collapse
Affiliation(s)
- Dustin R. Long
- Division of Critical Care Medicine, Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Elizabeth A. Holmes
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Hsin-Yu Lo
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Kelsi Penewit
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Jared Almazan
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Taylor Hodgson
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Nova F. Berger
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Zoe H. Bishop
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Janessa D. Lewis
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Adam Waalkes
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Daniel J. Wolter
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Stephen J. Salipante
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| |
Collapse
|
17
|
Zhang L, Yang J, Xu X, Zhang J, Qiu Z, Ju Y, Luo B, Liu Y, Gou X, Sui J, Chen B, Wang Y, Tao T, He L, Yang T, Luo Y. Discovery and Optimization of Novel SaFabI Inhibitors as Specific Therapeutic Agents for MRSA Infection. J Med Chem 2024; 67:10096-10134. [PMID: 38845361 DOI: 10.1021/acs.jmedchem.4c00320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
As the rate-limiting enzyme in fatty acid biosynthesis, Staphylococcus aureus enoyl-acyl carrier protein reductase (SaFabI) emerges as a compelling target for combating methicillin-resistant S. aureus (MRSA) infections. Herein, compound 1, featuring a 4-(1H-benzo[d]imidazol-2-yl)pyrrolidin-2-one scaffold, was identified as a potent SaFabI inhibitor (IC50 = 976.8 nM) from an in-house library. Subsequent optimization yielded compound n31, with improved inhibitory efficacy on enzymatic activity (IC50 = 174.2 nM) and selective potency against S. aureus (MIC = 1-2 μg/mL). Mechanistically, n31 directly inhibited SaFabI in cellular contexts. Moreover, n31 exhibited favorable safety and pharmacokinetic profiles, and dose-dependently treated MRSA-induced skin infections, outperforming the approved drug, linezolid. The chiral separation of n31 resulted in (S)-n31, with superior activities (IC50 = 94.0 nM, MIC = 0.25-1 μg/mL) and in vivo therapeutic efficacy. In brief, our research proposes (S)-n31 as a promising candidate for SaFabI-targeted therapy, offering specific anti-S. aureus efficacy and potential for further development.
Collapse
Affiliation(s)
- Laiying Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiaxing Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xin Xu
- Editorial Office of Chinese Journal of Medical Genetics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Jiangnan Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiqiang Qiu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuan Ju
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Baozhu Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yan Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xupeng Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Sui
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Baoyi Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanmei Wang
- Institute of traditional Chinese medicine, Sichuan College of Traditional Chinese Medicine, The Second Hospital of Traditional Chinese Medicine in Sichuan Province, Chengdu 610041, China
| | - Tao Tao
- Institute of traditional Chinese medicine, Sichuan College of Traditional Chinese Medicine, The Second Hospital of Traditional Chinese Medicine in Sichuan Province, Chengdu 610041, China
| | - Lei He
- Institute of traditional Chinese medicine, Sichuan College of Traditional Chinese Medicine, The Second Hospital of Traditional Chinese Medicine in Sichuan Province, Chengdu 610041, China
| | - Tao Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Youfu Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
18
|
Bernard C, Liu Y, Larrouy-Maumus G, Guilhot C, Cam K, Chalut C. Altered serine metabolism promotes drug tolerance in Mycobacterium abscessus via a WhiB7-mediated adaptive stress response. Antimicrob Agents Chemother 2024; 68:e0145623. [PMID: 38651855 PMCID: PMC11620514 DOI: 10.1128/aac.01456-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/31/2024] [Indexed: 04/25/2024] Open
Abstract
Mycobacterium abscessus is an emerging opportunistic pathogen responsible for chronic lung diseases, especially in patients with cystic fibrosis. Treatment failure of M. abscessus infections is primarily associated with intrinsic or acquired antibiotic resistance. However, there is growing evidence that antibiotic tolerance, i.e., the ability of bacteria to transiently survive exposure to bactericidal antibiotics through physiological adaptations, contributes to the relapse of chronic infections and the emergence of acquired drug resistance. Yet, our understanding of the molecular mechanisms that underlie antibiotic tolerance in M. abscessus remains limited. In the present work, a mutant with increased cross-tolerance to the first- and second-line antibiotics cefoxitin and moxifloxacin, respectively, has been isolated by experimental evolution. This mutant harbors a mutation in serB2, a gene involved in L-serine biosynthesis. Metabolic changes caused by this mutation alter the intracellular redox balance to a more reduced state that induces overexpression of the transcriptional regulator WhiB7 during the stationary phase, promoting tolerance through activation of a WhiB7-dependant adaptive stress response. These findings suggest that alteration of amino acid metabolism and, more generally, conditions that trigger whiB7 overexpression, makes M. abscessus more tolerant to antibiotic treatment.
Collapse
Affiliation(s)
- Célia Bernard
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France
| | - Yi Liu
- Faculty of Natural Sciences, Department of Life Sciences, Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Gérald Larrouy-Maumus
- Faculty of Natural Sciences, Department of Life Sciences, Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Christophe Guilhot
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France
| | - Kaymeuang Cam
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France
| | - Christian Chalut
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France
| |
Collapse
|
19
|
Landa G, Aguerri L, Irusta S, Mendoza G, Arruebo M. PLGA nanoparticle-encapsulated lysostaphin for the treatment of Staphylococcus aureus infections. Int J Biol Macromol 2024; 271:132563. [PMID: 38782313 DOI: 10.1016/j.ijbiomac.2024.132563] [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: 04/07/2024] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
Staphylococcus aureus possesses the ability to become pathogenic, leading to severe and life-threatening infections. Its methicillin-resistant variant MRSA has garnered high-priority status due to its increased morbidity and associated mortality. This emphasizes the urgency for novel anti-staphylococcal agents. The bacteriocin lysostaphin stands out for its remarkable bactericidal activity against S. aureus, including MRSA, outperforming conventional antibiotics. However, the clinical application of lysostaphin faces challenges, including enzymatic activity loss under physiological conditions and potential immunogenicity. This study introduces a novel approach by encapsulating lysostaphin within polylactic-co-glycolic acid (PLGA) nanoparticles, a biodegradable copolymer known for its biocompatibility and sustained drug release ability. The study assesses the antimicrobial activity of lysostaphin-loaded PLGA nanoparticles against different S. aureus strains, and we also used GFP-expressing S. aureus for facilitating its traceability in planktonic, biofilm, and intracellular infection models. The results showed the significant reduction in bacteria viability both in planktonic and biofilm states. The in vitro intracellular infection model demonstrated the significantly enhanced efficiency of the developed nanoparticles compared to the treatment with the free bacteriocin. This research presents lysostaphin encapsulation within PLGA nanoparticles and offers promising avenues for enhancing lysostaphin's therapeutic efficacy against S. aureus infections.
Collapse
Affiliation(s)
- Guillermo Landa
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain; Department of Chemical and Environmental Engineering, University of Zaragoza, Campus Río Ebro-Edificio I+D, C/Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain; Aragon Health Research Institute (IIS Aragon), 50009 Zaragoza, Spain.
| | - Laura Aguerri
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain; Department of Chemical and Environmental Engineering, University of Zaragoza, Campus Río Ebro-Edificio I+D, C/Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain
| | - Silvia Irusta
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain; Department of Chemical and Environmental Engineering, University of Zaragoza, Campus Río Ebro-Edificio I+D, C/Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain; Aragon Health Research Institute (IIS Aragon), 50009 Zaragoza, Spain
| | - Gracia Mendoza
- Aragon Health Research Institute (IIS Aragon), 50009 Zaragoza, Spain; Department of Pharmacology and Physiology, Forensic and Legal Medicine, University of Zaragoza, 50009 Zaragoza, Spain.
| | - Manuel Arruebo
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain; Department of Chemical and Environmental Engineering, University of Zaragoza, Campus Río Ebro-Edificio I+D, C/Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain; Aragon Health Research Institute (IIS Aragon), 50009 Zaragoza, Spain
| |
Collapse
|
20
|
Gattu R, Ramesh SS, Ramesh S. Role of small molecules and nanoparticles in effective inhibition of microbial biofilms: A ray of hope in combating microbial resistance. Microb Pathog 2024; 188:106543. [PMID: 38219923 DOI: 10.1016/j.micpath.2024.106543] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
Microbial biofilms pose a severe threat to global health, as they are associated with deadly chronic infections and antibiotic resistance. To date, very few drugs are in clinical practice that specifically target microbial biofilms. Therefore, there is an urgent need for the development of novel therapeutic options targeting biofilm-related infections. In this review, we discuss nearly seventy-five different molecular scaffolds published over the last decade (2010-2023) which have exhibited their biofilm inhibition potential. For convenience, we have classified these into five different sub-groups based on their origin and design (excluding peptides as they are placed in between small molecules and biologics), namely, heterocycles; inorganic small molecules & metal complexes; small molecules decorated nanoparticles; small molecules derived from natural products (both plant and marine sources); and small molecules designed by in-silico approach. These antibiofilm agents are capable of disrupting microbial biofilms and can offer a promising avenue for future developments in human medicine. A hitherto review of this kind will lay a platform for the researchers to find new molecular entities to curb the serious menace of antimicrobial resistance especially caused by biofilms.
Collapse
Affiliation(s)
- Rohith Gattu
- Postgraduate Department of Chemistry, JSS College of Arts, Commerce and Science (A Recognized Research Centre of University of Mysore), Ooty Road, Mysuru, 570025, Karnataka, India
| | - Sanjay S Ramesh
- Postgraduate Department of Chemistry, JSS College of Arts, Commerce and Science (A Recognized Research Centre of University of Mysore), Ooty Road, Mysuru, 570025, Karnataka, India
| | - Suhas Ramesh
- Postgraduate Department of Chemistry, JSS College of Arts, Commerce and Science (A Recognized Research Centre of University of Mysore), Ooty Road, Mysuru, 570025, Karnataka, India.
| |
Collapse
|
21
|
Karimaei S, Aghamir SMK, Pourmand MR. Comparative analysis of genes expression involved in type II toxin-antitoxin system in Staphylococcus aureus following persister cell formation. Mol Biol Rep 2024; 51:324. [PMID: 38393536 DOI: 10.1007/s11033-023-09179-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 12/18/2023] [Indexed: 02/25/2024]
Abstract
BACKGROUND The formation of persister cells is the main reason for persistent infections. They are associated with antibiotic treatment failure and subsequently chronic infection. The study aimed to assess the expression of type II toxin/antitoxin (TA) system genes in persister cells of Staphylococcus aureus in the presence of the following antibiotics vancomycin, ciprofloxacin, and gentamicin in exponential and stationary phases. METHODS AND RESULTS The colony count was used to evaluate the effect of different types of antibiotics on S. aureus persister cell formation during exponential and stationary phases. Moreover, the expression level of TA systems and clpP genes in the persister population in exponential and stationary phases were measured by quantitative reverse transcriptase real-time PCR (qRT-PCR). The results of the study showed the presence of persister phenotype of S. aureus strains in the attendance of bactericidal antibiotics in comparison to the control group during the exponential and stationary phases. Moreover, qRT-PCR resulted in the fact that the role of TA systems involved in the persister cell formation depends on the bacterial growth phase and the type of strain and antibiotic. CONCLUSIONS In total, the present study provides some data on the persister cell formation and the possible role of TA system genes in this process.
Collapse
Affiliation(s)
- Samira Karimaei
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Reza Pourmand
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Pathobiology, School of Public Health and Biotechnology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Luo ZX, Li Y, Liu MF, Zhao R. Ciprofloxacin enhances the biofilm formation of Staphylococcus aureus via an agrC-dependent mechanism. Front Microbiol 2023; 14:1328947. [PMID: 38179460 PMCID: PMC10764545 DOI: 10.3389/fmicb.2023.1328947] [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: 10/27/2023] [Accepted: 12/04/2023] [Indexed: 01/06/2024] Open
Abstract
Staphylococcus aureus readily forms biofilms on host tissues and medical devices, enabling its persistence in chronic infections and resistance to antibiotic therapy. The accessory gene regulator (Agr) quorum sensing system plays a key role in regulating S. aureus biofilm formation. This study reveals the widely used fluoroquinolone antibiotic, ciprofloxacin, strongly stimulates biofilm formation in methicillin-resistant S. aureus, methicillin-sensitive S. aureus, and clinical isolates with diverse genetic backgrounds. Crystal violet staining indicated that ciprofloxacin induced a remarkable 12.46- to 15.19-fold increase in biofilm biomass. Confocal laser scanning microscopy revealed that ciprofloxacin induced denser biofilms. Phenotypic assays suggest that ciprofloxacin may enhance polysaccharide intercellular adhesin production, inhibit autolysis, and reduce proteolysis during the biofilm development, thus promoting initial adhesion and enhancing biofilm stability. Mechanistically, ciprofloxacin significantly alters the expression of various biofilm-related genes (icaA, icaD, fnbA, fnbB, eap, emp) and regulators (agrA, saeR). Gene knockout experiments revealed that deletion of agrC, rather than saeRS, abolishes the ciprofloxacin-induced enhancement of biofilm formation, underscoring the key role of agrC. Thermal shift assays showed ciprofloxacin binds purified AgrC protein, thereby inhibiting the Agr system. Molecular docking results further support the potential interaction between ciprofloxacin and AgrC. In summary, subinhibitory concentrations of ciprofloxacin stimulate S. aureus biofilm formation via an agrC-dependent pathway. This inductive effect may facilitate local infection establishment and bacterial persistence, ultimately leading to therapeutic failure.
Collapse
Affiliation(s)
- Zhao-xia Luo
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, China
- School of Public Health, Nanchang University, Nanchang, China
| | - Yuting Li
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, China
- School of Public Health, Nanchang University, Nanchang, China
| | - Mei-fang Liu
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Rui Zhao
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
23
|
Hackemann VCJ, Hagel S, Jandt KD, Rödel J, Löffler B, Tuchscherr L. The Controversial Effect of Antibiotics on Methicillin-Sensitive S. aureus: A Comparative In Vitro Study. Int J Mol Sci 2023; 24:16308. [PMID: 38003500 PMCID: PMC10671744 DOI: 10.3390/ijms242216308] [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: 10/04/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Methicillin-sensitive Staphylococcus (S.) aureus (MSSA) bacteremia remains a global challenge, despite the availability of antibiotics. Primary treatments include β-lactam agents such as cefazolin and flucloxacillin. Ongoing discussions have focused on the potential synergistic effects of combining these agents with rifampicin or fosfomycin to combat infections associated with biofilm formation. Managing staphylococcal infections is challenging due to antibacterial resistance, biofilms, and S. aureus's ability to invade and replicate within host cells. Intracellular invasion shields the bacteria from antibacterial agents and the immune system, often leading to incomplete bacterial clearance and chronic infections. Additionally, S. aureus can assume a dormant phenotype, known as the small colony variant (SCV), further complicating eradication and promoting persistence. This study investigated the impact of antibiotic combinations on the persistence of S. aureus 6850 and its stable small colony variant (SCV strain JB1) focusing on intracellular survival and biofilm formation. The results from the wild-type strain 6850 demonstrate that β-lactams combined with RIF effectively eliminated biofilms and intracellular bacteria but tend to select for SCVs in planktonic culture and host cells. Higher antibiotic concentrations were associated with an increase in the zeta potential of S. aureus, suggesting reduced membrane permeability to antimicrobials. When using the stable SCV mutant strain JB1, antibiotic combinations with rifampicin successfully cleared planktonic bacteria and biofilms but failed to eradicate intracellular bacteria. Given these findings, it is reasonable to report that β-lactams combined with rifampicin represent the optimal treatment for MSSA bacteremia. However, caution is warranted when employing this treatment over an extended period, as it may elevate the risk of selecting for small colony variants (SCVs) and, consequently, promoting bacterial persistence.
Collapse
Affiliation(s)
| | - Stefan Hagel
- Institute of Infectious Diseases and Infection Control, Jena University Hospital, 07747 Jena, Germany
| | - Klaus D Jandt
- Otto Schott Institute of Materials Research (OSIM), Friedrich Schiller University Jena, 07743 Jena, Germany
- Jena School for Microbial Communication (JSMC), 07743 Jena, Germany
| | - Jürgen Rödel
- Institute for Medical Microbiology, Jena University Hospital, 07747 Jena, Germany
| | - Bettina Löffler
- Institute for Medical Microbiology, Jena University Hospital, 07747 Jena, Germany
| | - Lorena Tuchscherr
- Institute for Medical Microbiology, Jena University Hospital, 07747 Jena, Germany
| |
Collapse
|
24
|
Thompson NT, Kitzenberg DA, Kao DJ. Persister-mediated emergence of antimicrobial resistance in agriculture due to antibiotic growth promoters. AIMS Microbiol 2023; 9:738-756. [PMID: 38173975 PMCID: PMC10758577 DOI: 10.3934/microbiol.2023038] [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: 04/18/2023] [Revised: 10/16/2023] [Accepted: 11/02/2023] [Indexed: 01/05/2024] Open
Abstract
The creation and continued development of antibiotics have revolutionized human health and disease for the past century. The emergence of antimicrobial resistance represents a major threat to human health, and practices that contribute to the development of this threat need to be addressed. Since the 1950s, antibiotics have been used in low doses to increase growth and decrease the feed requirement of animal-derived food sources. A consequence of this practice is the accelerated emergence of antimicrobial resistance that can influence human health through its distribution via animal food products. In the laboratory setting, sublethal doses of antibiotics promote the expansion of bacterial persister populations, a low energy, low metabolism phenotype characterized broadly by antibiotic tolerance. Furthermore, the induction of persister bacteria has been positively correlated with an increased emergence of antibiotic-resistant strains. This body of evidence suggests that the use of antibiotics in agriculture at subtherapeutic levels is actively catalyzing the emergence of antimicrobial-resistant bacteria through the expansion of bacterial persister populations, which is potentially leading to increased infections in humans and decreased antibiotic potency. There is an urgent need to address this debilitating effect on antibiotics and its influence on human health. In this review, we summarize the recent literature on the topic of emerging antimicrobial resistance and its association with bacterial persister populations.
Collapse
Affiliation(s)
- Noah T Thompson
- Department of Medicine and Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - David A Kitzenberg
- Department of Medicine and Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Daniel J Kao
- Department of Medicine and Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
25
|
Durrani B, Mohammad A, Ljubetic BM, Dobberfuhl AD. The Potential Role of Persister Cells in Urinary Tract Infections. Curr Urol Rep 2023; 24:541-551. [PMID: 37907771 DOI: 10.1007/s11934-023-01182-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2023] [Indexed: 11/02/2023]
Abstract
PURPOSE OF REVIEW This review explores the role of persister cells in urinary tract infections (UTIs). UTIs are one of the most common bacterial infections, affecting millions of people worldwide. Persister cells are a subpopulation of bacteria with dormant metabolic activity which allows survival in the presence of antibiotics. RECENT FINDINGS This review summarizes recent research on the pathogenesis of persister cell formation in UTIs, the impact of persister cells on the effectiveness of antibiotics, the challenges they pose for treatment, and the need for new strategies to target these cells. Furthermore, this review examines the current state of research on the identification and characterization of persister cells in UTIs, as well as the future directions for investigations in this field. This review highlights the importance of understanding the role of persister cells in UTIs and the potential impact of targeting these cells in the development of new treatments.
Collapse
Affiliation(s)
- Butool Durrani
- Department of Internal Medicine, Aga Khan University Hospital, National Stadium Rd, Karachi, Karachi City, Pakistan
| | - Ashu Mohammad
- Department of Urology, Center for Academic Medicine, Stanford University School of Medicine, 453 Quarry Road, Urology-5656, Palo Alto, CA, 94304, USA
| | - Bernardita M Ljubetic
- Department of Urology, Center for Academic Medicine, Stanford University School of Medicine, 453 Quarry Road, Urology-5656, Palo Alto, CA, 94304, USA
| | - Amy D Dobberfuhl
- Department of Urology, Center for Academic Medicine, Stanford University School of Medicine, 453 Quarry Road, Urology-5656, Palo Alto, CA, 94304, USA.
| |
Collapse
|
26
|
Concilio M, Garcia Maset R, Lemonche LP, Kontrimas V, Song J, Rajendrakumar SK, Harrison F, Becer CR, Perrier S. Mechanism of Action of Oxazoline-Based Antimicrobial Polymers Against Staphylococcus aureus: In Vivo Antimicrobial Activity Evaluation. Adv Healthc Mater 2023; 12:e2301961. [PMID: 37522292 PMCID: PMC11468764 DOI: 10.1002/adhm.202301961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Indexed: 08/01/2023]
Abstract
Antimicrobial-resistant pathogens have reached alarming levels, becoming one of the most pressing global health issues. Hence, new treatments are necessary for the fight against antimicrobial resistance. Synthetic nanoengineered antimicrobial polymers (SNAPs) have emerged as a promising alternative to antimicrobial peptides, overcoming some of their limitations while keeping their key features. Herein, a library of amphiphilic oxazoline-based SNAPs using cationic ring-opening polymerization (CROP) is designed. Amphipathic compounds with 70% cationic content exhibit the highest activity against clinically relevant Staphylococcus aureus isolates, maintaining good biocompatibility in vitro and in vivo. The mechanism of action of the lead compounds against S. aureus is assessed using various microscopy techniques, indicating cell membrane disruption, while the cell wall remains unaffected. Furthermore, a potential interaction of the compounds with bacterial DNA is shown, with possible implications on bacterial division. Finally, one of the compounds exhibits high efficacy in vivo in an insect infection model.
Collapse
Affiliation(s)
| | - Ramón Garcia Maset
- Department of ChemistryUniversity of WarwickCoventryCV4 7ALUK
- Warwick Medical SchoolUniversity of WarwickCoventryCV4 7ALUK
| | | | - Vito Kontrimas
- Department of ChemistryUniversity of WarwickCoventryCV4 7ALUK
- Warwick Medical SchoolUniversity of WarwickCoventryCV4 7ALUK
| | - Ji‐Inn Song
- Department of ChemistryUniversity of WarwickCoventryCV4 7ALUK
| | | | - Freya Harrison
- School of Life SciencesUniversity of WarwickCoventryCV4 7ALUK
| | - C. Remzi Becer
- Department of ChemistryUniversity of WarwickCoventryCV4 7ALUK
| | - Sébastien Perrier
- Department of ChemistryUniversity of WarwickCoventryCV4 7ALUK
- Warwick Medical SchoolUniversity of WarwickCoventryCV4 7ALUK
- Faculty of Pharmacy and Pharmaceutical SciencesMonash UniversityParkvilleVictoria3052Australia
| |
Collapse
|
27
|
Gehrke AKE, Giai C, Gómez MI. Staphylococcus aureus Adaptation to the Skin in Health and Persistent/Recurrent Infections. Antibiotics (Basel) 2023; 12:1520. [PMID: 37887220 PMCID: PMC10604630 DOI: 10.3390/antibiotics12101520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/28/2023] Open
Abstract
Staphylococcus aureus is a microorganism with an incredible capability to adapt to different niches within the human body. Approximately between 20 and 30% of the population is permanently but asymptomatically colonized with S. aureus in the nose, and another 30% may carry S. aureus intermittently. It has been established that nasal colonization is a risk factor for infection in other body sites, including mild to severe skin and soft tissue infections. The skin has distinct features that make it a hostile niche for many bacteria, therefore acting as a strong barrier against invading microorganisms. Healthy skin is desiccated; it has a low pH at the surface; the upper layer is constantly shed to remove attached bacteria; and several host antimicrobial peptides are produced. However, S. aureus is able to overcome these defenses and colonize this microenvironment. Moreover, this bacterium can very efficiently adapt to the stressors present in the skin under pathological conditions, as it occurs in patients with atopic dermatitis or suffering chronic wounds associated with diabetes. The focus of this manuscript is to revise the current knowledge concerning how S. aureus adapts to such diverse skin conditions causing persistent and recurrent infections.
Collapse
Affiliation(s)
- Ana-Katharina E. Gehrke
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Departamento de Investigaciones Biomédicas y Biotecnológicas, Universidad Maimónides, Buenos Aires C1405BCK, Argentina;
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1425FQB, Argentina
| | - Constanza Giai
- Instituto de Histología y Embriología de Mendoza, Universidad Nacional de Cuyo—(UNCuyo) CONICET, Mendoza M5502JMA, Argentina;
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza M5502JMA, Argentina
- Facultad de Farmacia y Bioquímica, Universidad Juan Agustín Maza, Mendoza C1006ACC, Argentina
| | - Marisa I. Gómez
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Departamento de Investigaciones Biomédicas y Biotecnológicas, Universidad Maimónides, Buenos Aires C1405BCK, Argentina;
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1425FQB, Argentina
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121A6B, Argentina
| |
Collapse
|
28
|
Pan X, Liu W, Du Q, Zhang H, Han D. Recent Advances in Bacterial Persistence Mechanisms. Int J Mol Sci 2023; 24:14311. [PMID: 37762613 PMCID: PMC10531727 DOI: 10.3390/ijms241814311] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
The recurrence of bacterial infectious diseases is closely associated with bacterial persisters. This subpopulation of bacteria can escape antibiotic treatment by entering a metabolic status of low activity through various mechanisms, for example, biofilm, toxin-antitoxin modules, the stringent response, and the SOS response. Correspondingly, multiple new treatments are being developed. However, due to their spontaneous low abundance in populations and the lack of research on in vivo interactions between persisters and the host's immune system, microfluidics, high-throughput sequencing, and microscopy techniques are combined innovatively to explore the mechanisms of persister formation and maintenance at the single-cell level. Here, we outline the main mechanisms of persister formation, and describe the cutting-edge technology for further research. Despite the significant progress regarding study techniques, some challenges remain to be tackled.
Collapse
Affiliation(s)
- Xiaozhou Pan
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Wenxin Liu
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Qingqing Du
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Hong Zhang
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Dingding Han
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| |
Collapse
|
29
|
Wang C, Jin L. Microbial persisters and host: recent advances and future perspectives. Crit Rev Microbiol 2023; 49:658-670. [PMID: 36165023 DOI: 10.1080/1040841x.2022.2125286] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 09/07/2022] [Accepted: 09/12/2022] [Indexed: 11/03/2022]
Abstract
Microbial persisters are defined as the tiny sub-population of microorganisms that develop intrinsic strategies for survival with high tolerance to various antimicrobials. Currently, persister research remains in its infancy, and it is indeed a great challenge to precisely distinguish persister cells from other drug tolerant ones. Notably, the existence of persisters crucially contributes to prolonged antibiotic exposure time and treatment failure, yet there is the formation of antibiotic-resistant mutants. Further understanding on persisters is of profound importance for effective prevention and control of chronic infections/inflammation. The past two decades have witnessed rapid advances on the science, technologies and methodologies for persister investigations, along with deep knowledge about persisters and numerous anti-persister approaches developed. Whereas, various critical issues remain unsolved, such as what are the potential interaction profiles of persisters and host cells, and how to apply what we know about persisters to translational studies and clinical practice. Importantly, it is highly essential to better understand the multifaceted and complex cross-talk of microbial persisters with the host to develop novel tackling strategies for precision healthcare in the near future.
Collapse
Affiliation(s)
- Chuan Wang
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | | |
Collapse
|
30
|
Zhou Y, Liao H, Pei L, Pu Y. Combatting persister cells: The daunting task in post-antibiotics era. CELL INSIGHT 2023; 2:100104. [PMID: 37304393 PMCID: PMC10250163 DOI: 10.1016/j.cellin.2023.100104] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/25/2023] [Accepted: 04/21/2023] [Indexed: 06/13/2023]
Abstract
Over the years, much attention has been drawn to antibiotic resistance bacteria, but drug inefficacy caused by a subgroup of special phenotypic variants - persisters - has been largely neglected in both scientific and clinical field. Interestingly, this subgroup of phenotypic variants displayed their power of withstanding sufficient antibiotics exposure in a mechanism different from antibiotic resistance. In this review, we summarized the clinical importance of bacterial persisters, the evolutionary link between resistance, tolerance, and persistence, redundant mechanisms of persister formation as well as methods of studying persister cells. In the light of our recent findings of membrane-less organelle aggresome and its important roles in regulating bacterial dormancy depth, we propose an alternative approach for anti-persister therapy. That is, to force a persister into a deeper dormancy state to become a VBNC (viable but non-culturable) cell that is incapable of regrowth. We hope to provide the latest insights on persister studies and call upon more research interest into this field.
Collapse
Affiliation(s)
- Yidan Zhou
- Department of Clinical Laboratory, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei- MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430079, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430079, China
| | - Hebin Liao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei- MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430079, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430079, China
| | - Linsen Pei
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei- MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430079, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430079, China
| | - Yingying Pu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei- MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430079, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
31
|
Wang M, Zhang Y, Niu Z, Miao Q, Fu W. Study on the distribution characteristics and metabolic mechanism of chlorine-resistant bacteria in indoor water supply networks. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 328:121640. [PMID: 37059174 DOI: 10.1016/j.envpol.2023.121640] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/31/2023] [Accepted: 04/12/2023] [Indexed: 05/09/2023]
Abstract
The presence and attachment of chlorine-resistant bacteria on the surface of water distribution network will deteriorate water quality and threaten human health. Chlorination is critical in drinking water treatment to ensure the biosafety of drinking water. However, how disinfectants affect the structures of dominant flora during biofilm development and whether the changes are consistent with the free flora remain unclear. Therefore, we investigated changes in species diversity and relative abundance of different bacterial communities in planktonic and biofilm samples at different chlorine residual concentrations (blank, 0.3 mg/L, 0.8 mg/L, 2.0 mg/L and 4.0 mg/L), and the main reasons for the development of chlorine resistance in bacteria was also discussed. The results showed that the richness of microbial species in the biofilm was higher than that in planktonic microbial samples. In the planktonic samples, Proteobacteria and Actinobacteria were the dominant groups regardless of the chlorine residual concentration. For biofilm samples, the dominant position of Proteobacteria bacteria was gradually replaced by actinobacteria bacteria with the increase of chlorine residual concentration. In addition, at higher chlorine residual concentration, Gram-positive bacteria were more concentrated to form biofilms. There are three main reasons for the generation of chlorine resistance of bacteria: enhanced function of efflux system, activated bacterial self-repair system, and enhanced nutrient uptake capacity.
Collapse
Affiliation(s)
- Mengyuan Wang
- School of Environmental Science and Engineering, Tianjin University, Tianjin, 300350, China; College of Environmental Science and Engineering, Nankai University, Tianjin, 300350, China
| | - Ying Zhang
- School of Environmental Science and Engineering, Tianjin University, Tianjin, 300350, China.
| | - Zhiguang Niu
- School of Environmental Science and Engineering, Tianjin University, Tianjin, 300350, China; The International Joint Institute of Tianjin University, Fuzhou, 350207, China
| | - Qinkui Miao
- College of Environmental Science and Engineering, Nankai University, Tianjin, 300350, China
| | - Wei Fu
- College of Environmental Science and Engineering, Nankai University, Tianjin, 300350, China
| |
Collapse
|
32
|
Theis TJ, Daubert TA, Kluthe KE, Brodd KL, Nuxoll AS. Staphylococcus aureus persisters are associated with reduced clearance in a catheter-associated biofilm infection. Front Cell Infect Microbiol 2023; 13:1178526. [PMID: 37228667 PMCID: PMC10203555 DOI: 10.3389/fcimb.2023.1178526] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
Background Staphylococcus aureus causes a wide variety of infections, many of which are chronic or relapsing in nature. Antibiotic therapy is often ineffective against S. aureus biofilm-mediated infections. Biofilms are difficult to treat partly due to their tolerance to antibiotics, however the underlying mechanism responsible for this remains unknown. One possible explanation is the presence of persister cells-dormant-like cells that exhibit tolerance to antibiotics. Recent studies have shown a connection between a fumC (fumarase C, a gene in the tricarboxylic acid cycle) knockout strain and increased survival to antibiotics, antimicrobial peptides, and in a Drosophila melanogaster model. Objective It remained unclear whether a S. aureus high persister strain would have a survival advantage in the presence of innate and adaptive immunity. To further investigate this, a fumC knockout and wild type strains were examined in a murine catheter-associated biofilm model. Results Interestingly, mice struggled to clear both S. aureus wild type and the fumC knockout strains. We reasoned both biofilm-mediated infections predominantly consisted of persister cells. To determine the persister cell population within biofilms, expression of a persister cell marker (Pcap5A::dsRED) in a biofilm was examined. Cell sorting of biofilms challenged with antibiotics revealed cells with intermediate and high expression of cap5A had 5.9-and 4.5-fold higher percent survival compared to cells with low cap5A expression. Based on previous findings that persisters are associated with reduced membrane potential, flow cytometry analysis was used to examine the metabolic state of cells within a biofilm. We confirmed cells within biofilms had reduced membrane potential compared to both stationary phase cultures (2.5-fold) and exponential phase cultures (22.4-fold). Supporting these findings, cells within a biofilm still exhibited tolerance to antibiotic challenge following dispersal of the matrix through proteinase K. Conclusion Collectively, these data show that biofilms are largely comprised of persister cells, and this may explain why biofilm infections are often chronic and/or relapsing in clinical settings.
Collapse
|
33
|
Kamer AMA, Abdelaziz AA, Al-Monofy KB, Al-Madboly LA. Antibacterial, antibiofilm, and anti-quorum sensing activities of pyocyanin against methicillin-resistant Staphylococcus aureus: in vitro and in vivo study. BMC Microbiol 2023; 23:116. [PMID: 37095436 PMCID: PMC10124065 DOI: 10.1186/s12866-023-02861-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/13/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND Methicillin-resistant Staphylococcus aureus (MRSA) infections are considered a major public health problem, as the treatment options are restricted. Biofilm formation and the quorum sensing (QS) system play a pivotal role in S. aureus pathogenicity. Hence, this study was performed to explore the antibacterial effect of pyocyanin (PCN) on MRSA as well as its effect on MRSA biofilm and QS. RESULTS Data revealed that PCN exhibited strong antibacterial activity against all test MRSA isolates (n = 30) with a MIC value equal to 8 µg/ml. About 88% of MRSA biofilms were eradicated by PCN treatment using the crystal violet assay. The disruption of MRSA biofilm was confirmed using confocal laser scanning microscopy, which showed a reduction in bacterial viability (approximately equal to 82%) and biofilm thickness (approximately equal to 60%). Additionally, the disruption of the formation of microcolonies and the disturbance of the connection between bacterial cells in the MRSA biofilm after PCN treatment were examined by scanning electron microscopy. The 1/2 and 1/4 MICs of PCN exerted promising anti-QS activity without affecting bacterial viability; Agr QS-dependent virulence factors (hemolysin, protease, and motility), and the expression of agrA gene, decreased after PCN treatment. The in silico analysis confirmed the binding of PCN to the AgrA protein active site, which blocked its action. The in vivo study using the rat wound infection model confirmed the ability of PCN to modulate the biofilm and QS of MRSA isolates. CONCLUSION The extracted PCN seems to be a good candidate for treating MRSA infection through biofilm eradication and Agr QS inhibition.
Collapse
Affiliation(s)
- Amal M Abo Kamer
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Ahmed A Abdelaziz
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Khaled B Al-Monofy
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| | - Lamiaa A Al-Madboly
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
34
|
Long DR, Penewit K, Lo HY, Almazan J, Holmes EA, Bryan AB, Wolter DJ, Lewis JD, Waalkes A, Salipante SJ. In Vitro Selection Identifies Staphylococcus aureus Genes Influencing Biofilm Formation. Infect Immun 2023; 91:e0053822. [PMID: 36847490 PMCID: PMC10016075 DOI: 10.1128/iai.00538-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/13/2023] [Indexed: 03/01/2023] Open
Abstract
Staphylococcus aureus generates biofilms during many chronic human infections, which contributes to its growth and persistence in the host. Multiple genes and pathways necessary for S. aureus biofilm production have been identified, but knowledge is incomplete, and little is known about spontaneous mutations that increase biofilm formation as infection progresses. Here, we performed in vitro selection of four S. aureus laboratory strains (ATCC 29213, JE2, N315, and Newman) to identify mutations associated with enhanced biofilm production. Biofilm formation increased in passaged isolates from all strains, exhibiting from 1.2- to 5-fold the capacity of parental lines. Whole-genome sequencing identified nonsynonymous mutations affecting 23 candidate genes and a genomic duplication encompassing sigB. Six candidate genes significantly impacted biofilm formation as isogenic transposon knockouts: three were previously reported to impact S. aureus biofilm formation (icaR, spdC, and codY), while the remaining three (manA, narH, and fruB) were newly implicated by this study. Plasmid-mediated genetic complementation of manA, narH, and fruB transposon mutants corrected biofilm deficiencies, with high-level expression of manA and fruB further enhancing biofilm formation over basal levels. This work recognizes genes not previously identified as contributing to biofilm formation in S. aureus and reveals genetic changes able to augment biofilm production by that organism.
Collapse
Affiliation(s)
- Dustin R. Long
- Division of Critical Care Medicine, Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Kelsi Penewit
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Hsin-Yu Lo
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Jared Almazan
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Elizabeth A. Holmes
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Andrew B. Bryan
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Daniel J. Wolter
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
| | - Janessa D. Lewis
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Adam Waalkes
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Stephen J. Salipante
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
35
|
Diban F, Di Lodovico S, Di Fermo P, D’Ercole S, D’Arcangelo S, Di Giulio M, Cellini L. Biofilms in Chronic Wound Infections: Innovative Antimicrobial Approaches Using the In Vitro Lubbock Chronic Wound Biofilm Model. Int J Mol Sci 2023; 24:1004. [PMID: 36674518 PMCID: PMC9862456 DOI: 10.3390/ijms24021004] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023] Open
Abstract
Chronic wounds have harmful effects on both patients and healthcare systems. Wound chronicity is attributed to an impaired healing process due to several host and local factors that affect healing pathways. The resulting ulcers contain a wide variety of microorganisms that are mostly resistant to antimicrobials and possess the ability to form mono/poly-microbial biofilms. The search for new, effective and safe compounds to handle chronic wounds has come a long way throughout the history of medicine, which has included several studies and trials of conventional treatments. Treatments focus on fighting the microbial colonization that develops in the wound by multidrug resistant pathogens. The development of molecular medicine, especially in antibacterial agents, needs an in vitro model similar to the in vivo chronic wound environment to evaluate the efficacy of antimicrobial agents. The Lubbock chronic wound biofilm (LCWB) model is an in vitro model developed to mimic the pathogen colonization and the biofilm formation of a real chronic wound, and it is suitable to screen the antibacterial activity of innovative compounds. In this review, we focused on the characteristics of chronic wound biofilms and the contribution of the LCWB model both to the study of wound poly-microbial biofilms and as a model for novel treatment strategies.
Collapse
Affiliation(s)
- Firas Diban
- Department of Pharmacy, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy
| | - Silvia Di Lodovico
- Department of Pharmacy, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy
| | - Paola Di Fermo
- Department of Pharmacy, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy
| | - Simonetta D’Ercole
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy
| | - Sara D’Arcangelo
- Department of Pharmacy, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy
| | - Mara Di Giulio
- Department of Pharmacy, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy
| | - Luigina Cellini
- Department of Pharmacy, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
36
|
Huemer M, Mairpady Shambat S, Hertegonne S, Bergada-Pijuan J, Chang CC, Pereira S, Gómez-Mejia A, Van Gestel L, Bär J, Vulin C, Pfammatter S, Stinear TP, Monk IR, Dworkin J, Zinkernagel AS. Serine-threonine phosphoregulation by PknB and Stp contributes to quiescence and antibiotic tolerance in Staphylococcus aureus. Sci Signal 2023; 16:eabj8194. [PMID: 36595572 DOI: 10.1126/scisignal.abj8194] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 12/06/2022] [Indexed: 01/05/2023]
Abstract
Staphylococcus aureus can cause infections that are often chronic and difficult to treat, even when the bacteria are not antibiotic resistant because most antibiotics act only on metabolically active cells. Subpopulations of persister cells are metabolically quiescent, a state associated with delayed growth, reduced protein synthesis, and increased tolerance to antibiotics. Serine-threonine kinases and phosphatases similar to those found in eukaryotes can fine-tune essential bacterial cellular processes, such as metabolism and stress signaling. We found that acid stress-mimicking conditions that S. aureus experiences in host tissues delayed growth, globally altered the serine and threonine phosphoproteome, and increased threonine phosphorylation of the activation loop of the serine-threonine protein kinase B (PknB). The deletion of stp, which encodes the only annotated functional serine-threonine phosphatase in S. aureus, increased the growth delay and phenotypic heterogeneity under different stress challenges, including growth in acidic conditions, the intracellular milieu of human cells, and abscesses in mice. This growth delay was associated with reduced protein translation and intracellular ATP concentrations and increased antibiotic tolerance. Using phosphopeptide enrichment and mass spectrometry-based proteomics, we identified targets of serine-threonine phosphorylation that may regulate bacterial growth and metabolism. Together, our findings highlight the importance of phosphoregulation in mediating bacterial quiescence and antibiotic tolerance and suggest that targeting PknB or Stp might offer a future therapeutic strategy to prevent persister formation during S. aureus infections.
Collapse
Affiliation(s)
- Markus Huemer
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Srikanth Mairpady Shambat
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sanne Hertegonne
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Judith Bergada-Pijuan
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Chun-Chi Chang
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sandro Pereira
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Alejandro Gómez-Mejia
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Lies Van Gestel
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Julian Bär
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Clément Vulin
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sibylle Pfammatter
- Functional Genomics Center Zurich, ETH/University of Zurich, Zurich, Switzerland
| | - Timothy P Stinear
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Ian R Monk
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Jonathan Dworkin
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Annelies S Zinkernagel
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
37
|
Nwabuife JC, Hassan D, Madhaorao Pant A, Devnarain N, Gafar MA, Osman N, Rambharose S, Govender T. Novel vancomycin free base – Sterosomes for combating diseases caused by Staphylococcus aureus and Methicillin-resistant Staphylococcus aureus infections (S. Aureus and MRSA). J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2022.104089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
38
|
Muacevic A, Adler JR, Sano C, Ohta R. Subacute Bacterial Cellulitis With a Subacute Clinical Course With Difficulty in Distinguishing From Sjögren's Syndrome: A Case Report. Cureus 2023; 15:e33554. [PMID: 36779117 PMCID: PMC9908298 DOI: 10.7759/cureus.33554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Differentiating between infectious and autoimmune diseases regarding inflammatory changes in the soft tissues around the joints that develop in a subacute course is often difficult. Herein, we present the case of a 65-year-old woman who presented with a chief complaint of the tarsal metatarsal joint (Lisfranc joint) pain in the second, third, and fourth toes of her right foot that had persisted for several days. The patient was initially treated with non-steroidal anti-inflammatory drugs, colchicine, and prednisolone 10 mg for pseudogout and Sjogren's syndrome arthritis; however, there was little improvement. A few weeks later, the skin of the fingers and toes peeled, and the patient was diagnosed with subacute burn-like skin syndrome and treated with antibiotics. After the treatment, the inflammatory findings steadily improved. Arthritis associated with an infection is considered to have an acute course. However, subacute-to-chronic arthritis associated with Staphylococcus aureus infection may also be possible, as in this case. Therefore, the possibility of infection should be evaluated in patients with subacute-to-chronic refractory arthritis.
Collapse
|
39
|
Rapid Bactericidal Activity of SC5005 Combined with Docosahexaenoic Acid against Multidrug-Resistant Staphylococcus aureus Persisters and Biofilms. Antimicrob Agents Chemother 2022; 66:e0080322. [PMID: 36354314 PMCID: PMC9764969 DOI: 10.1128/aac.00803-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Staphylococcus aureus can form persister cells and biofilms, making the treatment difficult and often leading to recurrent infections. In an effort to discover new anti-staphylococcal agents, we observed that oleic acid enhances the activity of a new antibacterial agent, SC5005, against S. aureus and MRSA strains. Subsequent studies showed that saturated or trans-form unsaturated fatty acids did not potentiate SC5005's antibacterial activity. SC5005 only exhibits synergistic bactericidal activity with cis-form unsaturated fatty acids with 16 to 22 carbon atoms. In particular, docosahexaenoic acid (DHA) could reduce the MIC of SC5005 to the subng/mL range against different MRSA strains, including those resistant to second- and third-line antibiotics. However, we did not detect any significant shift in SC5005's cytotoxicity toward four different mammalian cell lines, suggesting that the synergy of DHA and SC5005 is highly selective. Most importantly, this combination demonstrated fast-killing activity, completely eradicating MRSA USA300 planktonic and persister cells within 10 and 30 min, respectively, and removing nearly 98% of MRSA biofilms within 1 min. Together, our findings suggest that the combination of SC5005 and DHA has great potential as a new therapeutic for the treatment of infections caused by multidrug-resistant (MDR) S. aureus biofilms.
Collapse
|
40
|
PurN Is Involved in Antibiotic Tolerance and Virulence in Staphylococcus aureus. Antibiotics (Basel) 2022; 11:antibiotics11121702. [PMID: 36551359 PMCID: PMC9774800 DOI: 10.3390/antibiotics11121702] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/13/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
Staphylococcus aureus can cause chronic infections which are closely related to persister formation. Purine metabolism is involved in S. aureus persister formation, and purN, encoding phosphoribosylglycinamide formyltransferase, is an important gene in the purine metabolism process. In this study, we generated a ΔpurN mutant of the S. aureus Newman strain and assessed its roles in antibiotic tolerance and virulence. The ΔpurN in the late exponential phase had a significant defect in persistence to antibiotics. Complementation of the ΔpurN restored its tolerance to different antibiotics. PurN significantly affected virulence gene expression, hemolytic ability, and biofilm formation in S. aureus. Moreover, the LD50 (3.28 × 1010 CFU/mL) of the ΔpurN for BALB/c mice was significantly higher than that of the parental strain (2.81 × 109 CFU/mL). Transcriptome analysis revealed that 58 genes that were involved in purine metabolism, alanine, aspartate, glutamate metabolism, and 2-oxocarboxylic acid metabolism, etc., were downregulated, while 24 genes involved in ABC transporter and transferase activity were upregulated in ΔpurN vs. parental strain. Protein-protein interaction network showed that there was a close relationship between PurN and GltB, and SaeRS. The study demonstrated that PurN participates in the formation of the late exponential phase S. aureus persisters via GltB and regulates its virulence by activating the SaeRS two-component system.
Collapse
|
41
|
Pamukçu A, Erdoğan N, Şen Karaman D. Polyethylenimine-grafted mesoporous silica nanocarriers markedly enhance the bactericidal effect of curcumin against Staphylococcus aureus biofilm. J Biomed Mater Res B Appl Biomater 2022; 110:2506-2520. [PMID: 35735075 PMCID: PMC9541607 DOI: 10.1002/jbm.b.35108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/16/2022] [Accepted: 05/31/2022] [Indexed: 11/12/2022]
Abstract
The recalcitrant nature of biofilms makes biofilm-associated infections difficult to treat in modern medicine. Biofilms have a high vulnerability to antibiotics and a limited repertoire of antibiotics could act on matured biofilms. This issue has resulted in a gradual paradigm shift in drug discovery and therapy, with anti-biofilm compounds being sought alongside new drug carriers. A potential solution to biofilm-associated infections is to employ antibiofilm treatments, which can attack biofilms from many fronts. Nanocarriers are promising in this regard because they can be entrapped within biofilm matrix, target biofilm matrix, and provide local drug delivery to inhibit biofilm formation. In this study, curcumin as an herbal extract was loaded onto hyperbranched polyethylenimine-grafted mesoporous silica nanoparticles (F-MSN-PEI/Cur) and antibiofilm investigations were performed. The F-MSN-PEI/Cur design has the potential to repurpose curcumin as an antibiofilm agent by increasing its solubility and lowering the required doses for the destruction of matured biofilms as well as suppressing biofilm development. Using imaging and spectroscopic techniques, we assessed the interaction of F-MSN-PEI/Cur with Staphylococcus aureus bacterial cells and determined the impact of F-MSN-PEI/Cur on eradicating matured biofilms and suppressing biofilm development. The F-MSN-PEI/Cur design is highly cytocompatible, as observed by the cytotoxicity screening investigations on L929 mouse fibroblast cell line. Our findings show that F-MSN-PEI/Cur design reduces the bacterial cell viability, inhibits biofilm formation, and induces biofilm eradication, which is attributed to F-MSN-PEI/Cur design having the potential to repurpose the antibiofilm activity of curcumin-herbal extract.
Collapse
Affiliation(s)
- Ayşenur Pamukçu
- Department of Biomedical Technologies, Graduate School of Natural and Applied SciencesIzmir Katip Çelebi UniversityIzmirTurkey
| | - Nursu Erdoğan
- Department of Biomedical Technologies, Graduate School of Natural and Applied SciencesIzmir Katip Çelebi UniversityIzmirTurkey
| | - Didem Şen Karaman
- Department of Biomedical Engineering, Faculty of Engineering and ArchitectureIzmir Katip Çelebi UniversityIzmirTurkey
- Pharmaceutical Sciences Laboratory, Faculty of Science and EngineeringÅbo Akademi UniversityFinland
| |
Collapse
|
42
|
Keim KC, George IK, Reynolds L, Smith AC. The Clinical Significance of Staphylococcus aureus Small Colony Variants. Lab Med 2022; 54:227-234. [PMID: 36226897 DOI: 10.1093/labmed/lmac101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Abstract
A burdensome, atypical phenotype of Staphylococcus aureus (SA) called S aureus small colony variant (SA-SCV) has been identified, which is induced as a result of a combination of environmental stressors, including polymicrobial interactions. The SA-SCVs exhibit altered phenotypes as a result of metabolic dormancy caused by electron transport deficiency, leading to increased biofilm production and alterations to antimicrobial susceptibility. The SA-SCVs typically exhibit altered colony morphology and biochemical reactions compared with wild-type SA, making them difficult to detect via routine diagnostics. The SA-SCVs have been found to contribute to chronic or recurrent infections, including skin and soft-tissue infections, foreign-body associated infection, cystic fibrosis, and sepsis. There is evidence that SA-SCVs contribute to patient morbidity and mortality as a result of diagnostic difficulties and limited treatment options. New detection methods may need to be developed that can be incorporated into routine diagnostics, which would allow for better assessment of specimens and introduce new considerations for treatment.
Collapse
Affiliation(s)
- Klara C Keim
- Department of Immunology and Microbiology, School of Medicine, Anschutz Medical Campus, University of Colorado , Aurora, CO , USA
| | - Isaiah K George
- Department of Honors Studies, Texas Tech University , Lubbock, TX , USA
| | - Landrye Reynolds
- Department of Honors Studies, Texas Tech University , Lubbock, TX , USA
| | - Allie C Smith
- Department of Honors Studies, Texas Tech University , Lubbock, TX , USA
| |
Collapse
|
43
|
Jia J, Xue X, Guan Y, Fan X, Wang Z. Biofilm characteristics and transcriptomic profiling of Acinetobacter johnsonii defines signatures for planktonic and biofilm cells. ENVIRONMENTAL RESEARCH 2022; 213:113714. [PMID: 35718162 DOI: 10.1016/j.envres.2022.113714] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 06/11/2022] [Accepted: 06/14/2022] [Indexed: 06/15/2023]
Abstract
Most bacteria in the natural environment have a biofilm mode of life, which is intrinsically tolerant to antibiotics. While until now, the knowledge of biofilm formation by Acinetobacter johnsonii is not well understood. In this study, the characteristics and the effect of a sub-inhibitory concentration of antibiotic on A. johnsonii biofilm and planktonic cells were determined. We discovered a positive relationship between biofilm formation and tetracycline resistance, and biofilms rapidly evolve resistance to tetracycline they are treated with. Persister cells commonly exist in both planktonic and biofilm cells, with a higher frequency in the latter. Further transcriptomic analysis speculates that the overexpression of multidrug resistance genes and stress genes were mainly answered to sub lethal concentration of tetracycline in planktonic cells, and the lower metabolic levels after biofilm formation result in high resistance level of biofilm cells to tetracycline. Altogether, these data suggest that A. johnsonii can adjust its phenotype when grown as biofilm and change its metabolism under antibiotic stress, and provide implications for subsequent biofilm control.
Collapse
Affiliation(s)
- Jia Jia
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xue Xue
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yongjing Guan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xiaoteng Fan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Zaizhao Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
44
|
Barajas-Mendiola MA, Salgado-Lora MG, López-Meza JE, Ochoa-Zarzosa A. Prolactin regulates H3K9ac and H3K9me2 epigenetic marks and miRNAs expression in bovine mammary epithelial cells challenged with Staphylococcus aureus. Front Microbiol 2022; 13:990478. [PMID: 36212825 PMCID: PMC9539446 DOI: 10.3389/fmicb.2022.990478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Epigenetic mechanisms are essential in the regulation of immune response during infections. Changes in the levels of reproductive hormones, such as prolactin, compromise the mammary gland’s innate immune response (IIR); however, its effect on epigenetic marks is poorly known. This work explored the epigenetic regulation induced by bovine prolactin (bPRL) on bovine mammary epithelial cells (bMECs) challenged with Staphylococcus aureus. In this work, bMECs were treated as follows: (1) control cells without any treatment, (2) bMECs treated with bPRL (5 ng/ml) at different times (12 or 24 h), (3) bMECs challenged with S. aureus for 2 h, and (4) bMECs treated with bPRL at different times (12 or 24 h), and then challenged with S. aureus 2 h. By western blot analyses of histones, we determined that the H3K9ac mark decreased (20%) in bMECs treated with bPRL (12 h) and challenged with S. aureus, while the H3K9me2 mark was increased (50%) in the same conditions. Also, this result coincided with an increase (2.3-fold) in HDAC activity analyzed using the cellular histone deacetylase fluorescent kit FLUOR DE LYS®. ChIP-qPCRs were performed to determine if the epigenetic marks detected in the histones correlate with enriched marks in the promoter regions of inflammatory genes associated with the S. aureus challenge. The H3K9ac mark was enriched in the promoter region of IL-1β, IL-10, and BNBD10 genes (1.5, 2.5, 7.5-fold, respectively) in bMECs treated with bPRL, but in bMECs challenged with S. aureus it was reduced. Besides, the H3K9me2 mark was enriched in the promoter region of IL-1β and IL-10 genes (3.5 and 2.5-fold, respectively) in bMECs challenged with S. aureus but was inhibited by bPRL. Additionally, the expression of several miRNAs was analyzed by qPCR. Let-7a-5p, miR-21a, miR-30b, miR-155, and miR-7863 miRNAs were up-regulated (2, 1.5, 10, 1.5, 3.9-fold, respectively) in bMECs challenged with S. aureus; however, bPRL induced a down-regulation in the expression of these miRNAs. In conclusion, bPRL induces epigenetic regulation on specific IIR elements, allowing S. aureus to persist and evade the host immune response.
Collapse
|
45
|
Felix L, Mishra B, Khader R, Ganesan N, Mylonakis E. In Vitro and In Vivo Bactericidal and Antibiofilm Efficacy of Alpha Mangostin Against Staphylococcus aureus Persister Cells. Front Cell Infect Microbiol 2022; 12:898794. [PMID: 35937701 PMCID: PMC9353584 DOI: 10.3389/fcimb.2022.898794] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/23/2022] [Indexed: 11/23/2022] Open
Abstract
The formation of persister cells is associated with recalcitrance and infections. In this study, we examined the antimicrobial property of alpha mangostin, a natural xanthone molecule, against methicillin-resistant Staphylococcus aureus (MRSA) persisters and biofilm. The MIC of alpha mangostin against MRSA persisters was 2 µg/ml, and activity was mediated by causing membrane permeabilization within 30 min of exposure. The membrane activity of alpha mangostin was further studied by fast-killing kinetics of MRSA persiste r cells and found that the compound exhibited 99.99% bactericidal activity within 30 min. Furthermore, alpha mangostin disrupted established MRSA biofilms and inhibited bacterial attachment as biofilm formation. Alpha mangostin down-regulated genes associated with the formation of persister cells and biofilms, such as norA, norB, dnaK, groE, and mepR, ranging from 2 to 4-folds. Alpha mangostin at 16 μg/ml was non-toxic (> 95% cell survival) to liver-derived HepG2 and lung-derived A549 cells, similarly. Still, alpha mangostin exhibited 50% cell lysis of human RBC at 16 μg/ml. Interestingly, alpha mangostin was effective in vivo at increasing the survival up to 75% (p<0.0001) of Galleria mellonella larvae infected with MRSA persister for 120 h. In conclusion, we report that alpha mangostin is active against MRSA persisters and biofilms, and these data further our understanding of the antistaphylococcal activity and toxicity of this natural compound.
Collapse
|
46
|
Hou H, Li Y, Jin Y, Chen S, Long J, Duan G, Yang H. The crafty opponent: the defense systems of Staphylococcus aureus and response measures. Folia Microbiol (Praha) 2022; 67:233-243. [PMID: 35149955 DOI: 10.1007/s12223-022-00954-9] [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: 09/22/2021] [Accepted: 01/29/2022] [Indexed: 11/29/2022]
Abstract
Staphylococcus aureus is a serious threat to public health. S. aureus infection can cause acute or long-term persistent infections that are often resistant to antibiotics and are associated with high morbidity and death. Understanding the defensive systems of S. aureus can help clinicians make the best use of antimicrobial drugs and can also help with antimicrobial stewardship. The mechanisms and clinical implications of S. aureus defense systems, as well as potential response systems, were discussed in this study. Because resistance to all currently available antibiotics is unavoidable, new medicines are always being developed. Alternative techniques, such as anti-virulence and bacteriophage therapies, are being researched and may become major tools in the fight against staphylococcal infections in the future, in addition to the development of new small compounds that affect cell viability.
Collapse
Affiliation(s)
- Hongjie Hou
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Yang Li
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Yuefei Jin
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Shuaiyin Chen
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Jinzhao Long
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Guangcai Duan
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Haiyan Yang
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China.
| |
Collapse
|
47
|
Marrazzo P, Fischer N, Nastasi C, Cricca M, Fusco D. Host–Pathogen Interactions: Organotypic Cultures to Unravel the Mysteries of the Primordial Hostility among Organisms. Pathogens 2022; 11:pathogens11030362. [PMID: 35335685 PMCID: PMC8951007 DOI: 10.3390/pathogens11030362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 12/10/2022] Open
Affiliation(s)
- Pasquale Marrazzo
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy;
- Correspondence: (P.M.); (D.F.)
| | - Natalie Fischer
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany;
| | - Claudia Nastasi
- Laboratory of Cancer Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy;
| | - Monica Cricca
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy;
| | - Daniela Fusco
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany;
- German Center for Infection Research (DZIF), Hamburg-Borstel-Lübeck-Riems, 20246 Hamburg, Germany
- Correspondence: (P.M.); (D.F.)
| |
Collapse
|
48
|
Han X, Chen Q, Zhang X, Peng J, Zhang M, Zhong Q. The elimination effects of lavender essential oil on Listeria monocytogenes biofilms developed at different temperatures and the induction of VBNC state. Lett Appl Microbiol 2022; 74:1016-1026. [PMID: 35211995 DOI: 10.1111/lam.13681] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 11/28/2022]
Abstract
Listeria monocytogenes is a typical foodborne pathogen that causes hard-to-treat bacterial infections, mainly due to its ability to form biofilm and enter into a viable but non-culturable state (VBNC). In this study, we investigated the removal effects of four antimicrobial agents on L. monocytogenes biofilms formed at 32 ℃ and 10 ℃, analyzed the resistances of the mature biofilms to antimicrobial agents, and explored the VBNC state of cells in mature biofilms induced by lavender essential oil (LEO). The results showed that the growth of L. monocytogenes was completely inhibited when 1.6% (v/v) of the LEO was added. Meanwhile, the results of the crystal violet staining and XTT reduction method indicated that different concentrations of LEO significantly reduced L. monocytogenes biofilms biomass and metabolic activities, followed by sodium hypochlorite, lactic acid, and hydrogen peroxide. Moreover, the confocal laser scanning microscopy (CLSM) images confirmed that the treated biofilms became thinner, the structure was sparse, and the appearance was blurry. More interestingly, L. monocytogenes biofilms developed at 10 ℃ were less susceptible to the sanitizers than that formed at 32 ℃. In addition, LEO presented a more significant dispersing effect on the biofilm cells, and 1/2 MIC to 4 MIC of LEO could induce fewer VBNC state cells in biofilm and plankton compared with sodium hypochlorite. This study indicated that the LEO could be considered as an ideal antibiofilm agent for controlling L. monocytogenes. But we should pay attention to the resistance of the biofilms developed at low temperatures.
Collapse
Affiliation(s)
- Xiangpeng Han
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.,Guangdong Laboratory of Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China
| | - Qingying Chen
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.,Guangdong Laboratory of Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China
| | - Xingguo Zhang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.,Guangdong Laboratory of Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China
| | - Jiayi Peng
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.,Guangdong Laboratory of Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China
| | - Mengyu Zhang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.,Guangdong Laboratory of Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China
| | - Qingping Zhong
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.,Guangdong Laboratory of Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China
| |
Collapse
|
49
|
Bär J, Boumasmoud M, Mairpady Shambat S, Vulin C, Huemer M, Schweizer TA, Gómez-Mejia A, Eberhard N, Achermann Y, Zingg PO, Mestres C, Brugger SD, Schuepbach RA, Kouyos RD, Hasse B, Zinkernagel AS. Quantification of within-patient Staphylococcus aureus phenotypic heterogeneity as a proxy for presence of persisters across clinical presentations. Clin Microbiol Infect 2022; 28:1022.e1-1022.e7. [DOI: 10.1016/j.cmi.2022.01.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/19/2022] [Accepted: 01/23/2022] [Indexed: 12/21/2022]
|
50
|
Seregina TA, Lobanov KV, Shakulov RS, Mironov AS. Enhancement of the Bactericidal Effect of Antibiotics by Inhibition of Enzymes Involved in Production of Hydrogen Sulfide in Bacteria. Mol Biol 2022; 56:638-648. [PMID: 36217334 PMCID: PMC9534473 DOI: 10.1134/s0026893322050120] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 11/23/2022]
Abstract
Counteraction of the origin and distribution of multidrug-resistant pathogens responsible for intra-hospital infections is a worldwide issue in medicine. In this brief review, we discuss the results of our recent investigations, which argue that many antibiotics, along with inactivation of their traditional biochemical targets, can induce oxidative stress (ROS production), thus resulting in increased bactericidal efficiency. As we previously showed, hydrogen sulfide, which is produced in the cells of different pathogens protects them not only against oxidative stress but also against bactericidal antibiotics. Next, we clarified the interplay of oxidative stress, cysteine metabolism, and hydrogen sulfide production. Finally, demonstrated that small molecules, which inhibit a bacterial enzyme involved in hydrogen sulfide production, potentiate bactericidal antibiotics including quinolones, beta-lactams, and aminoglycosides against bacterial pathogens in in vitro and in mouse models of infection. These inhibitors also suppress bacterial tolerance to antibiotics by disrupting the biofilm formation and substantially reducing the number of persister bacteria, which survive the antibiotic treatment. We hypothesise that agents which limit hydrogen sulfide biosynthesis are effective tools to counteract the origin and distribution of multidrug-resistant pathogens.
Collapse
Affiliation(s)
- T. A. Seregina
- Engelhardt Institute of Molecular Biology, Russian Academy of Science, 119991 Moscow, Russia
| | - K. V. Lobanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Science, 119991 Moscow, Russia
| | - R. S. Shakulov
- Engelhardt Institute of Molecular Biology, Russian Academy of Science, 119991 Moscow, Russia
| | - A. S. Mironov
- Engelhardt Institute of Molecular Biology, Russian Academy of Science, 119991 Moscow, Russia
| |
Collapse
|