1
|
Neeff M, Broderick D, Douglas RG, Biswas K. Anaerobic bacteria dominate the cholesteatoma tissue of chronic suppurative otitis media patients. Microb Pathog 2024; 196:106935. [PMID: 39270753 DOI: 10.1016/j.micpath.2024.106935] [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: 04/10/2024] [Revised: 08/19/2024] [Accepted: 09/10/2024] [Indexed: 09/15/2024]
Abstract
The aim of this study was to investigate both the microbial composition and absolute abundance of clinically relevant bacteria in tissue specimens from patients with chronic suppurative otitis media with cholesteatoma (CSOM with cholesteatoma). Mastoid mucosa and cholesteatoma tissue from eleven subjects with CSOM with cholesteatoma, and mastoid mucosa from ten controls were examined using standard hospital culture swabs, Gram staining, bacterial 16S rRNA gene sequencing, Droplet Digital PCR (ddPCR), and multiplex PCR. Positive results from culture swabs were reported in half the CSOM with cholesteatoma samples and 1 control sample. In contrast, ddPCR detected bacterial genes copies in all 11 mucosa and cholesteatoma of CSOM subjects and 3 control samples. The average bacterial gene copies in tissue samples with CSOM with cholesteaotoma (1.6 ± 0.7 log10) was significantly higher compared to healthy controls (0.3 ± 1.6). These results were corroborated with Gram-staining that identified the large presence of Gram-positive cocci cells in the cholesteatoma tissue of CSOM subjects which were not seen in the mucosa of controls. The most abundant genus detected by sequencing in the mucosa and cholesteatoma of CSOM samples was Anaerococcus (93.5 % of all reads), and genus Meiothermus (0.9 %) in the control sample. The 3 samples with the highest sequencing reads (>300) were further analysed using multiplex PCR to identify the dominant Anaerococcus species. Anaerococcus hydrogenalis was the dominant species identified in these samples. In contract, commonly named ear pathogens, genera Staphylococcus and Pseudomonas, were detected in low numbers (<0.001 % of all sequencing reads) and low prevalence (2/16 samples) in the tissue samples of this study. The results show that culture severely underestimated the bacterial diversity in CSOM samples and investigating tissue rather than standard culture swabs might be advantageous to understanding the disease process. The high abundance of bacteria and the large presence of Gram-positive cells detected in the cholesteatoma tissue of CSOM compared to mucosa of CSOM or controls could be members from the genus Anaerococcus. Anaerococcus may well be a pathogen in CSOM with cholesteatoma, but their role in this condition requires further investigation.
Collapse
Affiliation(s)
- Michel Neeff
- Department of Surgery, The University of Auckland, Auckland, 1023, New Zealand; Te Whatu Ora - Te Toka Tumai Auckland, Health New Zealand, Auckland 1142, New Zealand
| | - David Broderick
- Department of Surgery, The University of Auckland, Auckland, 1023, New Zealand
| | - Richard G Douglas
- Department of Surgery, The University of Auckland, Auckland, 1023, New Zealand; Te Whatu Ora - Te Toka Tumai Auckland, Health New Zealand, Auckland 1142, New Zealand
| | - Kristi Biswas
- Department of Surgery, The University of Auckland, Auckland, 1023, New Zealand.
| |
Collapse
|
2
|
Xie M, Lv M, Zhao Z, Li L, Jiang H, Yu Y, Zhang X, Liu P, Chen J. Plastisphere characterization in habitat of the highly endangered Shinisaurus crocodilurus: Bacterial composition, assembly, function and the comparison with surrounding environment. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 900:165807. [PMID: 37506917 DOI: 10.1016/j.scitotenv.2023.165807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/15/2023] [Accepted: 07/24/2023] [Indexed: 07/30/2023]
Abstract
Plastisphere is a new niche for microorganisms that complicate the ecological effects of plastics, and may profoundly influence biodiversity and habitat conservation. The DaGuishan National Nature Reserve, one of the largest habitats of the highly endangered crocodile lizard (Shinisaurus crocodilurus), is experiencing plastic pollution without sufficient attention. Here, plastisphere collected from captive tanks of crocodile lizards in this nature reserve was characterized for the first time. Three types of plastic (PE-PP, PE1, and PE2) together with the surrounding water and soil samples, were collected, and 16S rRNA sequencing technology was used to characterize the bacterial composition. The results demonstrated that plastisphere was driven by stochastic process and had a distinct bacterial community with higher diversity than that in surrounding water (p < 0.05). Bacteria related to nitrogen and carbon cycles (Pseudomonas psychrotolerans, Methylobacterium-Methylorubrum) were more abundant in plastisphere than in water or soil (p < 0.05). More importantly, plastics recruited pathogens and those bacteria function in antibiotic resistant genes (ARGs) coding. Bacteria related to polymer degradation also proliferated in plastisphere, especially Bacillus subtilis with a fold change of 42.01. The PE2 plastisphere, which had the lowest diversity and was dominated by Methylobacterium-Methylorubrum differed from PE 1 and PE-PP plastispheres totally. Plastics' morphology and aquatic nutrient levels contributed to the heterogeneity of different plastispheres. Overall, this study demonstrated that plastispheres diversify in composition and function, affecting ecosystem services directly or indirectly. Pathogens and bacteria related to ARGs expression enriched in the plastisphere should not be ignored because they may threaten the health of crocodile lizards by increasing the risk of infection. Plastic pollution control should be included in conservation efforts for crocodile lizards. This study provides new insights into the potential impacts of plastisphere, which is important for ecological risk assessments of plastic pollution in the habitats of endangered species.
Collapse
Affiliation(s)
- Mujiao Xie
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China
| | - Mei Lv
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China
| | - Zhiwen Zhao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China
| | - Linmiao Li
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China
| | - Haiying Jiang
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China
| | - Yepin Yu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China
| | - Xiujuan Zhang
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China
| | - Ping Liu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China
| | - Jinping Chen
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China.
| |
Collapse
|
3
|
Lewis ED, Ortega EF, Dao MC, Barger K, Mason JB, Leong JM, Osburne MS, Magoun L, Nepveux V FJ, Chishti AH, Schwake C, Quynh A, Gilhooly CH, Petty G, Guo W, Matuszek G, Pereira D, Reddy M, Wang J, Wu D, Meydani SN, Combs GF. Safe and effective delivery of supplemental iron to healthy adults: a two-phase, randomized, double-blind trial - the safe iron study. Front Nutr 2023; 10:1230061. [PMID: 37899826 PMCID: PMC10603204 DOI: 10.3389/fnut.2023.1230061] [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: 05/27/2023] [Accepted: 08/28/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction The safety of novel forms of iron in healthy, iron-replete adults as might occur if used in population-based iron supplementation programs was examined. We tested the hypotheses that supplementation with nanoparticulate iron hydroxide adipate tartrate (IHAT), an iron-enriched Aspergillus oryzae product (ASP), or ferrous sulphate heptahydrate (FS) are safe as indicated by erythrocyte susceptibility to malarial infection, bacterial proliferation, and gut inflammation. Responses to FS administered daily or weekly, and with or without other micronutrients were compared. Methods Two phases of randomized, double-blinded trials were conducted in Boston, MA. Phase I randomized 160 volunteers to six treatments: placebo, IHAT, ASP, FS, and FS plus a micronutrient powder (MNP) administrated daily at 60 mg Fe/day; and FS administered as a single weekly dose of 420 mg Fe. Phase II randomized 86 volunteers to IHAT, ASP, or FS administered at 120 mg Fe/day. Completing these phases were 151 and 77 participants, respectively. The study was powered to detect effects on primary endpoints: susceptibility of participant erythrocytes to infection by Plasmodium falciparum, the proliferation potential of selected pathogenic bacteria in sera, and markers of gut inflammation. Secondary endpoints for which the study was not powered included indicators of iron status and gastrointestinal symptoms. Results Supplementation with any form of iron did not affect any primary endpoint. In Phase I, the frequency of gastrointestinal symptoms associated with FS was unaffected by dosing with MNP or weekly administration; but participants taking IHAT more frequently reported abdominal pain (27%, p < 0.008) and nausea (4%, p = 0.009) than those taking FS, while those taking ASP more frequently reported nausea (8%, p = 0.009). Surprisingly, only 9% of participants taking IHAT at 120 mg Fe/day (Phase II) reported abdominal pain and no other group reported that symptom. Discussion With respect to the primary endpoints, few differences were found when comparing these forms of iron, indicating that 28 days of 60 or 120 mg/day of IHAT, ASP, or FS may be safe for healthy, iron-replete adults. With respect to other endpoints, subjects receiving IHAT more frequently reported abdominal pain and nausea, suggesting the need for further study. Clinical Trial Registration ClinicalTrials.gov, NCT03212677; registered: 11 July 2017.
Collapse
Affiliation(s)
- Erin D. Lewis
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Edwin F. Ortega
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Maria Carlota Dao
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Kathryn Barger
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Joel B. Mason
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - John M. Leong
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA, United States
| | - Marcia S. Osburne
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA, United States
| | - Loranne Magoun
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA, United States
| | - Felix J. Nepveux V
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA, United States
| | - Athar H. Chishti
- Department of Developmental, Molecular and Chemical Biology, Tufts University, Boston, MA, United States
| | - Christopher Schwake
- Department of Developmental, Molecular and Chemical Biology, Tufts University, Boston, MA, United States
| | - Anh Quynh
- Department of Developmental, Molecular and Chemical Biology, Tufts University, Boston, MA, United States
| | - Cheryl H. Gilhooly
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Gayle Petty
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Weimin Guo
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Gregory Matuszek
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Dora Pereira
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Manju Reddy
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States
| | - Jifan Wang
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Dayong Wu
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Simin N. Meydani
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Gerald F. Combs
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| |
Collapse
|
4
|
Leite DPDSBM, Barbosa IC, da Silva RA, Fernandes PR, Abad ACA, da Silva JG, Mota RA, Porto TS. Occurrence of antimicrobial-resistant Staphylococcus aureus in a Brazilian veterinary hospital environment. Braz J Microbiol 2023; 54:2393-2401. [PMID: 37407882 PMCID: PMC10485224 DOI: 10.1007/s42770-023-01035-w] [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: 09/14/2022] [Accepted: 06/07/2023] [Indexed: 07/07/2023] Open
Abstract
Antimicrobial resistance is a threat to public health. The emergence of antibiotic-resistant Staphylococcus aureus represents a priority for the implementation of preventive measures. The objective was to isolate S. aureus in humans, animals, and animal health care environment, and to characterize the genotypic and phenotypic profile of antimicrobial resistance in these isolates. We isolated S. aureus from staff, animals, and environment of a veterinary hospital, and identified their antimicrobial resistance profiles. Samples were collected from 20 humans, 13 animals, 14 surfaces, 8 mobile phones, and 7 veterinarians' stethoscopes by using sterile swabs. S. aureus was isolated by culturing on mannitol salt agar and preliminary identification was done by Gram staining and catalase test. Subsequently, a polymerase chain reaction was performed for species confirmation and investigating their antimicrobial-resistant genotypic profiles. Phenotypic profiles of resistant isolates were determined using the disk-diffusion technique. Ten S. aureus isolates were recovered from 5/20 humans (25%), it was also recovered from 2/13 animals (15.38%), including 1 dog and 1 cat, and from 1/14 of surfaces (7.14%). The oxacillin-susceptible mecA-positive Staphylococcus aureus phenotype was identified in a feline. Most of the isolates carried at least two resistance genes of different antimicrobial classes, with 90% (9/10) presenting the gene blaZ, with 10% (1/10) presenting the gene mecA, 20% (2/10) presenting tet38, 10% (1/10) presenting tetM, 90% (9/10) presenting norA, 50% (5/10) presenting norC, 10% (1/10) presenting ermA, and 60% (6/10) presenting ermB. In antibiograms, resistance to penicillin was identified in all the isolates, resistance to erythromycin was identified in 80% (8/10), and all the isolate's resistance to erythromycin presented erythromycin-induced resistance to clindamycin. Antimicrobial resistance in the veterinary hospital requires attention due to the risk of interspecies transmission, gene transfer between bacteria that colonize companion animals and humans and, can make antimicrobial therapy difficult.
Collapse
Affiliation(s)
- Denny Parente de Sá Barreto Maia Leite
- Department of Veterinary Medicine, Federal Rural University of Pernambuco (UFRPE), Recife, Pernambuco, Brazil
- Department of Animal Morphology and Physiology, Federal Rural University of Pernambuco (UFRPE), Recife, Pernambuco, Brazil
| | - Iago Carvalho Barbosa
- Department of Veterinary Medicine, Federal Rural University of Pernambuco (UFRPE), Recife, Pernambuco, Brazil
- Department of Animal Morphology and Physiology, Federal Rural University of Pernambuco (UFRPE), Recife, Pernambuco, Brazil
| | - Renato Amorim da Silva
- Department of Veterinary Medicine, Federal Rural University of Pernambuco (UFRPE), Recife, Pernambuco, Brazil
- Department of Animal Morphology and Physiology, Federal Rural University of Pernambuco (UFRPE), Recife, Pernambuco, Brazil
| | - Pollyanne Raysa Fernandes
- Department of Veterinary Medicine, Federal Rural University of Pernambuco (UFRPE), Recife, Pernambuco, Brazil
- Department of Animal Morphology and Physiology, Federal Rural University of Pernambuco (UFRPE), Recife, Pernambuco, Brazil
| | - Atzel Candido Acosta Abad
- Department of Veterinary Medicine, Federal Rural University of Pernambuco (UFRPE), Recife, Pernambuco, Brazil
- Department of Animal Morphology and Physiology, Federal Rural University of Pernambuco (UFRPE), Recife, Pernambuco, Brazil
| | - José Givanildo da Silva
- Department of Preventive Veterinary Medicine and Animal Production, School of Veterinary Medicine and Zootechnics, Federal University of Bahia (UFBA), Salvador, Bahia, Brazil
| | - Rinaldo Aparecido Mota
- Department of Veterinary Medicine, Federal Rural University of Pernambuco (UFRPE), Recife, Pernambuco, Brazil.
| | - Tatiana Souza Porto
- Department of Animal Morphology and Physiology, Federal Rural University of Pernambuco (UFRPE), Recife, Pernambuco, Brazil.
| |
Collapse
|
5
|
Heinzinger LR, Pugh AR, Wagner JA, Otto M. Evaluating the Translational Potential of Bacteriocins as an Alternative Treatment for Staphylococcus aureus Infections in Animals and Humans. Antibiotics (Basel) 2023; 12:1256. [PMID: 37627676 PMCID: PMC10451987 DOI: 10.3390/antibiotics12081256] [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: 06/27/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Antibiotic resistance remains a global threat to human and animal health. Staphylococcus aureus is an opportunistic pathogen that causes minor to life-threatening infections. The widespread use of antibiotics in the clinical, veterinary, and agricultural setting combined with the increasing prevalence of antibiotic-resistant S. aureus strains makes it abundantly clear that alternatives to antibiotics are urgently needed. Bacteriocins represent one potential alternative therapeutic. They are antimicrobial peptides that are produced by bacteria that are generally nontoxic and have a relatively narrow target spectrum, and they leave many commensals and most mammalian cells unperturbed. Multiple studies involving bacteriocins (e.g., nisin, epidermicin, mersacidin, and lysostaphin) have demonstrated their efficacy at eliminating or treating a wide variety of S. aureus infections in animal models. This review provides a comprehensive and updated evaluation of animal studies involving bacteriocins and highlights their translational potential. The strengths and limitations associated with bacteriocin treatments compared with traditional antibiotic therapies are evaluated, and the challenges that are involved with implementing novel therapeutics are discussed.
Collapse
Affiliation(s)
| | | | | | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814, USA; (L.R.H.); (A.R.P.); (J.A.W.)
| |
Collapse
|
6
|
Huynh TQ, Tran VN, Thai VC, Nguyen HA, Nguyen NTG, Tran MK, Nguyen TPT, Le CA, Ho LTN, Surian NU, Chen S, Nguyen TTH. Genomic alterations involved in fluoroquinolone resistance development in Staphylococcus aureus. PLoS One 2023; 18:e0287973. [PMID: 37494330 PMCID: PMC10370734 DOI: 10.1371/journal.pone.0287973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 06/19/2023] [Indexed: 07/28/2023] Open
Abstract
AIM Fluoroquinolone (FQ) is a potent antibiotic class. However, resistance to this class emerges quickly which hinders its application. In this study, mechanisms leading to the emergence of multidrug-resistant (MDR) Staphylococcus aureus (S. aureus) strains under FQ exposure were investigated. METHODOLOGY S. aureus ATCC 29213 was serially exposed to ciprofloxacin (CIP), ofloxacin (OFL), or levofloxacin (LEV) at sub-minimum inhibitory concentrations (sub-MICs) for 12 days to obtain S. aureus -1 strains and antibiotic-free cultured for another 10 days to obtain S. aureus-2 strains. The whole genome (WGS) and target sequencing were applied to analyze genomic alterations; and RT-qPCR was used to access the expressions of efflux-related genes, alternative sigma factors, and genes involved in FQ resistance. RESULTS A strong and irreversible increase of MICs was observed in all applied FQs (32 to 128 times) in all S. aureus-1 and remained 16 to 32 times in all S. aureus-2. WGS indicated 10 noticeable mutations occurring in all FQ-exposed S. aureus including 2 insdel mutations in SACOL0573 and rimI; a synonymous mutation in hslO; and 7 missense mutations located in an untranslated region. GrlA, was found mutated (R570H) in all S. aureus-1 and -2. Genes encoding for efflux pumps and their regulator (norA, norB, norC, and mgrA); alternative sigma factors (sigB and sigS); acetyltransferase (rimI); methicillin resistance (fmtB); and hypothetical protein BJI72_0645 were overexpressed in FQ-exposed strains. CONCLUSION The emergence of MDR S. aureus was associated with the mutations in the FQ-target sequences and the overexpression of efflux pump systems and their regulators.
Collapse
Affiliation(s)
- Thuc Quyen Huynh
- School of Biotechnology, International University, Ho Chi Minh City, Vietnam
- Research Center for Infectious Diseases, International University, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Van Nhi Tran
- School of Biotechnology, International University, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Van Chi Thai
- School of Biotechnology, International University, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Hoang An Nguyen
- School of Biotechnology, International University, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Ngoc Thuy Giang Nguyen
- School of Biotechnology, International University, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Minh Khang Tran
- School of Biotechnology, International University, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Thi Phuong Truc Nguyen
- School of Biotechnology, International University, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Cat Anh Le
- School of Biotechnology, International University, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Le Thanh Ngan Ho
- School of Biotechnology, International University, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | | | - Swaine Chen
- Genome Institute of Singapore, Singapore, Singapore
| | - Thi Thu Hoai Nguyen
- School of Biotechnology, International University, Ho Chi Minh City, Vietnam
- Research Center for Infectious Diseases, International University, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| |
Collapse
|
7
|
Conte AL, Brunetti F, Marazzato M, Longhi C, Maurizi L, Raponi G, Palamara AT, Grassi S, Conte MP. Atopic dermatitis-derived Staphylococcus aureus strains: what makes them special in the interplay with the host. Front Cell Infect Microbiol 2023; 13:1194254. [PMID: 37389215 PMCID: PMC10303148 DOI: 10.3389/fcimb.2023.1194254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 05/22/2023] [Indexed: 07/01/2023] Open
Abstract
Background Atopic dermatitis (AD) is a chronic inflammatory skin condition whose pathogenesis involves genetic predisposition, epidermal barrier dysfunction, alterations in the immune responses and microbial dysbiosis. Clinical studies have shown a link between Staphylococcus aureus and the pathogenesis of AD, although the origins and genetic diversity of S. aureus colonizing patients with AD is poorly understood. The aim of the study was to investigate if specific clones might be associated with the disease. Methods WGS analyses were performed on 38 S. aureus strains, deriving from AD patients and healthy carriers. Genotypes (i.e. MLST, spa-, agr- and SCCmec-typing), genomic content (e.g. virulome and resistome), and the pan-genome structure of strains have been investigated. Phenotypic analyses were performed to determine the antibiotic susceptibility, the biofilm production and the invasiveness within the investigated S. aureus population. Results Strains isolated from AD patients revealed a high degree of genetic heterogeneity and a shared set of virulence factors and antimicrobial resistance genes, suggesting that no genotype and genomic content are uniquely associated with AD. The same strains were characterized by a lower variability in terms of gene content, indicating that the inflammatory conditions could exert a selective pressure leading to the optimization of the gene repertoire. Furthermore, genes related to specific mechanisms, like post-translational modification, protein turnover and chaperones as well as intracellular trafficking, secretion and vesicular transport, were significantly more enriched in AD strains. Phenotypic analysis revealed that all of our AD strains were strong or moderate biofilm producers, while less than half showed invasive capabilities. Conclusions We conclude that in AD skin, the functional role played by S. aureus may depend on differential gene expression patterns and/or on post-translational modification mechanisms rather than being associated with peculiar genetic features.
Collapse
Affiliation(s)
- Antonietta Lucia Conte
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Francesca Brunetti
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Massimiliano Marazzato
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Catia Longhi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Linda Maurizi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Giammarco Raponi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Anna Teresa Palamara
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory Affiliated to Institute Pasteur Italia- Cenci Bolognetti Foundation, Rome, Italy
| | - Sara Grassi
- Dermatology Clinic, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Maria Pia Conte
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
8
|
Cheruvanachari P, Pattnaik S, Mishra M, Pragyandipta P, Pattnaik A, Naik PK. Deciphering the antibiofilm potential of 2-Phenylethyl methyl ether (PEME), a bioactive compound of Kewda essential oil against Staphylococcus aureus. Microb Pathog 2023; 179:106093. [PMID: 37004966 DOI: 10.1016/j.micpath.2023.106093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023]
Abstract
Opportunistic pathogenic bacteria and their pathogenicity linked with biofilm infections become a severe issue as they resist the actions of multiple antimicrobial drugs. Naturally derived drugs having antibiofilm properties are more effective than chemically synthesized drugs. The plant derived essential oils are a rich source of phytoconstituents with widespread pharmacological values. In the present investigation, a major phytoconstituent, 2-Phenyl Ethyl Methyl Ether (PEME) of Kewda essential oil extracted from the flowers of Pandanus odorifer was explored for its prospective antimicrobial and anti-biofilm properties against ESKAPE pathogenic bacterial strain, Staphylococcus aureus and MTCC 740. The minimum inhibitory concentration (MIC) of PEME was found to be 50 mM against the tested bacterial strains. A gradual decrease in biofilm production was observed when PEME was treated with the sub-MIC concentration. The reduction in biofilm formation was noticeable from qualitative assay i.e., Congo Red Agar Assay (CRA) and further quantified by crystal violet staining assay. The decline in exopolysaccharides production was quantified, with the highest inhibition against MTCC 740 with a decrease of 71.76 ± 4.56% compared to untreated control. From the microscopic analysis (light and microscopic fluorescence method), PEME exhibited inhibitory effect on biofilm formation on the polystyrene surface. The In silico studies stated that PEME could invariably bind to biofilm associated target proteins. Further, transcriptomic data analysis suggested the role of PEME in the down-regulation of specific genes, agrA, sarA, norA and mepR, which are critically associated with bacterial virulence, biofilm dynamics and drug resistance patterns in S. aureus. Further, qRT-PCR analysis validated the role of PEME on biofilm inhibition by relative downregulation of agrA, sarA, norA and mepR genes. Further, advanced in silico methodologies could be employed in future investigations to validate its candidature as promising anti-biofilm agent.
Collapse
|
9
|
Majumder S, Sackey T, Viau C, Park S, Xia J, Ronholm J, George S. Genomic and phenotypic profiling of Staphylococcus aureus isolates from bovine mastitis for antibiotic resistance and intestinal infectivity. BMC Microbiol 2023; 23:43. [PMID: 36803552 PMCID: PMC9940407 DOI: 10.1186/s12866-023-02785-1] [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: 08/18/2022] [Accepted: 01/30/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Staphylococcus aureus is one of the prevalent etiological agents of contagious bovine mastitis, causing a significant economic burden on the global dairy industry. Given the emergence of antibiotic resistance (ABR) and possible zoonotic spillovers, S aureus from mastitic cattle pose threat to both veterinary and public health. Therefore, assessment of their ABR status and pathogenic translation in human infection models is crucial. RESULTS In this study, 43 S. aureus isolates associated with bovine mastitis obtained from four different Canadian provinces (Alberta, Ontario, Quebec, and Atlantic provinces) were tested for ABR and virulence through phenotypic and genotypic profiling. All 43 isolates exhibited crucial virulence characteristics such as hemolysis, and biofilm formation, and six isolates from ST151, ST352, and ST8 categories showed ABR. Genes associated with ABR (tetK, tetM, aac6', norA, norB, lmrS, blaR, blaZ, etc.), toxin production (hla, hlab, lukD, etc.), adherence (fmbA, fnbB, clfA, clfB, icaABCD, etc.), and host immune invasion (spa, sbi, cap, adsA, etc.) were identified by analyzing whole-genome sequences. Although none of the isolates possessed human adaptation genes, both groups of ABR and antibiotic-susceptible isolates demonstrated intracellular invasion, colonization, infection, and death of human intestinal epithelial cells (Caco-2), and Caenorhabditis elegans. Notably, the susceptibilities of S. aureus towards antibiotics such as streptomycin, kanamycin, and ampicillin were altered when the bacteria were internalized in Caco-2 cells and C. elegans. Meanwhile, tetracycline, chloramphenicol, and ceftiofur were comparatively more effective with ≤ 2.5 log10 reductions of intracellular S. aureus. CONCLUSIONS This study demonstrated the potential of S. aureus isolated from mastitis cows to possess virulence characteristics enabling invasion of intestinal cells thus calling for developing therapeutics capable of targeting drug-resistant intracellular pathogens for effective disease management.
Collapse
Affiliation(s)
- Satwik Majumder
- grid.14709.3b0000 0004 1936 8649Department of Food Science and Agricultural Chemistry, Macdonald Campus, McGill University, Macdonald-Stewart Building, Room-1039, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada
| | - Trisha Sackey
- grid.14709.3b0000 0004 1936 8649Department of Food Science and Agricultural Chemistry, Macdonald Campus, McGill University, Macdonald-Stewart Building, Room-1039, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada
| | - Charles Viau
- grid.14709.3b0000 0004 1936 8649Institute of Parasitology, Macdonald Campus, McGill University, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada
| | - Soyoun Park
- grid.14709.3b0000 0004 1936 8649Department of Food Science and Agricultural Chemistry, Macdonald Campus, McGill University, Macdonald-Stewart Building, Room-1039, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada
| | - Jianguo Xia
- grid.14709.3b0000 0004 1936 8649Institute of Parasitology, Macdonald Campus, McGill University, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada ,grid.14709.3b0000 0004 1936 8649Department of Animal Science, Macdonald Campus, McGill University, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada
| | - Jennifer Ronholm
- grid.14709.3b0000 0004 1936 8649Department of Food Science and Agricultural Chemistry, Macdonald Campus, McGill University, Macdonald-Stewart Building, Room-1039, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada ,grid.14709.3b0000 0004 1936 8649Department of Animal Science, Macdonald Campus, McGill University, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada
| | - Saji George
- Department of Food Science and Agricultural Chemistry, Macdonald Campus, McGill University, Macdonald-Stewart Building, Room-1039, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9, Canada.
| |
Collapse
|
10
|
Stephen J, Salam F, Lekshmi M, Kumar SH, Varela MF. The Major Facilitator Superfamily and Antimicrobial Resistance Efflux Pumps of the ESKAPEE Pathogen Staphylococcus aureus. Antibiotics (Basel) 2023; 12:antibiotics12020343. [PMID: 36830254 PMCID: PMC9952236 DOI: 10.3390/antibiotics12020343] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
The ESKAPEE bacterial pathogen Staphylococcus aureus has posed a serious public health concern for centuries. Throughout its evolutionary course, S. aureus has developed strains with resistance to antimicrobial agents. The bacterial pathogen has acquired multidrug resistance, causing, in many cases, untreatable infectious diseases and raising serious public safety and healthcare concerns. Amongst the various mechanisms for antimicrobial resistance, integral membrane proteins that serve as secondary active transporters from the major facilitator superfamily constitute a chief system of multidrug resistance. These MFS transporters actively export structurally different antimicrobial agents from the cells of S. aureus. This review article discusses the S. aureus-specific MFS multidrug efflux pump systems from a molecular mechanistic perspective, paying particular attention to structure-function relationships, modulation of antimicrobial resistance mediated by MFS drug efflux pumps, and direction for future investigation.
Collapse
Affiliation(s)
- Jerusha Stephen
- ICAR-Central Institute of Fisheries Education (CIFE), Mumbai 400061, India
| | - Fathima Salam
- ICAR-Central Institute of Fisheries Education (CIFE), Mumbai 400061, India
| | - Manjusha Lekshmi
- ICAR-Central Institute of Fisheries Education (CIFE), Mumbai 400061, India
| | - Sanath H. Kumar
- ICAR-Central Institute of Fisheries Education (CIFE), Mumbai 400061, India
| | - Manuel F. Varela
- Department of Biology, Eastern New Mexico University, Portales, NM 88130, USA
- Correspondence: ; Tel.: +1-575-562-2464
| |
Collapse
|
11
|
Fu T, Fan Z, Li Y, Li Z, Du B, Liu S, Cui X, Zhang R, Zhao H, Feng Y, Xue G, Cui J, Yan C, Gan L, Feng J, Xu Z, Yu Z, Tian Z, Ding Z, Chen J, Chen Y, Yuan J. ArcR contributes to tolerance to fluoroquinolone antibiotics by regulating katA in Staphylococcus aureus. Front Microbiol 2023; 14:1106340. [PMID: 36910210 PMCID: PMC9998937 DOI: 10.3389/fmicb.2023.1106340] [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: 11/23/2022] [Accepted: 01/04/2023] [Indexed: 02/26/2023] Open
Abstract
Staphylococcus aureus is an opportunistic pathogen that shows a unique ability to quickly respond to a variety of antibiotics. The Crp/Fnr family transcriptional regulator ArcR controls expression of arginine deiminase pathway genes arcABDC, which enable the utilization of arginine as an energy source for cell growth under anaerobic conditions. However, ArcR shares low overall similarity with other Crp/Fnr family proteins, suggesting that they differ in the response to environmental stress. In this study, MIC and survival assays were performed to determine the role of ArcR in antibiotic resistance and tolerance. The results showed that deletion of arcR reduced tolerance of S.aureus to fluoroquinolone antibiotics, mainly through a defect in the response to oxidative stress. In ΔarcR mutant, the expression of the major catalase gene katA was downregulated, and katA overexpression restored bacterial resistance to oxidative stress and antibiotics. We showed that ArcR directly regulated katA transcription by binding to the promoter region of katA. Therefore, our results revealed the contribution of ArcR in bacterial tolerance to oxidative stress and subsequently to fluoroquinolones antibiotics. This study added our understanding on the role of Crp/Fnr family in bacterial susceptibility to antibiotics.
Collapse
Affiliation(s)
- Tongtong Fu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Zheng Fan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Yujie Li
- Department of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhoufei Li
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Bing Du
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Shiyu Liu
- Military Supplies and Energy Quality Supervision Station of NV, PLA, Beijing, China
| | - Xiaohu Cui
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Rui Zhang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Hanqing Zhao
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Yanling Feng
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Guanhua Xue
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Jinghua Cui
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Chao Yan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Lin Gan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Junxia Feng
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Ziying Xu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Zihui Yu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Ziyan Tian
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Zanbo Ding
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Jinfeng Chen
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Yujie Chen
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Jing Yuan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| |
Collapse
|
12
|
Afzal M, Vijay AK, Stapleton F, Willcox M. The Relationship between Ciprofloxacin Resistance and Genotypic Changes in S. aureus Ocular Isolates. Pathogens 2022; 11:1354. [PMID: 36422605 PMCID: PMC9695201 DOI: 10.3390/pathogens11111354] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/26/2022] [Accepted: 11/10/2022] [Indexed: 07/28/2023] Open
Abstract
Staphylococcus aureus (S. aureus) is a frequent cause of eye infections with some isolates exhibiting increased antimicrobial resistance to commonly prescribed antibiotics. The increasing resistance of ocular S. aureus to ciprofloxacin is a serious concern as it is a commonly used as a first line antibiotic to treat S. aureus keratitis. This study aimed to analyse genetic mutations in the genomes of 25 S. aureus isolates from infections or non-infectious ocular conditions from the USA and Australia and their relationship to ciprofloxacin resistance. Overall, 14/25 isolates were phenotypically resistant to ciprofloxacin. All isolates were analyzed for mutations in their quinolone resistance-determining regions (QRDRs) and efflux pump genes. Of the fourteen resistant isolates, 9/14 had ciprofloxacin resistance mutations within their QRDRs, at codons 80 or 84 within the parC subunit and codon 84 within the gyrA subunit of DNA gyrase. The highest resistance (MIC = 2560 μg/mL) was associated with two SNPs in both gyrA and parC. Other resistant isolates (3/14) had mutations within norB. Mutations in genes of other efflux pumps and their regulator (norA, norC, mepA, mdeA, sepA, sdrM, mepR, arlR, and arlS) or the DNA mismatch repair (MMR) system (mutL and mutS) were not associated with increased resistance to ciprofloxacin. The functional mutations associated with ciprofloxacin resistance in QRDRs (gyrA and parC) and norB suggests that these are the most common reasons for ciprofloxacin resistance in ocular isolates. Novel SNPs of gyrA Glu-88-Leu, Asn-860-Thr and Thr-845-Ala and IIe-855-Met, identified in this study, need further gene knock out/in studies to better understand their effect on ciprofloxacin resistance.
Collapse
|
13
|
Molecular Mechanisms of Drug Resistance in Staphylococcus aureus. Int J Mol Sci 2022; 23:ijms23158088. [PMID: 35897667 PMCID: PMC9332259 DOI: 10.3390/ijms23158088] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 12/03/2022] Open
Abstract
This paper discusses the mechanisms of S. aureus drug resistance including: (1) introduction. (2) resistance to beta-lactam antibiotics, with particular emphasis on the mec genes found in the Staphylococcaceae family, the structure and occurrence of SCCmec cassettes, as well as differences in the presence of some virulence genes and its expression in major epidemiological types and clones of HA-MRSA, CA-MRSA, and LA-MRSA strains. Other mechanisms of resistance to beta-lactam antibiotics will also be discussed, such as mutations in the gdpP gene, BORSA or MODSA phenotypes, as well as resistance to ceftobiprole and ceftaroline. (3) Resistance to glycopeptides (VRSA, VISA, hVISA strains, vancomycin tolerance). (4) Resistance to oxazolidinones (mutational and enzymatic resistance to linezolid). (5) Resistance to MLS-B (macrolides, lincosamides, ketolides, and streptogramin B). (6) Aminoglycosides and spectinomicin, including resistance genes, their regulation and localization (plasmids, transposons, class I integrons, SCCmec), and types and spectrum of enzymes that inactivate aminoglycosides. (7). Fluoroquinolones (8) Tetracyclines, including the mechanisms of active protection of the drug target site and active efflux of the drug from the bacterial cell. (9) Mupirocin. (10) Fusidic acid. (11) Daptomycin. (12) Resistance to other antibiotics and chemioterapeutics (e.g., streptogramins A, quinupristin/dalfopristin, chloramphenicol, rifampicin, fosfomycin, trimethoprim) (13) Molecular epidemiology of MRSA.
Collapse
|
14
|
Forecasting Staphylococcus aureus Infections Using Genome-Wide Association Studies, Machine Learning, and Transcriptomic Approaches. mSystems 2022; 7:e0037822. [PMID: 35862809 PMCID: PMC9426533 DOI: 10.1128/msystems.00378-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus is a major human and animal pathogen, colonizing diverse ecological niches within its hosts. Predicting whether an isolate will infect a specific host and its subsequent clinical fate remains unknown. In this study, we investigated the S. aureus pangenome using a curated set of 356 strains, spanning a wide range of hosts, origins, and clinical display and antibiotic resistance profiles. We used genome-wide association study (GWAS) and random forest (RF) algorithms to discriminate strains based on their origins and clinical sources. Here, we show that the presence of sak and scn can discriminate strains based on their host specificity, while other genes such as mecA are often associated with virulent outcomes. Both GWAS and RF indicated the importance of intergenic regions (IGRs) and coding DNA sequence (CDS) but not sRNAs in forecasting an outcome. Additional transcriptomic analyses performed on the most prevalent clonal complex 8 (CC8) clonal types, in media mimicking nasal colonization or bacteremia, indicated three RNAs as potential RNA markers to forecast infection, followed by 30 others that could serve as infection severity predictors. Our report shows that genetic association and transcriptomics are complementary approaches that will be combined in a single analytical framework to improve our understanding of bacterial pathogenesis and ultimately identify potential predictive molecular markers. IMPORTANCE Predicting the outcome of bacterial colonization and infections, based on extensive genomic and transcriptomic data from a given pathogen, would be of substantial help for clinicians in treating and curing patients. In this report, genome-wide association studies and random forest algorithms have defined gene combinations that differentiate human from animal strains, colonization from diseases, and nonsevere from severe diseases, while it revealed the importance of IGRs and CDS, but not small RNAs (sRNAs), in anticipating an outcome. In addition, transcriptomic analyses performed on the most prevalent clonal types, in media mimicking either nasal colonization or bacteremia, revealed significant differences and therefore potent RNA markers. Overall, the use of both genomic and transcriptomic data in a single analytical framework can enhance our understanding of bacterial pathogenesis.
Collapse
|
15
|
Holasová K, Křížkovská B, Hoang L, Dobiasová S, Lipov J, Macek T, Křen V, Valentová K, Ruml T, Viktorová J. Flavonolignans from silymarin modulate antibiotic resistance and virulence in Staphylococcus aureus. Biomed Pharmacother 2022; 149:112806. [PMID: 35303568 DOI: 10.1016/j.biopha.2022.112806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/25/2022] [Accepted: 03/07/2022] [Indexed: 11/25/2022] Open
Abstract
Antibiotic resistance is currently a serious health problem. Since the discovery of new antibiotics no longer seems to be a sufficient tool in the fight against multidrug-resistant infections, adjuvant (combination) therapy is gaining in importance as well as reducing bacterial virulence. Silymarin is a complex of flavonoids and flavonolignans known for its broad spectrum of biological activities, including its ability to modulate drug resistance in cancer. This work aimed to test eleven, optically pure silymarin flavonolignans for their ability to reverse the multidrug resistance phenotype of Staphylococcus aureus and reduce its virulence. Silybin A, 2,3-dehydrosilybin B, and 2,3-dehydrosilybin AB completely reversed antibiotic resistance at concentrations of 20 µM or less. Both 2,3-dehydrosilybin B and AB decreased the antibiotic-induced gene expression of representative efflux pumps belonging to the major facilitator (MFS), multidrug and toxic compound extrusion (MATE), and ATP-binding cassette (ABC) families. 2,3-Dehydrosilybin B also inhibited ethidium bromide accumulation and efflux in a clinical isolate whose NorA and MdeA overproduction was induced by antibiotics. Most of the tested flavonolignans reduced cell-to-cell communication on a tetrahydrofuran-borate (autoinducer-2) basis, with isosilychristin leading the way followed by 2,3-dehydrosilybin A and AB, which halved communication at 10 µM. Anhydrosilychristin was the only compound that reduced communication based on acyl-homoserine lactone (autoinducer 1), with an IC50 of 4.8 µM. Except for isosilychristin and anhydrosilychristin, all of the flavonolignans inhibited S. aureus surface colonization, with 2,3-dehydrosilybin A being the most active (IC50 10.6 µM). In conclusion, the selected flavonolignans, particularly derivatives of 2,3-dehydrosilybin B, 2,3-dehydrosilybin AB, and silybin A are non-toxic modulators of S. aureus multidrug resistance and can decrease the virulence of the bacterium, which deserves further detailed research.
Collapse
Affiliation(s)
- Kateřina Holasová
- Department of Biochemistry and Microbiology, Faculty of Food and Biochemical Technology, University of Chemistry and Technology, Prague, Technická 3, Prague 166 28, Czech Republic.
| | - Bára Křížkovská
- Department of Biochemistry and Microbiology, Faculty of Food and Biochemical Technology, University of Chemistry and Technology, Prague, Technická 3, Prague 166 28, Czech Republic.
| | - Lan Hoang
- Department of Biochemistry and Microbiology, Faculty of Food and Biochemical Technology, University of Chemistry and Technology, Prague, Technická 3, Prague 166 28, Czech Republic.
| | - Simona Dobiasová
- Department of Biochemistry and Microbiology, Faculty of Food and Biochemical Technology, University of Chemistry and Technology, Prague, Technická 3, Prague 166 28, Czech Republic.
| | - Jan Lipov
- Department of Biochemistry and Microbiology, Faculty of Food and Biochemical Technology, University of Chemistry and Technology, Prague, Technická 3, Prague 166 28, Czech Republic.
| | - Tomáš Macek
- Department of Biochemistry and Microbiology, Faculty of Food and Biochemical Technology, University of Chemistry and Technology, Prague, Technická 3, Prague 166 28, Czech Republic.
| | - Vladimír Křen
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ 142 20 Prague, Czech Republic.
| | - Kateřina Valentová
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ 142 20 Prague, Czech Republic.
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, Faculty of Food and Biochemical Technology, University of Chemistry and Technology, Prague, Technická 3, Prague 166 28, Czech Republic.
| | - Jitka Viktorová
- Department of Biochemistry and Microbiology, Faculty of Food and Biochemical Technology, University of Chemistry and Technology, Prague, Technická 3, Prague 166 28, Czech Republic.
| |
Collapse
|
16
|
Dey N, Kamatchi C, Vickram AS, Anbarasu K, Thanigaivel S, Palanivelu J, Pugazhendhi A, Ponnusamy VK. Role of nanomaterials in deactivating multiple drug resistance efflux pumps - A review. ENVIRONMENTAL RESEARCH 2022; 204:111968. [PMID: 34453898 DOI: 10.1016/j.envres.2021.111968] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/05/2021] [Accepted: 08/19/2021] [Indexed: 06/13/2023]
Abstract
The changes in lifestyle and living conditions have affected not only humans but also microorganisms. As man invents new drugs and therapies, pathogens alter themselves to survive and thrive. Multiple drug resistance (MDR) is the talk of the town for decades now. Many generations of medications have been termed useless as MDR rises among the infectious population. The surge in nanotechnology has brought a new hope in reducing this aspect of resistance in pathogens. It has been observed in several laboratory-based studies that the use of nanoparticles had a synergistic effect on the antibiotic being administered to the pathogen; several resistant strains scummed to the stress created by the nanoparticles and became susceptible to the drug. The major cause of resistance to date is the efflux system, which makes the latest generation of antibiotics ineffective without reaching the target site. If species-specific nanomaterials are used to control the activity of efflux pumps, it could revolutionize the field of medicine and make the previous generation resistant medications active once again. Therefore, the current study was devised to assess and review nanoparticles' role on efflux systems and discuss how specialized particles can be designed towards an infectious host's particular drug ejection systems.
Collapse
Affiliation(s)
- Nibedita Dey
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - C Kamatchi
- Department of Biotechnology, The Oxford College of Science, Bengaluru, India
| | - A S Vickram
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - K Anbarasu
- Department of Bioinformatics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - S Thanigaivel
- Department of Biomedical Engineering, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Jeyanthi Palanivelu
- Department of Biotechnology, Vel Tech Rangarajan Dr. Sagunthala R&D Institute of Science and Technology, Chennai, India
| | | | - Vinoth Kumar Ponnusamy
- Department of Medicinal and Applied Chemistry & Research Center for Environmental Medicine, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital (KMUH), Kaohsiung City, 807, Taiwan; Program of Aquatic Science and Technology, College of Hydrosphere Science, National Kaohsiung University of Science and Technology (NKUST), Kaohsiung City, Taiwan.
| |
Collapse
|
17
|
Garcia ÍR, de Oliveira Garcia FA, Pereira PS, Coutinho HDM, Siyadatpanah A, Norouzi R, Wilairatana P, de Lourdes Pereira M, Nissapatorn V, Tintino SR, Rodrigues FFG. Microbial resistance: The role of efflux pump superfamilies and their respective substrates. Life Sci 2022; 295:120391. [PMID: 35149116 DOI: 10.1016/j.lfs.2022.120391] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 02/02/2022] [Accepted: 02/06/2022] [Indexed: 12/24/2022]
Abstract
The microorganism resistance to antibiotics has become one of the most worrying issues for science due to the difficulties related to clinical treatment and the rapid spread of diseases. Efflux pumps are classified into six groups of carrier proteins that are part of the different types of mechanisms that contribute to resistance in microorganisms, allowing their survival. The present study aimed to carry out a bibliographic review on the superfamilies of carriers in order to understand their compositions, expressions, substrates, and role in intrinsic resistance. At first, a search for manuscripts was carried out in the databases Medline, Pubmed, ScienceDirect, and Scielo, using as descriptors: efflux pump, expression, pump inhibitors and efflux superfamily. For article selection, two criteria were taken into account: for inclusion, those published between 2000 and 2020, including textbooks, and for exclusion, duplicates and academic collections. In this research, 139,615 published articles were obtained, with 312 selected articles and 7 book chapters that best met the aim. From the comprehensive analysis, it was possible to consider that the chromosomes and genetic elements can contain genes encoding efflux pumps and are responsible for multidrug resistance. Even though this is a well-explored topic in the scientific community, understanding the behavior of antibiotics as substrates that increase the expression of pump-encoding genes has challenged medicine. This review study succinctly summarizes the most relevant features of these systems, as well as their contribution to multidrug resistance.
Collapse
Affiliation(s)
| | | | | | | | - Abolghasem Siyadatpanah
- Ferdows School of Paramedical and Health, Birjand University of Medical Sciences, Birjand, Iran
| | - Roghayeh Norouzi
- Department of Pathobiology, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| | - Maria de Lourdes Pereira
- CICECO-Aveiro Institute of Materials & Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Veeranoot Nissapatorn
- School of Allied Health Sciences and Research Excellence Center for Innovation and Health, Walailak University, Thailand
| | | | | |
Collapse
|
18
|
Deka B, Suri M, Sarma S, Devi MV, Bora A, Sen T, Dihingia A, Pahari P, Singh AK. Potentiating the intracellular killing of Staphylococcus aureus by dihydroquinazoline analogues as NorA efflux pump inhibitor. Bioorg Med Chem 2021; 54:116580. [PMID: 34953341 DOI: 10.1016/j.bmc.2021.116580] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 12/04/2021] [Accepted: 12/10/2021] [Indexed: 11/19/2022]
Abstract
Staphylococcus aureus is an emerging human pathogen that has become difficult to treat due to its high resistance against wide range of drugs. Emergence of drug resistant isolates has further convoluted the treatment process. Among different resistance mechanisms, efflux pump proteins play a central role and has made itself a direct approach for therapeutic exploration. To demarcate the role of dihydroquinazoline analogues as NorA efflux pump inhibitor in S. aureus1199B (NorA over producing) strain total seventeen analogues were synthesized and tested for their modulatory effects on norfloxacin and Etbr resistance. Further accumulation assays, bacterial time kill kinetics, cytotoxicity assay were also carried out. The intracellular killing ability of analogues, as EPI was determined using THP-1 monocytes. The binding interaction of analogues with NorA was also predicted. Dihydroquinazoline analogues notably reduced the MIC of norfloxacin and Etbr in S. aureus1199B. In addition to their very low toxicity, they showed high Etbr and norfloxacin accumulation respectively. Further effective over time log reduction in bacterial kill kinetics in presence of these analogues confirmed their role as NorA efflux pump inhibitor. FESEM analysis clearly depicted their effect on the cell surface morphology owing to its lyses. The most significant finding of this study was the ability of analogues to significantly reduce the intracellular S. aureus1199B in human THP-1 monocytes in presence of norfloxacin. Our study has shown for the first time the possibility of developing the dihydroquinazoline analogues as NorA efflux pump inhibitors for S. aureus and control its infection.
Collapse
Affiliation(s)
- Banani Deka
- Biotechnology Group, Biological Sciences and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mrinaly Suri
- Applied Organic Chemistry Group, Chemical Sciences and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam, India
| | - Sangita Sarma
- Biotechnology Group, Biological Sciences and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Moirangthem Veigyabati Devi
- Biotechnology Group, Biological Sciences and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Anamika Bora
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Applied Organic Chemistry Group, Chemical Sciences and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam, India
| | - Tejosmita Sen
- Biotechnology Group, Biological Sciences and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Anjum Dihingia
- Biotechnology Group, Biological Sciences and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Pallab Pahari
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Applied Organic Chemistry Group, Chemical Sciences and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam, India.
| | - Anil Kumar Singh
- Biotechnology Group, Biological Sciences and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
19
|
Dashtbani-Roozbehani A, Brown MH. Efflux Pump Mediated Antimicrobial Resistance by Staphylococci in Health-Related Environments: Challenges and the Quest for Inhibition. Antibiotics (Basel) 2021; 10:antibiotics10121502. [PMID: 34943714 PMCID: PMC8698293 DOI: 10.3390/antibiotics10121502] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 11/30/2021] [Accepted: 11/30/2021] [Indexed: 01/04/2023] Open
Abstract
The increasing emergence of antimicrobial resistance in staphylococcal bacteria is a major health threat worldwide due to significant morbidity and mortality resulting from their associated hospital- or community-acquired infections. Dramatic decrease in the discovery of new antibiotics from the pharmaceutical industry coupled with increased use of sanitisers and disinfectants due to the ongoing COVID-19 pandemic can further aggravate the problem of antimicrobial resistance. Staphylococci utilise multiple mechanisms to circumvent the effects of antimicrobials. One of these resistance mechanisms is the export of antimicrobial agents through the activity of membrane-embedded multidrug efflux pump proteins. The use of efflux pump inhibitors in combination with currently approved antimicrobials is a promising strategy to potentiate their clinical efficacy against resistant strains of staphylococci, and simultaneously reduce the selection of resistant mutants. This review presents an overview of the current knowledge of staphylococcal efflux pumps, discusses their clinical impact, and summarises compounds found in the last decade from plant and synthetic origin that have the potential to be used as adjuvants to antibiotic therapy against multidrug resistant staphylococci. Critically, future high-resolution structures of staphylococcal efflux pumps could aid in design and development of safer, more target-specific and highly potent efflux pump inhibitors to progress into clinical use.
Collapse
|
20
|
Faraag AHI, Shafaa MW, Elkholy NS, Abdel-Hafez LJM. Stress impact of liposomes loaded with ciprofloxacin on the expression level of MepA and NorB efflux pumps of methicillin-resistant Staphylococcus aureus. Int Microbiol 2021; 25:427-446. [PMID: 34822035 DOI: 10.1007/s10123-021-00219-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 10/05/2021] [Accepted: 10/20/2021] [Indexed: 10/19/2022]
Abstract
One mechanism of ciprofloxacin resistance is attributed to chromosomal DNA-encoded efflux pumps such as the MepA and NorB proteins. The goal of this research is to find a way to bypass Staphylococcus aureus' efflux pumps. Because of its high membrane permeability and low association with NorB and MepA efflux proteins, a liposome-encapsulating antibiotic is one of the promising, cost-effective drug carriers and coating mechanisms for overcoming active transport of methicillin-resistant S. aureus (MRSA) multidrug-resistant efflux protein . The calculated "Log Perm RRCK" membrane permeability values of 1,2-distearoyl-sn-glycerol-3-phosphocholine (DSPC) ciprofloxacin liposome-encapsulated (CFL) showed a lower negative value of - 4,652 cm/s and greater membrane permeability than ciprofloxacin free (CPF). The results of RT-qPCR showed that cationic liposomes containing ciprofloxacin in liposome-encapsulated form (CFL) improved CPF antibacterial activity and affinity for negatively charged bacterial cell surface membrane in comparison to free drug and liposome, as it overcame several resistance mechanisms and reduced the expression of efflux pumps. Ciprofloxacin liposome-encapsulated (CFL) is therefore more effective than ciprofloxacin alone. Liposomes can be combined with a variety of drugs that interact with bacterial cell efflux pumps to maintain high sustained levels of antibiotics in bacterial cells.
Collapse
Affiliation(s)
| | - Medhat W Shafaa
- Medical Biophysics Division, Physics Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Nourhan S Elkholy
- Medical Biophysics Division, Physics Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Lina Jamil M Abdel-Hafez
- Department of Microbiology and Immunology, Faculty of Pharmacy, October 6 University, 6 October City, Giza, Egypt
| |
Collapse
|
21
|
Dorado Doncel R, Larcher P, Schmitter C, Zouaghi S. [Bacteria in cystic fibrosis: United they stand - Recent advances in microbiology research through the eyes of ENS students]. Med Sci (Paris) 2021; 37:939-941. [PMID: 34647884 DOI: 10.1051/medsci/2021157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Le dossier que nous présentons a été rédigé par les étudiantes et étudiants de Master 1 de Biologie de l’École Normale Supérieure de Lyon à l’issue de l’UE Microbiologie Moléculaire et Structurale (2020-2021).
Le Master de Biologie de l’ENS de Lyon accueille chaque année environ 40 étudiants en M1 et en M2 et propose une formation de haut niveau à la recherche en biosciences. Chaque étudiant y construit son parcours à la carte, en choisissant ses options parmi un large panel de modules, favorisant ainsi une approche pluridisciplinaire des sciences du vivant, et cela en relation étroite avec les laboratoires de recherche du tissu local, national et international.
En participant à diverses activités scientifiques liées aux UE de leur formation, les étudiants préparent également l’obtention du Diplôme de l’ENS de Lyon, qui valide leur scolarité à l’ENS. La rédaction du présent dossier, qui vise à transmettre de façon claire les messages issus d’une sélection d’articles scientifiques publiés récemment dans le domaine de la microbiologie, constitue l’une de ces activités connexes proposées aux étudiants.
Collapse
Affiliation(s)
- Romane Dorado Doncel
- École normale supérieure de Lyon, Département de biologie, Master Biosciences, Lyon, France
| | - Philéas Larcher
- École normale supérieure de Lyon, Département de biologie, Master Biosciences, Lyon, France
| | - Céline Schmitter
- École normale supérieure de Lyon, Département de biologie, Master Biosciences, Lyon, France
| | - Sarah Zouaghi
- École normale supérieure de Lyon, Département de biologie, Master Biosciences, Lyon, France - ETH Zurich, Institute of Food, Nutrition and Health, Zurich, Suisse
| |
Collapse
|
22
|
RNA-seq-based transcriptome analysis of a cefquinome-treated, highly resistant, and virulent MRSA strain. Microb Pathog 2021; 160:105201. [PMID: 34547409 DOI: 10.1016/j.micpath.2021.105201] [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: 07/14/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 11/23/2022]
Abstract
The emergence and dissemination of methicillin-resistant Staphylococcus aureus (MRSA) strains of animal origin that are resistant to several antibiotics is of great concern. Cefquinome is a fourth-generation cephalosporin developed specifically for veterinary use. The mechanism of MRSA resistance to cefquinome is still not established. Therefore, we designed this study to evaluate the effect of cefquinome on the transcriptome of MRSA1679a, a strain that was isolated from a chicken. The transcriptome analysis indicated that multiple efflux pumps (QacA, NorB, Bcr, and ABCb) were upregulated in MRSA1679a as a resistance mechanism to expel cefquinome. Additionally, penicillin-binding protein 1A was overexpressed, which conferred resistance to cefquinome, a β-lactam antibiotic. Adhesion and the biofilm-forming capacity of the MRSA strain was also enhanced in addition to overexpression of many stress-related genes. Genes related to carbohydrate metabolism, secretion systems, and transport activity were also significantly upregulated in MRSA1679a. In conclusion, global transcription was triggered to overcome the stress induced by cefquinome, and the MRSA1679a showed a great genetic potential to survive in this challenging environment. This study provides a profound understanding of MRSA1679a as a potentially important pathogen and identifies key resistance characteristics of MRSA against cefquinome. Studies should be aimed to demonstrate multidrug resistance mechanisms of virulent strains by exposing to different antibiotic combinations.
Collapse
|
23
|
Pasqua M, Bonaccorsi di Patti MC, Fanelli G, Utsumi R, Eguchi Y, Trirocco R, Prosseda G, Grossi M, Colonna B. Host - Bacterial Pathogen Communication: The Wily Role of the Multidrug Efflux Pumps of the MFS Family. Front Mol Biosci 2021; 8:723274. [PMID: 34381818 PMCID: PMC8350985 DOI: 10.3389/fmolb.2021.723274] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/13/2021] [Indexed: 12/23/2022] Open
Abstract
Bacterial pathogens are able to survive within diverse habitats. The dynamic adaptation to the surroundings depends on their ability to sense environmental variations and to respond in an appropriate manner. This involves, among others, the activation of various cell-to-cell communication strategies. The capability of the bacterial cells to rapidly and co-ordinately set up an interplay with the host cells and/or with other bacteria facilitates their survival in the new niche. Efflux pumps are ubiquitous transmembrane transporters, able to extrude a large set of different molecules. They are strongly implicated in antibiotic resistance since they are able to efficiently expel most of the clinically relevant antibiotics from the bacterial cytoplasm. Besides antibiotic resistance, multidrug efflux pumps take part in several important processes of bacterial cell physiology, including cell to cell communication, and contribute to increase the virulence potential of several bacterial pathogens. Here, we focus on the structural and functional role of multidrug efflux pumps belonging to the Major Facilitator Superfamily (MFS), the largest family of transporters, highlighting their involvement in the colonization of host cells, in virulence and in biofilm formation. We will offer an overview on how MFS multidrug transporters contribute to bacterial survival, adaptation and pathogenicity through the export of diverse molecules. This will be done by presenting the functions of several relevant MFS multidrug efflux pumps in human life-threatening bacterial pathogens as Staphylococcus aureus, Listeria monocytogenes, Klebsiella pneumoniae, Shigella/E. coli, Acinetobacter baumannii.
Collapse
Affiliation(s)
- Martina Pasqua
- Department of Biology and Biotechnology "C. Darwin", Istituto Pasteur Italia, Sapienza Università di Roma, Rome, Italy
| | | | - Giulia Fanelli
- Department of Biology and Biotechnology "C. Darwin", Istituto Pasteur Italia, Sapienza Università di Roma, Rome, Italy
| | - Ryutaro Utsumi
- The Institute of Scientific and Industrial Research (SANKEN), Osaka University, Osaka, Japan
| | - Yoko Eguchi
- Department of Science and Technology on Food Safety, Kindai University, Kinokawa, Japan
| | - Rita Trirocco
- Department of Biology and Biotechnology "C. Darwin", Istituto Pasteur Italia, Sapienza Università di Roma, Rome, Italy
| | - Gianni Prosseda
- Department of Biology and Biotechnology "C. Darwin", Istituto Pasteur Italia, Sapienza Università di Roma, Rome, Italy
| | - Milena Grossi
- Department of Biology and Biotechnology "C. Darwin", Istituto Pasteur Italia, Sapienza Università di Roma, Rome, Italy
| | - Bianca Colonna
- Department of Biology and Biotechnology "C. Darwin", Istituto Pasteur Italia, Sapienza Università di Roma, Rome, Italy
| |
Collapse
|
24
|
Kumar S, Athreya A, Gulati A, Nair RM, Mahendran I, Ranjan R, Penmatsa A. Structural basis of inhibition of a transporter from Staphylococcus aureus, NorC, through a single-domain camelid antibody. Commun Biol 2021; 4:836. [PMID: 34226658 PMCID: PMC8257674 DOI: 10.1038/s42003-021-02357-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 06/10/2021] [Indexed: 12/11/2022] Open
Abstract
Transporters play vital roles in acquiring antimicrobial resistance among pathogenic bacteria. In this study, we report the X-ray structure of NorC, a 14-transmembrane major facilitator superfamily member that is implicated in fluoroquinolone resistance in drug-resistant Staphylococcus aureus strains, at a resolution of 3.6 Å. The NorC structure was determined in complex with a single-domain camelid antibody that interacts at the extracellular face of the transporter and stabilizes it in an outward-open conformation. The complementarity determining regions of the antibody enter and block solvent access to the interior of the vestibule, thereby inhibiting alternating-access. NorC specifically interacts with an organic cation, tetraphenylphosphonium, although it does not demonstrate an ability to transport it. The interaction is compromised in the presence of NorC-antibody complex, consequently establishing a strategy to detect and block NorC and related transporters through the use of single-domain camelid antibodies.
Collapse
Affiliation(s)
- Sushant Kumar
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
- Van Andel Institute, Grand Rapids, MI, USA
| | - Arunabh Athreya
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Ashutosh Gulati
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Rahul Mony Nair
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Ithayaraja Mahendran
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
- Structural Parasitology Lab, International Centre for Genetic engineering and Biotechnology, New Delhi, India
| | - Rakesh Ranjan
- Principal Scientist, ICAR-National Research Centre of Camel (NRCC), Bikaner, India
| | - Aravind Penmatsa
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India.
| |
Collapse
|
25
|
Zhang S, Qu X, Tang H, Wang Y, Yang H, Yuan W, Yue B. Diclofenac Resensitizes Methicillin-Resistant Staphylococcus aureus to β-Lactams and Prevents Implant Infections. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2100681. [PMID: 34258168 PMCID: PMC8261494 DOI: 10.1002/advs.202100681] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/12/2021] [Indexed: 05/25/2023]
Abstract
Implant infections caused by methicillin-resistant Staphylococcus aureus (MRSA) can cause major complications during the perioperative period. Diclofenac, one of the most widely used nonsteroidal anti-inflammatory drugs, is often used to relieve pain and inflammation. In this study, it is found that high-dose diclofenac can inhibit the growth of MRSA, and does not easily induce drug-resistant mutations after continuous passage. However, low-doses diclofenac can resensitize bacteria to β-lactams, which help to circumvent drug resistance and improve the antibacterial efficacy of conventional antibiotics. Further, low-dose diclofenac in combination with β-lactams inhibit MRSA associated biofilm formation in implants. Transcriptomic and proteomic analyses indicate that diclofenac can reduce the expression of genes and proteins associated with β-lactam resistance: mecA, mecR, and blaZ; peptidoglycan biosynthesis: murA, murC, femA, and femB; and biofilm formation: altE and fnbP. Murine implant infection models indicate that diclofenac combined with β-lactams, can substantially alleviate MRSA infections in vivo. In addition, it is investigated that low dose diclofenac can inhibit MRSA antibiotic resistance via the mecA/blaZ pathway and related biofilms in implants. The synergistic effect of diclofenac and β-lactams might have promising applications for preventing perioperative infection, considering its multitarget effects against MRSA.
Collapse
Affiliation(s)
- Shutao Zhang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghai200127China
| | - Xinhua Qu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghai200127China
| | - Haozheng Tang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghai200127China
| | - You Wang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghai200127China
| | - Hongtao Yang
- Department of Plastic & Reconstructive SurgeryThe Ohio State UniversityColumbusOH43210USA
- School of Medical Science and EngineeringBeihang UniversityBeijing100191China
| | - Weien Yuan
- Engineering Research Center of Cell & Therapeutic AntibodyMinistry of EducationSchool of PharmacyShanghai Jiao Tong UniversityShanghai200240China
| | - Bing Yue
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghai200127China
| |
Collapse
|
26
|
Staphylococcus aureus Tet38 Efflux Pump Structural Modeling and Roles of Essential Residues in Drug Efflux and Host Cell Internalization. Infect Immun 2021; 89:IAI.00811-20. [PMID: 33619028 DOI: 10.1128/iai.00811-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/13/2021] [Indexed: 12/29/2022] Open
Abstract
The Staphylococcus aureus Tet38 membrane protein has distinct functions, including drug efflux and host cell attachment and internalization mediated by interaction with host cell CD36. Using structural modeling and site-directed mutagenesis, we identified key amino acids involved in different functions. Tet38, a member of the major facilitator superfamily, is predicted to have 14 transmembrane segments (TMS), 6 cytoplasmic loops, and 7 external loops. Cysteine substitutions of arginine 106 situated at the junction of TMS 4 and external loop L2, and glycine 151 of motif C on TMS 5, resulted in complete or near-complete (8- to 16-fold) reductions in Tet38-mediated resistance to tetracycline, with minimal to no effect on A549 host cell internalization. In contrast, a three-amino-acid deletion, F411P412G413, in external loop L7 situated between TMS 13 and 14 led to a decrease of 4-fold in S. aureus internalization by A549 cells and a partial effect on tetracycline resistance (4-fold reduction). A three-amino-acid deletion, D38D39L40, in external loop L1 situated between TMS-1 and TMS-2, had a similar partial effect on tetracycline resistance but did not affect cell internalization. Using an Ni column retention assay, we showed further that the L7, but not the L1, deletion impaired binding to CD36. Thus, the L7 domain of Tet38 is key for interaction with CD36 and host cell internalization, and amino acids R106 and G151 (TMSs 4 and 5) are particularly important for tetracycline resistance without affecting internalization.
Collapse
|
27
|
de Araújo ACJ, Freitas PR, Dos Santos Barbosa CR, Muniz DF, Ribeiro-Filho J, Tintino SR, Júnior JPS, Filho JMB, de Sousa GR, Coutinho HDM. Modulation of Drug Resistance by Limonene: Inhibition of Efflux Pumps in Staphylococcus aureus Strains RN-4220 and IS-58. Curr Drug Metab 2021; 22:110-113. [PMID: 33397229 DOI: 10.2174/1389200221999210104204718] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/09/2020] [Accepted: 09/09/2020] [Indexed: 11/22/2022]
Abstract
AIMS This study aimed to investigate the potential of limonene as an efflux pump (EP) inhibitor in Staphylococcus aureus strains, RN-4220 and IS-58, which carry EPs for erythromycin (MrsA) and tetracycline (TetK), respectively. BACKGROUND The evolution of bacterial resistance mechanisms over time has impaired the action of most classes of antibiotics. Staphylococcus aureus is a notable bacterium, with high pathogenic potential and demonstrated resistance to conventional antibiotics. Considering the importance of discovering novel compounds to combat antibiotic resistance, our group previously demonstrated the antibacterial properties of limonene, a compound present in the essential oils of several plant species. OBJECTIVE This study aimed to investigate the potential of limonene as an efflux pump (EP) inhibitor in Staphylococcus aureus strains RN-4220 and IS-58, which carry EPs for erythromycin (MrsA) and tetracycline (TetK), respectively. METHODS The minimum inhibitory concentrations (MIC) of limonene and other efflux pump inhibitors were determined through the broth microdilution method. A reduction in the MIC of ethidium bromide was used as a parameter of EP inhibition. RESULT While limonene was not shown to exhibit direct antibacterial effects against EP-carrying strains, in association with ethidium bromide and antibiotics, this compound demonstrated enhanced antibacterial activity, indicating the inhibition of the MrsA and TetK pumps. CONCLUSION In conclusion, this pioneering study demonstrated the effectiveness of limonene as an EP inhibitor in S. aureus strains, RN-4220 and IS-58. Nevertheless, further studies are required to characterize the molecular mechanisms associated with limonene-mediated EP inhibition.
Collapse
Affiliation(s)
- Ana C J de Araújo
- Department of Biological Chemistry, Regional University of Cariri, URCA, Crato, CE, Brazil
| | - Priscilla R Freitas
- Department of Biological Chemistry, Regional University of Cariri, URCA, Crato, CE, Brazil
| | | | - Débora F Muniz
- Department of Biological Chemistry, Regional University of Cariri, URCA, Crato, CE, Brazil
| | - Jaime Ribeiro-Filho
- Goncalo Moniz Institute, Oswaldo Cruz Foundation, IGM-FIOCRUZ/BA, Salvador, BA, Brazil
| | - Saulo R Tintino
- Department of Biological Chemistry, Regional University of Cariri, URCA, Crato, CE, Brazil
| | - José P S Júnior
- Federal University of Paraiba - UFPB, Joao Pessoa, Paraiba, Brazil
| | - José M B Filho
- Federal University of Paraiba - UFPB, Joao Pessoa, Paraiba, Brazil
| | | | - Henrique D M Coutinho
- Department of Biological Chemistry, Regional University of Cariri, URCA, Crato, CE, Brazil
| |
Collapse
|
28
|
Zhang S, Tang H, Wang Y, Nie B, Yang H, Yuan W, Qu X, Yue B. Antibacterial and antibiofilm effects of flufenamic acid against methicillin-resistant Staphylococcus aureus. Pharmacol Res 2020; 160:105067. [PMID: 32650057 DOI: 10.1016/j.phrs.2020.105067] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/19/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) infections are one of the most serious surgery complications, and their prevention is of utmost importance. Flufenamic acid is a non-steroid anti-inflammatory drug approved for clinical use to relieve inflammation and pain in rheumatoid arthritis patients. In this study, we explored the antibacterial efficacy of flufenamic acid and the mechanisms underlying this effect. By using minimal inhibitory concentration (MIC), time-kill, resistance induction assays, and the antibiotic synergy test, we demonstrated that flufenamic acid inhibited the growth of methicillin-resistant staphylococci and did not induce resistance when it was used at the MIC. Furthermore, flufenamic acid acted synergistically with the beta-lactam antibiotic oxacillin and did not show significant toxicity toward mammalian cells. The biofilm inhibition assay revealed that flufenamic acid could prevent biofilm formation on medical implants and destroy the ultrastructure of the bacterial cell wall. RNA sequencing and quantitative RT-PCR indicated that flufenamic acid inhibited the expression of genes associated with peptidoglycan biosynthesis, beta-lactam resistance, quorum sensing, and biofilm formation. Furthermore, flufenamic acid efficiently ameliorated a local infection caused by MRSA in mice. In conclusion, flufenamic acid may be a potent therapeutic compound against MRSA infections and a promising candidate for antimicrobial coating of implants and surgical devices.
Collapse
Affiliation(s)
- Shutao Zhang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haozheng Tang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - You Wang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bin'en Nie
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hongtao Yang
- Department of Plastic & Reconstructive Surgery, The Ohio State University, Columbus, OH 43210, United States
| | - Weien Yuan
- Ministry of Education Engineering Research Center of Cell & Therapeutic Antibody, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Xinhua Qu
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Bing Yue
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
29
|
The Great ESKAPE: Exploring the Crossroads of Bile and Antibiotic Resistance in Bacterial Pathogens. Infect Immun 2020; 88:IAI.00865-19. [PMID: 32661122 DOI: 10.1128/iai.00865-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Throughout the course of infection, many pathogens encounter bactericidal conditions that threaten the viability of the bacteria and impede the establishment of infection. Bile is one of the most innately bactericidal compounds present in humans, functioning to reduce the bacterial burden in the gastrointestinal tract while also aiding in digestion. It is becoming increasingly apparent that pathogens successfully resist the bactericidal conditions of bile, including bacteria that do not normally cause gastrointestinal infections. This review highlights the ability of Enterococcus, Staphylococcus, Klebsiella, Acinetobacter, Pseudomonas, Enterobacter (ESKAPE), and other enteric pathogens to resist bile and how these interactions can impact the sensitivity of bacteria to various antimicrobial agents. Given that pathogen exposure to bile is an essential component to gastrointestinal transit that cannot be avoided, understanding how bile resistance mechanisms align with antimicrobial resistance is vital to our ability to develop new, successful therapeutics in an age of widespread and increasing antimicrobial resistance.
Collapse
|
30
|
The transcriptome of Listeria monocytogenes during co-cultivation with cheese rind bacteria suggests adaptation by induction of ethanolamine and 1,2-propanediol catabolism pathway genes. PLoS One 2020; 15:e0233945. [PMID: 32701964 PMCID: PMC7377500 DOI: 10.1371/journal.pone.0233945] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/01/2020] [Indexed: 02/07/2023] Open
Abstract
The survival of Listeria (L.) monocytogenes in foods and food production environments (FPE) is dependent on several genes that increase tolerance to stressors; this includes competing with intrinsic bacteria. We aimed to uncover genes that are differentially expressed (DE) in L. monocytogenes sequence type (ST) 121 strain 6179 when co-cultured with cheese rind bacteria. L. monocytogenes was cultivated in broth or on plates with either a Psychrobacter or Brevibacterium isolate from cheese rinds. RNA was extracted from co-cultures in broth after two or 12 hours and from plates after 24 and 72 hours. Broth co-cultivations with Brevibacterium or Psychrobacter yielded up to 392 and 601 DE genes, while plate co-cultivations significantly affected the expression of up to 190 and 485 L. monocytogenes genes, respectively. Notably, the transcription of virulence genes encoding the Listeria adhesion protein and Listeriolysin O were induced during plate and broth co-cultivations. The expression of several systems under the control of the global stress gene regulator, σB, increased during co-cultivation. A cobalamin-dependent gene cluster, responsible for the catabolism of ethanolamine and 1,2-propanediol, was upregulated in both broth and plate co-cultures conditions. Finally, a small non-coding (nc)RNA, Rli47, was induced after 72 hours of co-cultivation on plates and accounted for 50-90% of the total reads mapped to L. monocytogenes. A recent study has shown that Rli47 may contribute to L. monocytogenes stress survival by slowing growth during stress conditions through the suppression of branch-chained amino acid biosynthesis. We hypothesize that Rli47 may have an impactful role in the response of L. monocytogenes to co-cultivation by regulating a complex network of metabolic and virulence mechanisms.
Collapse
|
31
|
Figueredo FG, Ramos ITL, Paz JA, Silva TMS, Câmara CA, de Morais Oliveira-Tintino CD, Tintino SR, de Farias PAM, Menezes IRAD, Coutinho HDM, Fonteles MMDF. Effect of hydroxyamines derived from lapachol and norlachol against Staphylococcus aureus strains carrying the NorA efflux pump. INFECTION GENETICS AND EVOLUTION 2020; 84:104370. [PMID: 32445918 DOI: 10.1016/j.meegid.2020.104370] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/13/2020] [Accepted: 05/16/2020] [Indexed: 02/07/2023]
Abstract
Isolated substances and those organically synthesized have stood out over the years for their therapeutic properties, including their antibacterial activity. These compounds may be an alternative to the production of new antibiotics or may have the ability to potentiate the action of preexisting ones. In this context, the objective of this study was to evaluate the in vitro antibacterial and efflux pump inhibitory activity of hydroxyamines derived from lapachol and norlachol, more specifically the compounds 2-(2-Hydroxyethylamino)-3-(3-methyl-2-butenyl)-1,4 dihydro-1,4-naphthalenedione, 2-(2-Hydroxyethylamino)-3-(2-methyl-propenyl)[1,4]naphthoquinone and 2-(3-Hydroxypropylamino)-3-(3-methyl-2-butenyl)-[1,4]naphthoquinone, against Staphylococcus aureus strains carrying the NorA efflux pump mechanism. The substances were synthesized from 2-hydroxy-quinones, lapachol and nor-lapachol, obtaining the corresponding 2-methoxylated derivatives via dimethyl sulfate alkylation in a basic medium, which then reacted chemoselectively with 2-ethanolamine and 3-propanolamine to form the corresponding amino alcohols. All three molecules underwent a virtual structure-based analysis (docking). The antibacterial activity of the substances was measured by determining their Minimum Inhibitory Concentration (MIC) and a microdilution assay was performed to verify efflux pump inhibition using the substances at a sub-inhibitory concentration. The results were subjected to statistical analysis using an analysis of variance (ANOVA) followed by Bonferroni's post hoc test. The substances obtained MIC values ≥1024 μg/mL, however, a significant reduction of their MICs was observed when the substances were associated with norfloxacin and ethidium bromide, with this effect being attributed to efflux pump inhibition. Following a virtual analysis based on its structure (docking), information regarding the affinity of new ligands for the ABC efflux pump were observed, thus contributing to the understanding of their mechanism of molecular interactions and the discovery of functional ligands associated with a reduction in bacterial resistance.
Collapse
Affiliation(s)
- Fernando Gomes Figueredo
- Postgraduate Program in Development and Technological Innovation in Medicines, Federal University of Ceará, CEP 60.430-370, Fortaleza, CE, Brazil; Department of Microbiology, Faculdade de Medicina Estácio de Juazeiro do Norte- CEP 63048-080, Juazeiro do Norte, CE, Brazil
| | - Ingrid T L Ramos
- Department of Chemistry, Universidade Federal Rural de Pernambuco, Recife, PE, Brazil
| | - Josinete A Paz
- Department of Chemistry, Universidade Federal Rural de Pernambuco, Recife, PE, Brazil
| | - Tania M S Silva
- Department of Chemistry, Universidade Federal Rural de Pernambuco, Recife, PE, Brazil
| | - Celso A Câmara
- Department of Chemistry, Universidade Federal Rural de Pernambuco, Recife, PE, Brazil
| | | | - Saulo Relison Tintino
- Department of Biological Chemistry, Regional University of Cariri, CEP, 63105-000 - Crato, CE, Brazil
| | - Pablo Antônio Maia de Farias
- Department of Microbiology, Faculdade de Medicina Estácio de Juazeiro do Norte- CEP 63048-080, Juazeiro do Norte, CE, Brazil
| | - Irwin Rose Alencar de Menezes
- Department of of Biological Chemistry, Laboratory of Pharmacology and Molecular Chemistry, Regional University of Cariri, CEP, 63105-000 - Crato, CE, Brazil.
| | | | - Marta Maria de F Fonteles
- Postgraduate Program in Development and Technological Innovation in Medicines, Federal University of Ceará, CEP 60.430-370, Fortaleza, CE, Brazil
| |
Collapse
|
32
|
Role of antibiotic stress in phenotypic switching to persister cells of antibiotic-resistant Staphylococcus aureus. ANN MICROBIOL 2020. [DOI: 10.1186/s13213-020-01552-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Abstract
Purpose
This study was designed to evaluate phenotypic and genotypic properties of persister cells formed by Staphylococcus aureus ATCC 15564 (SAWT), oxacillin-induced S. aureus (SAOXA), ciprofloxacin-induced S. aureus (SACIP), and clinically isolated multidrug-resistant S. aureus CCARM 3080 (SAMDR).
Methods
The dose-dependent biphasic killing patterns were observed for SAWT, SAOXA, SACIP, and SAMDR in response to twofold minimum inhibitory concentrate (MIC) of ciprofloxacin. The surviving cells of SAWT, SAOXA, SACIP, and SAMDR after twofold MIC of ciprofloxacin treatment were analyzed using a metabolic-based assay to estimate the fractions of persister cells.
Results
The least persister formation was induced in SACIP after twofold MIC of ciprofloxacin treatment, showing 58% of persistence. The lowest fitness cost of resistance was observed for the recovered persister cells of SACIP (relative fitness = 0.95), followed by SAMDR (relative fitness = 0.70), while the highest fitness cost was observed for SAWT (relative fitness = 0.26). The mRNA transcripts were analyzed by RT-PCR assay in recovered persister cells pre-incubated with ciprofloxacin. The highest expression levels of stress-related genes (dnaK and groEL) and efflux pump-related genes (mepR, norA, and norB) were observed in the recovered persister cells of SAOXA and SAMDR.
Conclusion
This study provides valuable information for understanding crosstalk between antibiotic resistance, tolerance, and persistence in different antibiotic-resistant S. aureus strains.
Collapse
|
33
|
The prevalence of antiseptic tolerance genes among staphylococci and enterococci in a pediatric population. Infect Control Hosp Epidemiol 2020; 40:333-340. [PMID: 30887943 DOI: 10.1017/ice.2019.3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The smr and qacA/B genes in Staphylococcus aureus confer tolerance to antiseptics and are associated with nosocomial acquisition of infection and underlying medical conditions. Such antiseptic tolerance (AT) genes have also been reported in coagulase-negative staphylococci (CoNS) and enterococci, however, few data are available regarding their prevalence. We sought to describe the frequency of AT genes among bloodstream isolates of S. aureus, CoNS and enterococci at Texas Children's Hospital (TCH). METHODS Banked CoNS, S. aureus and enterococci isolated from blood cultures collected bewteen October 1, 2016, and October 1, 2017, were obtained from the TCH clinical microbiology laboratory. All isolates underwent polymerase chain reaction (PCR) assay for the qacA/B and smr genes. Medical records were reviewed for all cases. RESULTS In total, 103 CoNS, 19 Enterococcus spp, and 119 S. aureus isolates were included in the study, and 80.6% of the CoNS possessed at least 1 AT gene compared to 37% of S. aureus and 43.8% of E. faecalis isolates (P < .001). Among CoNS bloodstream isolates, the presence of either AT gene was strongly associated with nosocomial infection (P < .001). The AT genes in S. aureus were associated with nosocomial infection (P = .025) as well as the diagnosis of central-line-associated bloodstream infection (CLABSI; P = .04) and recent hospitalizations (P < .001). We found no correlation with genotypic AT in E. faecalis and any clinical variable we examined. CONCLUSIONS Antiseptic tolerance is common among bloodstream staphylococci and E. faecalis isolates at TCH. Among CoNS, the presence of AT genes is strongly correlated with nosocomial acquisition of infection, consistent with previous studies in S. aureus. These data suggest that the healthcare environment contributes to AT among staphylococci.
Collapse
|
34
|
Epidemiology of efflux pumps genes mediating resistance among Staphylococcus aureus; A systematic review. Microb Pathog 2019; 139:103850. [PMID: 31706002 DOI: 10.1016/j.micpath.2019.103850] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/22/2019] [Accepted: 11/05/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Efflux of antibiotics is an effective resistance mechanism among antibiotic-resistant Staphylococcus aureus. This systematic review aims to evaluate the frequency and expression of efflux pump genes in S.aureus around the world. METHOD A comprehensive literature search of several databases (Medline Pub Med, ISI, Scopus, Google Scholar, ISC, Science direct and Persian Journals Online, and citation lists) was performed. We considered published studies from 2001 to 2018. Articles reporting the prevalence and expression of efflux pump genes were selected. RESULT Among 183 articles, 36 studies were selected. Of the 36, 23 articles were conducted in Asia.6 in Europe, 5 in America and 2 in African countries. In most of these studies norA, norB, qacA/B genes were commonly evaluated by molecular methods. The presence of efflux pump genes such as norA, norB, norC, mepA, mdeA, qacA/B was detected by PCR in 21 studies and over-expression of genes were reported in 13 studies. The most frequently reported genes in Asia were norA (75%), norB (60%), mepA (35%), mdeA (33%) and qacA/B (20.8%). In European studies, the prevalence of norB was mostly reported among S.aureus isolates and norA and qacA/B were commonly found in similar studies in America. The investigation of gene expression patterns showed that norA was most frequent single-pattern in Asia and America, norB or mdeA in Europe. CONCLUSION According to this study MDR efflux pumps not only cause high-level resistance but also it considerably associated with over-expression of these genes. Due to the selective pressure on MRSA isolate, the enormous diversity of plasmid-encoded genes had been recorded in different regions, owing to the various numbers and types of isolates in each study or types of disinfectants for general use.
Collapse
|
35
|
Alav I, Sutton JM, Rahman KM. Role of bacterial efflux pumps in biofilm formation. J Antimicrob Chemother 2019; 73:2003-2020. [PMID: 29506149 DOI: 10.1093/jac/dky042] [Citation(s) in RCA: 258] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Efflux pumps are widely implicated in antibiotic resistance because they can extrude the majority of clinically relevant antibiotics from within cells to the extracellular environment. However, there is increasing evidence from many studies to suggest that the pumps also play a role in biofilm formation. These studies have involved investigating the effects of efflux pump gene mutagenesis and efflux pump inhibitors on biofilm formation, and measuring the levels of efflux pump gene expression in biofilms. In particular, several key pathogenic species associated with increasing multidrug resistance, such as Acinetobacter baumannii, Escherichia coli, Pseudomonas aeruginosa and Staphylococcus aureus, have been investigated, whilst other studies have focused on Salmonella enterica serovar Typhimurium as a model organism and problematic pathogen. Studies have shown that efflux pumps, including AcrAB-TolC of E. coli, MexAB-OprM of P. aeruginosa, AdeFGH of A. baumannii and AcrD of S. enterica, play important roles in biofilm formation. The substrates for such pumps, and whether changes in their efflux activity affect biofilm formation directly or indirectly, remain to be determined. By understanding the roles that efflux pumps play in biofilm formation, novel therapeutic strategies can be developed to inhibit their function, to help disrupt biofilms and improve the treatment of infections. This review will discuss and evaluate the evidence for the roles of efflux pumps in biofilm formation and the potential approaches to overcome the increasing problem of biofilm-based infections.
Collapse
Affiliation(s)
- Ilyas Alav
- School of Cancer and Pharmaceutical Science, King's College London, London, UK
| | - J Mark Sutton
- Public Health England, National Infection Service, Porton Down, Salisbury, UK
| | | |
Collapse
|
36
|
Shokoofeh N, Moradi-Shoeili Z, Naeemi AS, Jalali A, Hedayati M, Salehzadeh A. Biosynthesis of Fe 3O 4@Ag Nanocomposite and Evaluation of Its Performance on Expression of norA and norB Efflux Pump Genes in Ciprofloxacin-Resistant Staphylococcus aureus. Biol Trace Elem Res 2019; 191:522-530. [PMID: 30788722 DOI: 10.1007/s12011-019-1632-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 01/01/2019] [Indexed: 10/27/2022]
Abstract
At present, the universal health problem with Staphylococcus aureus is the emergence of multidrug-resistant strains due to the overuse of antibiotics. Drug extrusion through efflux pumps is one of the bacterial mechanisms to neutralize the bactericidal effect of antibiotics. The antibacterial activity of silver nanoparticle as well as Fe3O4 nanoparticle had been previously studied and widely described. Today, the development of green methods for nanomaterial synthesis is an important aspect of research in the field of nanotechnology. Here, we report the biosynthesis and characterization of Fe3O4@Ag nanocomposite by Spirulina platensis cyanobacterium and it impacts on the expression of efflux pump genes in ciprofloxacin-resistant S. aureus (CRSA). The physical properties of biosynthesized nanocomposite measured and confirmed by ultraviolet-visible spectroscopy, Fourier-transform infrared spectroscopy, X-ray diffraction, energy-dispersive X-ray spectroscopy, and scanning and transmission electron microscopy. The minimum inhibitory concentration (MIC) of ciprofloxacin in CRSA strains was determined in the presence of Fe3O4@Ag nanoparticles by broth microdilution method. The effect of Fe3O4@Ag nanocomposite on the expression of norA and norB genes was evaluated by real-time PCR. The physical analysis confirmed well-dispersed, highly stable, and mostly spherical Fe3O4/Ag NPs with the average size of 30-68 nm. The results of antibacterial tests showed the synergistic effects of nanocomposite and antibiotics in MIC reduction. Additionally, in the presence of Fe3O4@Ag nanocomposite, the expression of norA and norB genes was decreased more than twofold compared to control. In conclusion, the Fe3O4/Ag nanocomposite can use as an effective inhibitor of antibiotic resistance in medicine.
Collapse
Affiliation(s)
- Nastaran Shokoofeh
- Department of Biology, Rasht Branch, Islamic Azad University, Rasht, Iran
| | - Zeinab Moradi-Shoeili
- Department of Chemistry, Faculty of Sciences, University of Guilan, P.O. Box 41335-1914, Rasht, Iran
| | - Akram Sadat Naeemi
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - Amir Jalali
- Department of Biology, Faculty of Science, Arak University, Arak, Iran
| | - Mohammad Hedayati
- Department of Cell and Molecular Biology, University of Guilan, Rasht, Iran
| | - Ali Salehzadeh
- Department of Biology, Rasht Branch, Islamic Azad University, Rasht, Iran.
| |
Collapse
|
37
|
Wagner Mackenzie B, Baker J, Douglas RG, Taylor MW, Biswas K. Detection and quantification of Staphylococcus in chronic rhinosinusitis. Int Forum Allergy Rhinol 2019; 9:1462-1469. [PMID: 31483577 DOI: 10.1002/alr.22425] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 08/05/2019] [Accepted: 08/09/2019] [Indexed: 11/10/2022]
Abstract
BACKGROUND The sinonasal microbiota has been implicated in chronic rhinosinusitis (CRS) pathogenesis, particularly related to the presence of Staphylococcus aureus. Staphylococcus epidermidis is also prevalent within the sinonasal microbiota and may inhibit S. aureus colonization. We investigated polymerase chain reaction (PCR) primer pairs for measuring absolute abundances of S. aureus and S. epidermidis, then compared bacterial community composition and absolute abundances of these species between CRS patients and controls. METHODS Six candidate Staphylococcus species-specific primer pairs were tested in silico and in vitro against pure bacterial isolates. Quantitative PCR (qPCR) for absolute quantification of S. aureus, S. epidermidis, and overall bacterial load were assessed in 40 CRS (CRS without nasal polyposis [CRSsNP] = 22, CRS with nasal polyposis [CRSwNP] = 18) patients and 14 controls. Amplicon sequencing of the V3-V4 hypervariable regions of the 16S ribosomal RNA (rRNA) bacterial gene were conducted to investigate community composition. RESULTS Primer pairs targeting the gmk gene of S. aureus and nrd gene from S. epidermidis were the most specific and sensitive primers. S. aureus (CRSsNP = 81.8% occurrence, CRSwNP = 83%, control = 92.9%) and S. epidermidis (CRSsNP = 95.5%, CRSwNP = 100%, control = 92.9%) were very prevalent, as indicated by qPCR results. Both CRSsNP and CRSwNP had significantly (p < 0.05) higher bacterial load when compared with controls (p < 0.05 for both). No significant correlation was observed between S. aureus and S. epidermidis abundances (p > 0.05). CONCLUSION Bacterial community sequencing detected Staphylococcus-assigned sequences in nearly all patients; however, it could not differentiate between S. aureus and S. epidermidis. Here, we present primer pairs that can distinguish between these species. We report a very high prevalence of S. aureus in both CRS patients and controls.
Collapse
Affiliation(s)
| | - Jesse Baker
- Department of Surgery, The University of Auckland, Auckland, New Zealand.,School of Biological Sciences and Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Richard G Douglas
- Department of Surgery, The University of Auckland, Auckland, New Zealand
| | - Michael W Taylor
- School of Biological Sciences and Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Kristi Biswas
- School of Biological Sciences and Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
38
|
Nejabatdoust A, Zamani H, Salehzadeh A. Functionalization of ZnO Nanoparticles by Glutamic Acid and Conjugation with Thiosemicarbazide Alters Expression of Efflux Pump Genes in Multiple Drug-Resistant Staphylococcus aureus Strains. Microb Drug Resist 2019; 25:966-974. [DOI: 10.1089/mdr.2018.0304] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Armin Nejabatdoust
- Department of Biology, Rasht Branch, Islamic Azad University, Rasht, Iran
| | - Hojjatolah Zamani
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Ali Salehzadeh
- Department of Biology, Rasht Branch, Islamic Azad University, Rasht, Iran
| |
Collapse
|
39
|
Pasqua M, Grossi M, Zennaro A, Fanelli G, Micheli G, Barras F, Colonna B, Prosseda G. The Varied Role of Efflux Pumps of the MFS Family in the Interplay of Bacteria with Animal and Plant Cells. Microorganisms 2019; 7:microorganisms7090285. [PMID: 31443538 PMCID: PMC6780985 DOI: 10.3390/microorganisms7090285] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/19/2019] [Accepted: 08/21/2019] [Indexed: 12/20/2022] Open
Abstract
Efflux pumps represent an important and large group of transporter proteins found in all organisms. The importance of efflux pumps resides in their ability to extrude a wide range of antibiotics, resulting in the emergence of multidrug resistance in many bacteria. Besides antibiotics, multidrug efflux pumps can also extrude a large variety of compounds: Bacterial metabolites, plant-produced compounds, quorum-sensing molecules, and virulence factors. This versatility makes efflux pumps relevant players in interactions not only with other bacteria, but also with plant or animal cells. The multidrug efflux pumps belonging to the major facilitator superfamily (MFS) are widely distributed in microbial genomes and exhibit a large spectrum of substrate specificities. Multidrug MFS efflux pumps are present either as single-component transporters or as tripartite complexes. In this review, we will summarize how the multidrug MFS efflux pumps contribute to the interplay between bacteria and targeted host cells, with emphasis on their role in bacterial virulence, in the colonization of plant and animal host cells and in biofilm formation. We will also address the complexity of these interactions in the light of the underlying regulatory networks required for the effective activation of efflux pump genes.
Collapse
Affiliation(s)
- Martina Pasqua
- Istituto Pasteur Italia, Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza Università di Roma, Via dei Sardi 70, 00185 Rome, Italy
| | - Milena Grossi
- Istituto Pasteur Italia, Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza Università di Roma, Via dei Sardi 70, 00185 Rome, Italy
| | - Alessandro Zennaro
- Istituto Pasteur Italia, Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza Università di Roma, Via dei Sardi 70, 00185 Rome, Italy
| | - Giulia Fanelli
- Istituto Pasteur Italia, Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza Università di Roma, Via dei Sardi 70, 00185 Rome, Italy
| | - Gioacchino Micheli
- Istituto di Biologia e Patologia Molecolari, Consiglio Nazionale delle Ricerche (CNR), P.le A. Moro 5, 00185 Roma, Italy
| | - Frederic Barras
- Département de Microbiologie, Institut Pasteur, 75015 Paris, France
- Équipe de Recherche Labellisée (ERL) Microbiology, Centre National de la Recherche Scientifique (CNRS), 13009 Marseille, France
| | - Bianca Colonna
- Istituto Pasteur Italia, Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza Università di Roma, Via dei Sardi 70, 00185 Rome, Italy
| | - Gianni Prosseda
- Istituto Pasteur Italia, Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza Università di Roma, Via dei Sardi 70, 00185 Rome, Italy.
| |
Collapse
|
40
|
Lowrence RC, Subramaniapillai SG, Ulaganathan V, Nagarajan S. Tackling drug resistance with efflux pump inhibitors: from bacteria to cancerous cells. Crit Rev Microbiol 2019; 45:334-353. [PMID: 31248314 DOI: 10.1080/1040841x.2019.1607248] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Drug resistance is a serious concern in a clinical setting jeopardizing treatment for both infectious agents and cancers alike. The wide-spread emergence of multi-drug resistant (MDR) phenotypes from bacteria to cancerous cells necessitates the need to target resistance mechanisms and prevent the emergence of resistant mutants. Drug efflux seems to be one of the preferred approaches embraced by both microbial and mammalian cells alike, to thwart the action of chemotherapeutic agents thereby leading to a drug resistant phenotype. Relative to microbes, which predominantly employs proton motive force (PMF) powered, Major Facilitator Superfamily (MFS)/Resistance Nodulation and Division (RND) classes of efflux pumps to efflux drugs, cancerous cells preferentially use ATP fuelled ATP binding cassette (ABC) transporters to extrude chemotherapeutic agents. The prevalence, evolutionary characteristics and overlapping functions of ABC transporters have been highlighted in this review. Additionally, we outline the role of ABC pumps in conferring MDR phenotype to both bacteria and cancerous cells and underscore the importance of efflux pump inhibitors (EPI) to mitigate drug resistance. Based on the literature reports and analysis, we reason out feasibility of employing bacteria as a tool to screen for EPI's targeting ABC pumps of cancerous cells.
Collapse
Affiliation(s)
- Rene Christena Lowrence
- a Department of Molecular Biology and Biotechnology, University of Sheffield , Sheffield , UK
| | | | | | - Saisubramanian Nagarajan
- c Department of Biotechnology, School of Chemical and Biotechnology, SASTRA Deemed to be University , Thanjavur , India
| |
Collapse
|
41
|
Cheng AV, Wuest WM. Signed, Sealed, Delivered: Conjugate and Prodrug Strategies as Targeted Delivery Vectors for Antibiotics. ACS Infect Dis 2019; 5:816-828. [PMID: 30969100 PMCID: PMC6570538 DOI: 10.1021/acsinfecdis.9b00019] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Innate and developed resistance mechanisms of bacteria to antibiotics are obstacles in the design of novel drugs. However, antibacterial prodrugs and conjugates have shown promise in circumventing resistance and tolerance mechanisms via directed delivery of antibiotics to the site of infection or to specific species or strains of bacteria. The selective targeting and increased permeability and accumulation of these prodrugs not only improves efficacy over unmodified drugs but also reduces off-target effects, toxicity, and development of resistance. Herein, we discuss some of these methods, including sideromycins, antibody-directed prodrugs, cell penetrating peptide conjugates, and codrugs.
Collapse
Affiliation(s)
- Ana V. Cheng
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - William M. Wuest
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
- Emory Antibiotic Resistance Center, Emory School of Medicine, 201 Dowman Drive, Atlanta, Georgia 30322, United States
| |
Collapse
|
42
|
Chen C, Hooper DC. Effect of Staphylococcus aureus Tet38 native efflux pump on in vivo response to tetracycline in a murine subcutaneous abscess model. J Antimicrob Chemother 2019; 73:720-723. [PMID: 29216347 DOI: 10.1093/jac/dkx432] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/24/2017] [Indexed: 11/13/2022] Open
Abstract
Objectives Staphylococcus aureus native efflux pump Tet38 confers resistance to tetracycline when overexpressed. tet38 expression is selectively upregulated in infection sites. This study evaluated the effect of Tet38 on tetracycline response in a murine subcutaneous abscess model. Methods S. aureus MW2 and its tet38 mutant were injected subcutaneously on the opposite flanks of each mouse. The infected mice were treated with tetracycline (10 mg/kg) or PBS (control) intraperitoneally every 12 h. The efficacy of tetracycline against S. aureus was measured by the relative change in viable bacteria in the abscesses 24 h after infection compared with the initial inoculum. Plasmid-based tet38-complemented strains were created and used to infect the mice followed by tetracycline or PBS treatment. Results The increased bacterial loads of S. aureus MW2 and its tet38 mutant in the abscess after 24 h were similar. Tetracycline produced significant decreases of both MW2 and the tet38 mutant compared with control. Although the tetracycline MICs for MW2 and the tet38 mutant did not differ in vitro, the antibacterial effect of tetracycline was significantly 2-fold greater in the tet38 mutant compared with the MW2 parental strain in vivo with a decrease of 0.67 ± 0.21 and 0.35 ± 0.19 log10 cfu/abscess, respectively (P < 0.05). The increased tetracycline activity in the tet38 mutant was complemented by plasmid-encoded tet38. Conclusions This study demonstrated that selective increased expression of tet38 in an abscess can affect tetracycline efficacy against S. aureus in vivo, highlighting an effect of native efflux pumps on response to therapy not reflected by testing in vitro.
Collapse
Affiliation(s)
- Chunhui Chen
- Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - David C Hooper
- Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
43
|
Yong YY, Dykes GA, Choo WS. Biofilm formation by staphylococci in health-related environments and recent reports on their control using natural compounds. Crit Rev Microbiol 2019; 45:201-222. [PMID: 30786799 DOI: 10.1080/1040841x.2019.1573802] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Staphylococci are Gram-positive bacteria that are ubiquitous in the environment and able to form biofilms on a range of surfaces. They have been associated with a range of human health issues such as medical device-related infection, localized skin infection, or direct infection caused by toxin production. The extracellular material produced by these bacteria resists antibiotics and host defence mechanism which complicates the treatment process. The commonly reported Staphylococcus species are Staphylococcus aureus and S. epidermidis as they inhabit human bodies. However, the emergence of other staphylococci, such as S. haemolyticus, S. lugdunensis, S. saprophyticus, S. capitis, S. saccharolyticus, S. warneri, S. cohnii, and S. hominis, is also of concern and they have been associated with biofilm formation. This review critically assesses recent cases on the biofilm formation by S. aureus, S. epidermidis, and other staphylococci reported in health-related environments. The control of biofilm formation by staphylococci using natural compounds is specifically discussed as they represent potential anti-biofilm agents which may reduce the burden of antibiotic resistance.
Collapse
Affiliation(s)
- Yi Yi Yong
- a School of Science , Monash University Malaysia , Selangor , Malaysia
| | - Gary A Dykes
- b School of Public Health , Curtin University , Bentley , Australia
| | - Wee Sim Choo
- a School of Science , Monash University Malaysia , Selangor , Malaysia
| |
Collapse
|
44
|
Intracellular Accumulation of Linezolid and Florfenicol in OptrA-Producing Enterococcus faecalis and Staphylococcus aureus. Molecules 2018; 23:molecules23123195. [PMID: 30518106 PMCID: PMC6320770 DOI: 10.3390/molecules23123195] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 11/30/2018] [Accepted: 12/01/2018] [Indexed: 11/28/2022] Open
Abstract
The optrA gene, which confers transferable resistance to oxazolidinones and phenicols, is defined as an ATP-binding cassette (ABC) transporter but lacks transmembrane domains. The resistance mechanism of optrA and whether it involves antibiotic efflux or ribosomal protection remain unclear. In this study, we determined the MIC values of all bacterial strains by broth microdilution, and used ultra-high performance liquid chromatography-tandem quadrupole mass spectrometry to quantitatively determine the intracellular concentrations of linezolid and florfenicol in Enterococcus faecalis and Staphylococcus aureus. Linezolid and florfenicol both accumulated in susceptible strains and optrA-carrying strains of E. faecalis and S. aureus. No significant differences were observed in the patterns of drug accumulation among E. faecalis JH2-2, E. faecalis JH2-2/pAM401, and E. faecalis JH2-2/pAM401+optrA, but also among S. aureus RN4220, S. aureus RN4220/pAM401, and S. aureus RN4220/pAM401+optrA. ANOVA scores also suggested similar accumulation conditions of the two target compounds in susceptible strains and optrA-carrying strains. Based on our findings, the mechanism of optrA-mediated resistance to oxazolidinones and phenicols obviously does not involve active efflux and the OptrA protein does not confer resistance via efflux like other ABC transporters.
Collapse
|
45
|
Oliveira A, Sousa JC, Silva AC, Melo LDR, Sillankorva S. Chestnut Honey and Bacteriophage Application to Control Pseudomonas aeruginosa and Escherichia coli Biofilms: Evaluation in an ex vivo Wound Model. Front Microbiol 2018; 9:1725. [PMID: 30108574 PMCID: PMC6080586 DOI: 10.3389/fmicb.2018.01725] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 07/11/2018] [Indexed: 12/21/2022] Open
Abstract
Chronic skin wounds represent a major burn both economically and socially. Pseudomonas aeruginosa and Escherichia coli are among the most common colonizers of infected wounds and are prolific biofilm formers. Biofilms are a major problem in infections due to their increasingly difficult control and eradication, and tolerance to multiple prescribed drugs. As so, alternative methods are necessary. Bacteriophages (phages) and honey are both seen as a promising approach for biofilm related infections. Phages have specificity toward a bacterial genus, species or even strain, self-replicating nature, and avoid dysbiosis. Honey has gained acknowledgment due to its antibacterial, antioxidant and anti-inflammatory and wound healing properties. In this work, the effect of E. coli and P. aeruginosa phages vB_EcoS_CEB_EC3a and vB_PaeP_PAO1-D and chestnut honey, alone and combined, were tested using in vitro (polystyrene) and ex vivo (porcine skin) models and against mono and dual-species biofilms of these bacteria. In general, colonization was higher in the porcine skins and the presence of a second microorganism in a consortium of species did not affect the effectiveness of the treatments. The antibacterial effect of combined therapy against dual-species biofilms led to bacterial reductions that were greater for biofilms formed on polystyrene than on skin. Monospecies biofilms of E. coli were better destroyed with phages and honey than P. aeruginosa monospecies biofilms. Overall, the combined phage-honey formulations resulted in higher efficacies possibly due to honey's capacity to damage the bacterial cell membrane and also to its ability to penetrate the biofilm matrix, promoting and enhancing the subsequent phage infection.
Collapse
Affiliation(s)
| | | | | | | | - Sanna Sillankorva
- Centre of Biological Engineering, Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Braga, Portugal
| |
Collapse
|
46
|
Tet38 Efflux Pump Contributes to Fosfomycin Resistance in Staphylococcus aureus. Antimicrob Agents Chemother 2018; 62:AAC.00927-18. [PMID: 29891612 DOI: 10.1128/aac.00927-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 06/06/2018] [Indexed: 11/20/2022] Open
Abstract
Fosfomycin inhibits MurA following uptake by the GlpT transporter of glycerol-3-phosphate in Escherichia coli In Staphylococcus aureus, plasmid overexpression of the Tet38 efflux pump and a glpT mutant resulted in increased MICs and decreased accumulation of fosfomycin, with MICs affected by glycerol-3-phosphate. In contrast, a tet38 mutant had a lower MIC and increased accumulation of fosfomycin, suggesting that Tet38 acts as an efflux transporter of fosfomycin.
Collapse
|
47
|
Identification of a Staphylococcus aureus Efflux Pump Regulator Using a DNA-Protein Affinity Technique. Methods Mol Biol 2018; 1700:269-291. [PMID: 29177836 DOI: 10.1007/978-1-4939-7454-2_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In this chapter, we describe the step-by-step identification of a putative regulator protein and demonstrate the function of this protein as a repressor of the expression of a specific efflux pump, causing resistance to quinolones in Staphylococcus aureus. We show that the knockout gene mutant has an increase in transcript levels of the target efflux pump when compared to that of the S. aureus parental strain RN6390. We provide a detailed protocol that includes the identification of the DNA-binding transcriptional regulatory protein from S. aureus cell extracts using DNA sequences linked to magnetic beads. In addition, we describe the real-time qRT-PCR assays and MIC testing to evaluate the effects of the regulator on S. aureus drug resistance phenotype.
Collapse
|
48
|
Phillips-Jones MK, Harding SE. Antimicrobial resistance (AMR) nanomachines-mechanisms for fluoroquinolone and glycopeptide recognition, efflux and/or deactivation. Biophys Rev 2018; 10:347-362. [PMID: 29525835 PMCID: PMC5899746 DOI: 10.1007/s12551-018-0404-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 02/05/2018] [Indexed: 12/11/2022] Open
Abstract
In this review, we discuss mechanisms of resistance identified in bacterial agents Staphylococcus aureus and the enterococci towards two priority classes of antibiotics-the fluoroquinolones and the glycopeptides. Members of both classes interact with a number of components in the cells of these bacteria, so the cellular targets are also considered. Fluoroquinolone resistance mechanisms include efflux pumps (MepA, NorA, NorB, NorC, MdeA, LmrS or SdrM in S. aureus and EfmA or EfrAB in the enterococci) for removal of fluoroquinolone from the intracellular environment of bacterial cells and/or protection of the gyrase and topoisomerase IV target sites in Enterococcus faecalis by Qnr-like proteins. Expression of efflux systems is regulated by GntR-like (S. aureus NorG), MarR-like (MgrA, MepR) regulators or a two-component signal transduction system (TCS) (S. aureus ArlSR). Resistance to the glycopeptide antibiotic teicoplanin occurs via efflux regulated by the TcaR regulator in S. aureus. Resistance to vancomycin occurs through modification of the D-Ala-D-Ala target in the cell wall peptidoglycan and removal of high affinity precursors, or by target protection via cell wall thickening. Of the six Van resistance types (VanA-E, VanG), the VanA resistance type is considered in this review, including its regulation by the VanSR TCS. We describe the recent application of biophysical approaches such as the hydrodynamic technique of analytical ultracentrifugation and circular dichroism spectroscopy to identify the possible molecular effector of the VanS receptor that activates expression of the Van resistance genes; both approaches demonstrated that vancomycin interacts with VanS, suggesting that vancomycin itself (or vancomycin with an accessory factor) may be an effector of vancomycin resistance. With 16 and 19 proteins or protein complexes involved in fluoroquinolone and glycopeptide resistances, respectively, and the complexities of bacterial sensing mechanisms that trigger and regulate a wide variety of possible resistance mechanisms, we propose that these antimicrobial resistance mechanisms might be considered complex 'nanomachines' that drive survival of bacterial cells in antibiotic environments.
Collapse
Affiliation(s)
- Mary K Phillips-Jones
- National Centre for Macromolecular Hydrodynamics, School of Biosciences, University of Nottingham, Sutton Bonington, LE12 5RD, Loughborough, Leicestershire, UK.
| | - Stephen E Harding
- National Centre for Macromolecular Hydrodynamics, School of Biosciences, University of Nottingham, Sutton Bonington, LE12 5RD, Loughborough, Leicestershire, UK
| |
Collapse
|
49
|
Foster TJ. Antibiotic resistance in Staphylococcus aureus. Current status and future prospects. FEMS Microbiol Rev 2018; 41:430-449. [PMID: 28419231 DOI: 10.1093/femsre/fux007] [Citation(s) in RCA: 412] [Impact Index Per Article: 68.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 02/12/2017] [Indexed: 12/11/2022] Open
Abstract
The major targets for antibiotics in staphylococci are (i) the cell envelope, (ii) the ribosome and (iii) nucleic acids. Several novel targets emerged from recent targeted drug discovery programmes including the ClpP protease and FtsZ from the cell division machinery. Resistance can either develop by horizontal transfer of resistance determinants encoded by mobile genetic elements viz plasmids, transposons and the staphylococcal cassette chromosome or by mutations in chromosomal genes. Horizontally acquired resistance can occur by one of the following mechanisms: (i) enzymatic drug modification and inactivation, (ii) enzymatic modification of the drug binding site, (iii) drug efflux, (iv) bypass mechanisms involving acquisition of a novel drug-resistant target, (v) displacement of the drug to protect the target. Acquisition of resistance by mutation can result from (i) alteration of the drug target that prevents the inhibitor from binding, (ii) derepression of chromosomally encoded multidrug resistance efflux pumps and (iii) multiple stepwise mutations that alter the structure and composition of the cell wall and/or membrane to reduce drug access to its target. This review focuses on development of resistance to currently used antibiotics and examines future prospects for new antibiotics and informed use of drug combinations.
Collapse
|
50
|
Lekshmi M, Ammini P, Adjei J, Sanford LM, Shrestha U, Kumar S, Varela MF. Modulation of antimicrobial efflux pumps of the major facilitator superfamily in Staphylococcus aureus. AIMS Microbiol 2018; 4:1-18. [PMID: 31294201 PMCID: PMC6605029 DOI: 10.3934/microbiol.2018.1.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 12/19/2017] [Indexed: 12/15/2022] Open
Abstract
Variants of the microorganism Staphylococcus aureus which are resistant to antimicrobial agents exist as causative agents of serious infectious disease and constitute a considerable public health concern. One of the main antimicrobial resistance mechanisms harbored by S. aureus pathogens is exemplified by integral membrane transport systems that actively remove antimicrobial agents from bacteria where the cytoplasmic drug targets reside, thus allowing the bacteria to survive and grow. An important class of solute transporter proteins, called the major facilitator superfamily, includes related and homologous passive and secondary active transport systems, many of which are antimicrobial efflux pumps. Transporters of the major facilitator superfamily, which confer antimicrobial efflux and bacterial resistance in S. aureus, are good targets for development of resistance-modifying agents, such as efflux pump inhibition. Such modulatory action upon these antimicrobial efflux systems of the major facilitator superfamily in S. aureus may circumvent resistance and restore the clinical efficacy of therapy towards S. aureus infection.
Collapse
Affiliation(s)
- Manjusha Lekshmi
- QC Laboratory, Harvest and Post Harvest Technology Division, ICAR-Central Institute of Fisheries Education (CIFE), Seven Bungalows, Versova, Andheri (W), Mumbai, 400061, India
| | - Parvathi Ammini
- CSIR-National Institute of Oceanography (NIO), Regional Centre, Dr. Salim Ali Road, Kochi, 682018, India
| | - Jones Adjei
- Eastern New Mexico, Department of Biology, Station 33, 1500 South Avenue K, Portales, NM, 88130, USA
| | - Leslie M Sanford
- Eastern New Mexico, Department of Biology, Station 33, 1500 South Avenue K, Portales, NM, 88130, USA
| | - Ugina Shrestha
- Eastern New Mexico, Department of Biology, Station 33, 1500 South Avenue K, Portales, NM, 88130, USA
| | - Sanath Kumar
- QC Laboratory, Harvest and Post Harvest Technology Division, ICAR-Central Institute of Fisheries Education (CIFE), Seven Bungalows, Versova, Andheri (W), Mumbai, 400061, India
| | - Manuel F Varela
- Eastern New Mexico, Department of Biology, Station 33, 1500 South Avenue K, Portales, NM, 88130, USA
| |
Collapse
|