1
|
Betts HM, Luckett JC, Hill PJ. Pilot Evaluation of S-(3-[ 18F]Fluoropropyl)-D-Homocysteine and O-(2-[ 18F]Fluoroethyl)-D-Tyrosine as Bacteria-Specific Radiotracers for PET Imaging of Infection. Mol Imaging Biol 2024; 26:704-713. [PMID: 38942967 PMCID: PMC11282134 DOI: 10.1007/s11307-024-01929-7] [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: 02/05/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 06/30/2024]
Abstract
PURPOSE There is currently no ideal radiotracer for imaging bacterial infections. Radiolabelled D-amino acids are promising candidates because they are actively incorporated into the peptidoglycan of the bacterial cell wall, a structural feature which is absent in human cells. This work describes fluorine-18 labelled analogues of D-tyrosine and D-methionine, O-(2-[18F]fluoroethyl)-D-tyrosine (D-[18F]FET) and S-(3-[18F]fluoropropyl)-D-homocysteine (D-[18F]FPHCys), and their pilot evaluation studies as potential radiotracers for imaging bacterial infection. PROCEDURES D-[18F]FET and D-[18F]FPHCys were prepared in classical fluorination-deprotection reactions, and their uptake in Staphylococcus aureus and Pseudomonas aeruginosa was evaluated over 2 h. Heat killed bacteria were used as controls. A clinically-relevant foreign body model of S. aureus infection was established in Balb/c mice, as well as a sterile foreign body to mimic inflammation. The ex vivo biodistribution of D-[18F]FPHCys in the infected and inflamed mice was evaluated after 1 h, by dissection and gamma counting. The uptake was compared to that of [18F]FDG. RESULTS In vitro uptake of both D-[18F]FET and D-[18F]FPHCys was specific to live bacteria. Uptake was higher in S. aureus than in P. aeruginosa for both radiotracers, and of the two, higher for D-[18F]FPHCys than D-[18F]FET. Blocking experiments with non-radioactive D-[19F]FPHCys confirmed specificity of uptake. In vivo, D-[18F]FPHCys had greater accumulation in S. aureus infection compared with sterile inflammation, which was statistically significant. As anticipated, [18F]FDG showed no significant difference in uptake between infection and inflammation. CONCLUSIONS D-[18F]FPHCys uptake was higher in infected tissues than inflammation, and represents a fluorine-18 labelled D-AA with potential to detect a S. aureus reference strain (Xen29) in vivo. Additional studies are needed to evaluate uptake of this radiotracer in clinical isolates.
Collapse
Affiliation(s)
- Helen M Betts
- Department of Nuclear Medicine, Nottingham University Hospitals NHS Trust, Queen's Medical Centre, Nottingham, NG7 2UH, UK.
- School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK.
| | - Jeni C Luckett
- School of Life Sciences, University of Nottingham, Biodiscovery Institute, University Park, Nottingham, NG7 2RD, UK
| | - Philip J Hill
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Sutton Bonington, LE17 5RD, UK
| |
Collapse
|
2
|
Liang H, Wang Y, Liu F, Duan G, Long J, Jin Y, Chen S, Yang H. The Application of Rat Models in Staphylococcus aureus Infections. Pathogens 2024; 13:434. [PMID: 38921732 PMCID: PMC11206676 DOI: 10.3390/pathogens13060434] [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: 04/12/2024] [Revised: 05/10/2024] [Accepted: 05/20/2024] [Indexed: 06/27/2024] Open
Abstract
Staphylococcus aureus (S. aureus) is a major human pathogen and can cause a wide range of diseases, including pneumonia, osteomyelitis, skin and soft tissue infections (SSTIs), endocarditis, mastitis, bacteremia, and so forth. Rats have been widely used in the field of infectious diseases due to their unique advantages, and the models of S. aureus infections have played a pivotal role in elucidating their pathogenic mechanisms and the effectiveness of therapeutic agents. This review outlined the current application of rat models in S. aureus infections and future prospects for rat models in infectious diseases caused by S. aureus.
Collapse
Affiliation(s)
- Hongyue Liang
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China; (H.L.); (F.L.); (G.D.); (J.L.); (Y.J.); (S.C.)
| | - Yadong Wang
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou 450016, China;
| | - Fang Liu
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China; (H.L.); (F.L.); (G.D.); (J.L.); (Y.J.); (S.C.)
| | - Guangcai Duan
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China; (H.L.); (F.L.); (G.D.); (J.L.); (Y.J.); (S.C.)
| | - Jinzhao Long
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China; (H.L.); (F.L.); (G.D.); (J.L.); (Y.J.); (S.C.)
| | - Yuefei Jin
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China; (H.L.); (F.L.); (G.D.); (J.L.); (Y.J.); (S.C.)
| | - Shuaiyin Chen
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China; (H.L.); (F.L.); (G.D.); (J.L.); (Y.J.); (S.C.)
| | - Haiyan Yang
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China; (H.L.); (F.L.); (G.D.); (J.L.); (Y.J.); (S.C.)
| |
Collapse
|
3
|
Wang Y, Li C, Zhang H, Chi Y, Cai Y. The Potentiation Activity of Azithromycin in Combination with Colistin or Levofloxacin Against Pseudomonas aeruginosa Biofilm Infection. Infect Drug Resist 2024; 17:1259-1266. [PMID: 38562404 PMCID: PMC10984211 DOI: 10.2147/idr.s438576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 03/08/2024] [Indexed: 04/04/2024] Open
Abstract
Objective Pseudomonas aeruginosa (PA) often displays drug resistance and biofilm-mediated adaptability. Here, we aimed to evaluate the antibiofilm efficacy of azithromycin-based combination regimens. Methods Minimum inhibitory concentrations (MICs), minimal biofilm eradication concentrations (MBECs), and MBEC-combination of azithromycin, colistin, amikacin, and levofloxacin to bioluminescent strain PAO1 and carbapenem-resistant PAO1 (CRPAO1) were assessed. An animal biofilm infection model was established and detected using a live animal bio-photonic imaging system. Results In vitro, PAO1 and CRPAO1 were susceptible to colistin, amikacin, and levofloxacin, while they were unsusceptible to azithromycin. The combinations based on azithromycin have no synergistic effect on biofilm in vitro. In vivo, azithromycin plus colistin or levofloxacin could shorten the PAO1 biofilm eradication time, which totally eradicates the biofilm in all mice on the 8th or 6th day, while monotherapy only eradicate biofilm in 70% or 80% mice on the 8th day. For CRPAO1 biofilm, only azithromycin-colistin combination and colistin monotherapy eradicated the bacteria in 60% and 40% of mice at the 6th day. Conclusion Azithromycin-based combinations containing levofloxacin or colistin had no synergistic effect in vitro, and they are promising for clinical applications due to the good synergistic activity against PAO1 biofilms in vivo.
Collapse
Affiliation(s)
- Yuhang Wang
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of PLA General Hospital, Beijing, People’s Republic of China
| | - Chunsun Li
- Laboratory of Department of Pulmonary and Critical Care Medicine, PLA General Hospital, Beijing, People’s Republic of China
| | - Huan Zhang
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of PLA General Hospital, Beijing, People’s Republic of China
| | - Yulong Chi
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of PLA General Hospital, Beijing, People’s Republic of China
| | - Yun Cai
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of PLA General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
4
|
Lacey KA, Serpas L, Makita S, Wang Y, Rashidfarrokhi A, Soni C, Gonzalez S, Moreira A, Torres VJ, Reizis B. Secreted mammalian DNases protect against systemic bacterial infection by digesting biofilms. J Exp Med 2023; 220:e20221086. [PMID: 36928522 PMCID: PMC10037111 DOI: 10.1084/jem.20221086] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 01/18/2023] [Accepted: 03/01/2023] [Indexed: 03/18/2023] Open
Abstract
Extracellular DNase DNASE1L3 maintains tolerance to self-DNA in humans and mice, whereas the role of its homolog DNASE1 remains controversial, and the overall function of secreted DNases in immunity is unclear. We report that deletion of murine DNASE1 neither caused autoreactivity in isolation nor exacerbated lupus-like disease in DNASE1L3-deficient mice. However, combined deficiency of DNASE1 and DNASE1L3 rendered mice susceptible to bloodstream infection with Staphylococcus aureus. DNASE1/DNASE1L3 double-deficient mice mounted a normal innate response to S. aureus and did not accumulate neutrophil extracellular traps (NETs). However, their kidneys manifested severe pathology, increased bacterial burden, and biofilm-like bacterial lesions that contained bacterial DNA and excluded neutrophils. Furthermore, systemic administration of recombinant DNASE1 protein during S. aureus infection rescued the mortality of DNase-deficient mice and ameliorated the disease in wild-type mice. Thus, DNASE1 and DNASE1L3 jointly facilitate the control of bacterial infection by digesting extracellular microbial DNA in biofilms, suggesting the original evolutionary function of secreted DNases as antimicrobial agents.
Collapse
Affiliation(s)
- Keenan A. Lacey
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Lee Serpas
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Sohei Makita
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Yueyang Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Ali Rashidfarrokhi
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Chetna Soni
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Sandra Gonzalez
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Andre Moreira
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY, USA
| | - Boris Reizis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
5
|
Complete Genome Sequences of Bioluminescent Staphylococcus aureus Strains Xen31 and Xen36, Derived from Two Clinical Isolates. Microbiol Resour Announc 2023; 12:e0002423. [PMID: 36840571 PMCID: PMC10019319 DOI: 10.1128/mra.00024-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023] Open
Abstract
Here, we report complete genome sequences of two clinical isolates of Staphylococcus aureus, namely, Xen31 and Xen36, which have been genetically modified to express an optimized Photorhabdus luminescens luciferase operon. Xen31 and Xen36 are bioluminescent strains used widely for investigation of bacterial pathogenesis, drug discovery, and development of novel therapies.
Collapse
|
6
|
Yi L, Fan Q, Wang H, Fan H, Zuo J, Wang Y, Wang Y. Establishment of Streptococcus suis Biofilm Infection Model In Vivo and Comparative Analysis of Gene Expression Profiles between In Vivo and In Vitro Biofilms. Microbiol Spectr 2023; 11:e0268622. [PMID: 36507687 PMCID: PMC9927446 DOI: 10.1128/spectrum.02686-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
Streptococcus suis is a zoonotic pathogen that continuously threatens animal husbandry and public health worldwide. Studies have shown that S. suis can cause persistent infection by forming biofilms. In this study, a model of S. suis biofilm-related infection was successfully constructed for the first time by simulating the natural infection of S. suis, and biofilm of S. suis in vivo was successfully observed in the lung tissue of infected pigs by a variety of detection methods. Subsequently, selective capture of transcribed sequences (SCOTS) was used to identify genes expressed by S. suis in vivo biofilms. Sixty-nine genes were captured in in vivo biofilms formed by S. suis for the first time by SCOTS; they were mainly involved in metabolism, cell replication, and division, transport, signal transduction, cell wall, etc. Genes related to S. suis in vitro biofilm formation were also identified by SCOTS and RNA sequencing. Approximately half of the genes captured by SCOTS in the in vivo and in vitro biofilms were found to be different. In summary, our study provides powerful clues for future exploration of the mechanisms of S. suis biofilm formation. IMPORTANCE Streptococcus suis is considered an important zoonotic pathogen, and persistent infection caused by biofilm is currently considered to be the reason why S. suis is difficult to control in swine. However, to date, a model of the biofilm of S. suis in vivo has not been successfully constructed. Here, we successfully detected biofilms of S. suis in vivo in lung tissues of piglets infected with S. suis. Selective capture of transcribed sequences and the transcriptome were used to obtain gene profiles of S. suis in vivo and in vitro biofilms, and the results showed large differences between them. Such data are of importance for future experimental studies exploring the mechanism of biofilm formation by S. suis in vivo.
Collapse
Affiliation(s)
- Li Yi
- College of Life Science, Luoyang Normal University, Luoyang, China
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Qingying Fan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, China
| | - Haikun Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, China
| | - Haoran Fan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, China
| | - Jing Zuo
- College of Life Science, Luoyang Normal University, Luoyang, China
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Yuxin Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, China
| | - Yang Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, China
| |
Collapse
|
7
|
Wang Y, Li C, Wang J, Bai N, Zhang H, Chi Y, Cai Y. The Efficacy of Colistin Combined with Amikacin or Levofloxacin against Pseudomonas aeruginosa Biofilm Infection. Microbiol Spectr 2022; 10:e0146822. [PMID: 36102678 PMCID: PMC9603716 DOI: 10.1128/spectrum.01468-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/18/2022] [Indexed: 12/31/2022] Open
Abstract
Pseudomonas aeruginosa (PA) biofilm infection is clinically prevalent and difficult to eradicate. In the present work, we aimed to evaluate the in vitro and in vivo efficacy of colistin (COL)-based combinations against PA biofilm. MICs and fractional inhibitory concentration indexes (FICIs) of four antibiotics (COL, amikacin, levofloxacin, and meropenem) to bioluminescent strain PAO1, carbapenem-resistant PAO1 (CRPAO1), and clinically isolated strains were assessed. Minimal biofilm eradication concentrations (MBECs) of monotherapy and combinations were examined by counting the live bacteria in biofilm, accompanied by visual confirmation using confocal laser-scanning microscopy. An animal biofilm infection model was established by implanting biofilm subcutaneously, and the therapeutic effect was evaluated according to the change in luminescence through a live animal bio-photonic imaging system. In vitro, even combined with 4 or 8 mg/L COL, meropenem needed to reach 128 or 256 mg/L to eradicate the biofilm. Moreover, 2 mg/L COL combined with 32 mg/L amikacin or 4-8 mg/L levofloxacin could kill the PAO1 and CRPAO1 in biofilm within 24 h. In vivo, COL combined with amikacin or levofloxacin could shorten the eradication time of biofilm than monotherapy. For PAO1 biofilm, combination therapy could eradicate the biofilm in all mice on the 5th day, whereas monotherapy only eradicated biofilms in almost half of the mice. For CRPAO1 biofilm, the biofilm eradication rate on the 6th day in the COL+ amikacin, amikacin, or COL alone regimen was 90%, 10%, or 40%, respectively. COL combined with levofloxacin did not show a better effect than each individual antibiotic. COL-based combinations containing levofloxacin or amikacin were promising choices for treating PA biofilm infection. IMPORTANCE Infections associated with PA biofilm formation are extremely challenging. When monotherapy fails to achieve optimal efficacy, combination therapy becomes the last option. After evaluating multiple drug combinations through a series of experiments in vitro and in vivo, we confirmed that colistin-based combinations containing levofloxacin or amikacin were promising choices for treating PA biofilm infection. The efficacy of these combinations derives from the different bactericidal mechanisms and the bacterial susceptibility to each antibiotic. This study provided a new regimen to solve the incurable problem of biofilm by using COL combined with other antibiotics.
Collapse
Affiliation(s)
- Yuhang Wang
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center, PLA General Hospital, Beijing, People’s Republic of China
| | - Chunsun Li
- Laboratory of Department of Pulmonary and Critical Care Medicine, PLA General Hospital, Beijing, People’s Republic of China
| | - Jin Wang
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center, PLA General Hospital, Beijing, People’s Republic of China
| | - Nan Bai
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center, PLA General Hospital, Beijing, People’s Republic of China
| | - Huan Zhang
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center, PLA General Hospital, Beijing, People’s Republic of China
| | - Yulong Chi
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center, PLA General Hospital, Beijing, People’s Republic of China
| | - Yun Cai
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center, PLA General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
8
|
Kouijzer JJP, Noordermeer DJ, van Leeuwen WJ, Verkaik NJ, Lattwein KR. Native valve, prosthetic valve, and cardiac device-related infective endocarditis: A review and update on current innovative diagnostic and therapeutic strategies. Front Cell Dev Biol 2022; 10:995508. [PMID: 36263017 PMCID: PMC9574252 DOI: 10.3389/fcell.2022.995508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Infective endocarditis (IE) is a life-threatening microbial infection of native and prosthetic heart valves, endocardial surface, and/or indwelling cardiac device. Prevalence of IE is increasing and mortality has not significantly improved despite technological advances. This review provides an updated overview using recent literature on the clinical presentation, diagnosis, imaging, causative pathogens, treatment, and outcomes in native valve, prosthetic valve, and cardiac device-related IE. In addition, the experimental approaches used in IE research to improve the understanding of disease mechanisms and the current diagnostic pipelines are discussed, as well as potential innovative diagnostic and therapeutic strategies. This will ultimately help towards deriving better diagnostic tools and treatments to improve IE patient outcomes.
Collapse
Affiliation(s)
- Joop J. P. Kouijzer
- Thoraxcenter, Department of Biomedical Engineering, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Daniëlle J. Noordermeer
- Thoraxcenter, Department of Biomedical Engineering, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Wouter J. van Leeuwen
- Department of Cardiothoracic Surgery, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Nelianne J. Verkaik
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Kirby R. Lattwein
- Thoraxcenter, Department of Biomedical Engineering, Erasmus MC University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
9
|
Tang C, Kurata S, Fuse N. Genetic dissection of innate immune memory in Drosophila melanogaster. Front Immunol 2022; 13:857707. [PMID: 35990631 PMCID: PMC9386478 DOI: 10.3389/fimmu.2022.857707] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Current studies have demonstrated that innate immunity possesses memory characteristics. Although the molecular mechanisms underlying innate immune memory have been addressed by numerous studies, genetic variations in innate immune memory and the associated genes remain unclear. Here, we explored innate immune memory in 163 lines of Drosophila melanogaster from the Drosophila Synthetic Population Resource. In our assay system, prior training with low pathogenic bacteria (Micrococcus luteus) increased the survival rate of flies after subsequent challenge with highly pathogenic bacteria (Staphylococcus aureus). This positive training effect was observed in most lines, but some lines exhibited negative training effects. Survival rates under training and control conditions were poorly correlated, suggesting that distinct genetic factors regulate training effects and normal immune responses. Subsequent quantitative trait loci analysis suggested that four loci containing 80 genes may be involved in regulating innate immune memory. Among them, Adgf-A, which encodes an extracellular adenosine deaminase-related growth factor, was shown to be associated with training effects. Our study findings help to elucidate the genetic architecture of innate immune memory in Drosophila and may provide insight for new therapeutic treatments aimed at boosting immunity.
Collapse
Affiliation(s)
| | | | - Naoyuki Fuse
- *Correspondence: Shoichiro Kurata, ; Naoyuki Fuse,
| |
Collapse
|
10
|
Zank A, Schulte L, Brandon X, Carstensen L, Wescott A, Schwan WR. Mutations of the brpR and brpS genes affect biofilm formation in Staphylococcus aureus. World J Clin Infect Dis 2022; 12:20-32. [DOI: 10.5495/wjcid.v12.i1.20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/03/2021] [Accepted: 02/13/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND In the United States, Staphylococcus aureus (S. aureus) kills tens of thousands of individuals each year and the formation of a biofilm contributes to lethality. Biofilm-associated infections are hard to treat once the biofilm has formed. A new stilbene drug, labeled SK-03-92, was shown to kill S. aureus and affected transcription of two genes tied to a putative two-component system (TCS) we have named brpR (biofilm regulating protein regulator) and brpS (biofilm regulating protein sensor).
AIM To determine if BrpR and BrpS regulate biofilm formation, brpR and brpS mutants were assessed using biofilm assays compared to wild-type S. aureus.
METHODS A combination of biofilm and quantitative real-time-polymerase chain reaction assays were used. In addition, bioinformatic software tools were also utilized.
RESULTS Significantly more biofilm was created in the brpR and brpS mutants vs wild-type cells. Quantitative real-time polymerase chain reactions showed the brpS mutant had differences in transcription of biofilm associated genes that were eight-fold higher for srtA, two-fold lower for lrgA, and 1.6-fold higher for cidA compared to wild-type. Bioinformatic analysis demonstrated that the S. aureus brpR/brpS TCS had homology to streptococcal late-stage competence proteins involved in cell-death, increased biofilm production, and the development of persister cells.
CONCLUSION Our study suggests that brpR/brpS is a TCS that may repress S. aureus biofilm production and be linked to late-stage competence in S. aureus.
Collapse
Affiliation(s)
- Allison Zank
- Department of Microbiology, University of Wisconsin-La Crosse, La Crosse, WI 54601, United States
| | - Lillian Schulte
- Department of Microbiology, University of Wisconsin-La Crosse, La Crosse, WI 54601, United States
| | - Xavier Brandon
- Department of Microbiology, University of Wisconsin-La Crosse, La Crosse, WI 54601, United States
| | - Lauren Carstensen
- Department of Microbiology, University of Wisconsin-La Crosse, La Crosse, WI 54601, United States
| | - Amy Wescott
- Department of Microbiology, University of Wisconsin-La Crosse, La Crosse, WI 54601, United States
| | - William R Schwan
- Department of Microbiology, University of Wisconsin-La Crosse, La Crosse, WI 54601, United States
| |
Collapse
|
11
|
Koczerka M, Lantier I, Pinard A, Morillon M, Deperne J, Gal-Mor O, Grépinet O, Virlogeux-Payant I. In Vivo Tracking of Bacterial Colonization in Different Murine Models Using Bioluminescence: The Example of Salmonella. Methods Mol Biol 2022; 2427:235-248. [PMID: 35619038 DOI: 10.1007/978-1-0716-1971-1_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Applications of bioluminescence for the in vivo study of pathogenic microorganisms are numerous, ranging from the quantification of virulence gene expression to measuring the effect of antimicrobial molecules on the colonization of tissues and organs by the pathogen. Most studies are performed in mice, but recent works demonstrate that this technique is applicable to larger animals like fish, guinea pigs, ferrets, and chickens. Here, we describe the construction and the utilization of a constitutively luminescent strain of Salmonella Typhimurium to monitor in vivo and ex vivo the colonization of mice in the gastroenteritis, typhoid fever, and asymptomatic carriage models of Salmonella infection.
Collapse
Affiliation(s)
| | | | | | | | | | - Ohad Gal-Mor
- The Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer, Israel
- The Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | |
Collapse
|
12
|
Sanguiin H-6 Fractionated from Cloudberry ( Rubus chamaemorus) Seeds Can Prevent the Methicillin-Resistant Staphylococcus aureus Biofilm Development during Wound Infection. Antibiotics (Basel) 2021; 10:antibiotics10121481. [PMID: 34943693 PMCID: PMC8698471 DOI: 10.3390/antibiotics10121481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/25/2021] [Accepted: 11/29/2021] [Indexed: 11/16/2022] Open
Abstract
Staphylococcus aureus is the most common cause of surgical site infections and its treatment is challenging due to the emergence of multi-drug resistant strains such as methicillin-resistant S. aureus (MRSA). Natural berry-derived compounds have shown antimicrobial potential, e.g., ellagitannins such as sanguiin H-6 and lambertianin C, the main phenolic compounds in Rubus seeds, have shown antimicrobial activity. The aim of this study was to evaluate the effect of sanguiin H-6 and lambertianin C fractionated from cloudberry seeds, on the MRSA growth, and as treatment of a MRSA biofilm development in different growth media in vitro and in vivo by using a murine wound infection model where sanguiin H-6 and lambertianin C were used to prevent the MRSA infection. Sanguiin H-6 and lambertianin C inhibited the in vitro biofilm development and growth of MRSA. Furthermore, sanguiin H-6 showed significant anti-MRSA effect in the in vivo wound model. Our study shows the possible use of sanguiin H-6 as a preventive measure in surgical sites to avoid postoperative infections, whilst lambertianin C showed no anti-MRSA activity.
Collapse
|
13
|
Lerche CJ, Schwartz F, Theut M, Fosbøl EL, Iversen K, Bundgaard H, Høiby N, Moser C. Anti-biofilm Approach in Infective Endocarditis Exposes New Treatment Strategies for Improved Outcome. Front Cell Dev Biol 2021; 9:643335. [PMID: 34222225 PMCID: PMC8249808 DOI: 10.3389/fcell.2021.643335] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 05/04/2021] [Indexed: 12/13/2022] Open
Abstract
Infective endocarditis (IE) is a life-threatening infective disease with increasing incidence worldwide. From early on, in the antibiotic era, it was recognized that high-dose and long-term antibiotic therapy was correlated to improved outcome. In addition, for several of the common microbial IE etiologies, the use of combination antibiotic therapy further improves outcome. IE vegetations on affected heart valves from patients and experimental animal models resemble biofilm infections. Besides the recalcitrant nature of IE, the microorganisms often present in an aggregated form, and gradients of bacterial activity in the vegetations can be observed. Even after appropriate antibiotic therapy, such microbial formations can often be identified in surgically removed, infected heart valves. Therefore, persistent or recurrent cases of IE, after apparent initial infection control, can be related to biofilm formation in the heart valve vegetations. On this background, the present review will describe potentially novel non-antibiotic, antimicrobial approaches in IE, with special focus on anti-thrombotic strategies and hyperbaric oxygen therapy targeting the biofilm formation of the infected heart valves caused by Staphylococcus aureus. The format is translational from preclinical models to actual clinical treatment strategies.
Collapse
Affiliation(s)
- Christian Johann Lerche
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Franziska Schwartz
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Marie Theut
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Emil Loldrup Fosbøl
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kasper Iversen
- Department of Cardiology, Herlev and Gentofte Hospital, University of Copenhagen, Herlev, Denmark
- Department of Emergency Medicine, Herlev and Gentofte Hospital, University of Copenhagen, Herlev, Denmark
| | - Henning Bundgaard
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Niels Høiby
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Claus Moser
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
14
|
Qiu B, Cobb J, Loiselle AE, Ketonis C. Development of a Murine Model of Pyogenic Flexor Tenosynovitis. J Bone Joint Surg Am 2021; 103:432-438. [PMID: 33411464 DOI: 10.2106/jbjs.20.00428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Pyogenic flexor tenosynovitis is a debilitating infection of the hand flexor tendon sheath with high morbidity despite standard treatments of empiric antibiotics with irrigation and debridement. In vivo studies in the available literature have used avian models, but these models are difficult to scale and maintain. The purpose of this study was to demonstrate the plausibility of a murine model of pyogenic flexor tenosynovitis utilizing bioluminescence imaging and tissue analysis at harvest. METHODS A 2-μL inoculate of bioluminescent Xen29 Staphylococcus aureus or sterile phosphate-buffered saline solution (sPBS) was administered to the tendon sheath of 36 male C57BL/6J mice. The infectious course was monitored by bioluminescence imaging (BLI) via an in vivo imaging system, gross anatomic deformity, and weight change. The infected hind paws were harvested at 4 time points: 24 hours, 72 hours, 1 week, and 2 weeks for histological analysis using Alcian blue, hematoxylin, and Orange-G staining. Two-way analysis of variance with the Sidak multiple comparison test was used to assess differences in bioluminescence and weight at each time point. RESULTS The infected cohort displayed significantly elevated bioluminescence values, had reductions in weight, and exhibited swelling of the infected digit throughout the course of infection. By day 4, most infected mice saw a substantial decrease in BLI signal intensity; however, 2 infected mice exhibited persistent BLI intensity through day 14. Histological analysis of the infected cohort showed tissue disorganization and the presence of a cellular infiltrate in and around the flexor tendon sheath. CONCLUSIONS A murine model of pyogenic flexor tenosynovitis is possible and can serve as an experimental platform for further investigation of the pathophysiology of pyogenic flexor tenosynovitis. CLINICAL RELEVANCE This animal model can be utilized in elucidating the basic molecular and/or cellular mechanisms of pyogenic flexor tenosynovitis while simultaneously evaluating novel therapeutic strategies.
Collapse
Affiliation(s)
- Bowen Qiu
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, New York
| | | | | | | |
Collapse
|
15
|
Ryan LK, Hise AG, Hossain CM, Ruddick W, Parveen R, Freeman KB, Weaver DG, Narra HP, Scott RW, Diamond G. A Novel Immunocompetent Mouse Model for Testing Antifungal Drugs Against Invasive Candida albicans Infection. J Fungi (Basel) 2020; 6:E197. [PMID: 33007818 PMCID: PMC7712810 DOI: 10.3390/jof6040197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/22/2020] [Accepted: 09/28/2020] [Indexed: 12/12/2022] Open
Abstract
Disseminated infection by Candida species represents a common, often life-threatening condition. Increased resistance to current antifungal drugs has led to an urgent need to develop new antifungal drugs to treat this pathogen. However, in vivo screening of candidate antifungal compounds requires large numbers of animals and using immunosuppressive agents to allow for fungal dissemination. To increase the efficiency of screening, to use fewer mice, and to remove the need for immunosuppressive agents, which may interfere with the drug candidates, we tested the potential for a novel approach using in vivo imaging of a fluorescent strain of Candida albicans, in a mouse strain deficient in the host defense peptide, murine β-defensin 1 (mBD-1). We developed a strain of C. albicans that expresses red fluorescent protein (RFP), which exhibits similar infectivity to the non-fluorescent parent strain. When this strain was injected into immunocompetent mBD-1-deficient mice, we observed a non-lethal disseminated infection. Further, we could quantify its dissemination in real time, and observe the activity of an antifungal peptide mimetic drug by in vivo imaging. This novel method will allow for the rapid in vivo screening of antifungal drugs, using fewer mice, and increase the efficiency of testing new antifungal agents.
Collapse
Affiliation(s)
- Lisa K. Ryan
- Division of Infectious Disease and Global Medicine, Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA;
| | - Amy G Hise
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA;
- Medicine Service, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| | - Chowdhury Mobaswar Hossain
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610, USA; (C.M.H.); (W.R.); (R.P.)
| | - William Ruddick
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610, USA; (C.M.H.); (W.R.); (R.P.)
| | - Rezwana Parveen
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610, USA; (C.M.H.); (W.R.); (R.P.)
| | - Katie B. Freeman
- Fox Chase Chemical Diversity Center, Inc., Pennsylvania Biotechnology Center, Doylestown, PA 18902, USA; (K.B.F.); (D.G.W.); (R.W.S.)
| | - Damian G. Weaver
- Fox Chase Chemical Diversity Center, Inc., Pennsylvania Biotechnology Center, Doylestown, PA 18902, USA; (K.B.F.); (D.G.W.); (R.W.S.)
| | - Hema P. Narra
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - Richard W. Scott
- Fox Chase Chemical Diversity Center, Inc., Pennsylvania Biotechnology Center, Doylestown, PA 18902, USA; (K.B.F.); (D.G.W.); (R.W.S.)
| | - Gill Diamond
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610, USA; (C.M.H.); (W.R.); (R.P.)
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY 40902, USA
| |
Collapse
|
16
|
Li Y, Daryaee F, Yoon GE, Noh D, Smith-Jones PM, Si Y, Walker SG, Turkman N, Meimetis L, Tonge PJ. Positron Emission Tomography Imaging of Staphylococcus aureus Infection Using a Nitro-Prodrug Analogue of 2-[ 18F]F- p-Aminobenzoic Acid. ACS Infect Dis 2020; 6:2249-2259. [PMID: 32672928 DOI: 10.1021/acsinfecdis.0c00374] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Deep-seated bacterial infections caused by pathogens such as Staphylococcus aureus are difficult to diagnose and treat and are thus a major threat to human health. In previous work we demonstrated that positron emission tomography (PET) imaging with 2-[18F]F-p-aminobenzoic acid (2-[18F]F-PABA) could noninvasively identify, localize, and monitor S. aureus infection with excellent sensitivity and specificity in a rodent soft tissue infection model. However, 2-[18F]F-PABA is rapidly N-acetylated and eliminated, and in an attempt to improve radiotracer accumulation in bacteria we adopted a prodrug strategy in which the acid was protected by an ester and the amine was replaced with a nitro group. Metabolite analysis indicated that the nitro group of ethyl 2-[18F]fluoro-4-nitrobenzoate (2-[18F]F-ENB) is converted to the corresponding amine by bacteria-specific nitroreductases while the ester is hydrolyzed in vivo into the acid. PET/CT imaging of 2-[18F]F-ENB and the corresponding acid 2-[18F]F-NB in a rat soft tissue infection model demonstrated colocalization of the radiotracer with the bioluminescent signal arising from S. aureus Xen29, and demonstrated that the tracer could differentiate S. aureus infection from sterile inflammation. Significantly, the accumulation of both 2-[18F]F-ENB and 2-[18F]F-NB at the site of infection was 17-fold higher than at the site of sterile inflammation compared to 8-fold difference observed for 2-[18F]F-PABA, supporting the proposal that the active radiotracer in vivo is 2-[18F]F-NB. Collectively, these data suggest that 2-[18F]F-ENB and 2-[18F]F-NB have the potential for translation to humans as a rapid, noninvasive diagnostic tool to identify and localize S. aureus infections.
Collapse
Affiliation(s)
- Yong Li
- Chronus Pharmaceuticals, 25 Health Sciences Drive, Stony Brook, New York 11790, United States
| | - Fereidoon Daryaee
- Chronus Pharmaceuticals, 25 Health Sciences Drive, Stony Brook, New York 11790, United States
| | - Grace E. Yoon
- The Facility for Experimental Radiopharmaceutical Manufacturing, Department of Psychiatry, Stony Brook University, Stony Brook, New York 11794, United States
| | - Doyoung Noh
- The Facility for Experimental Radiopharmaceutical Manufacturing, Department of Psychiatry, Stony Brook University, Stony Brook, New York 11794, United States
| | - Peter M. Smith-Jones
- The Facility for Experimental Radiopharmaceutical Manufacturing, Department of Psychiatry, Stony Brook University, Stony Brook, New York 11794, United States
| | | | - Stephen G. Walker
- Department of Oral Biology and Pathology, Stony Brook University, Stony Brook, New York 11794, United States
| | | | - Labros Meimetis
- Chronus Pharmaceuticals, 25 Health Sciences Drive, Stony Brook, New York 11790, United States
| | - Peter J. Tonge
- Chronus Pharmaceuticals, 25 Health Sciences Drive, Stony Brook, New York 11790, United States
| |
Collapse
|
17
|
Castillo Almeida NE, Gurram P, Esquer Garrigos Z, Mahmood M, Baddour LM, Sohail MR. Diagnostic imaging in infective endocarditis: a contemporary perspective. Expert Rev Anti Infect Ther 2020; 18:911-925. [PMID: 32442039 DOI: 10.1080/14787210.2020.1773260] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Infective endocarditis (IE) remains a diagnostic challenge. Prompt diagnosis is essential for accurate risk stratification and appropriate therapeutic decisions and surgical management. In recent years, the use of multimodal imaging has had a transformative effect on the diagnostic approach of IE in selected patients. AREAS COVERED This review assesses published literature on different imaging modalities for the diagnosis of IE published between 1 January 2009 and 1 February 2020. We illustrate the diagnostic approach to IE with three clinical cases. EXPERT OPINION Novel approaches to imaging for cardiac and extracardiac complications improve and individualize diagnosis, management, and prognosis in patients with suspected IE. The use of multimodal imaging should be guided by a multidisciplinary group of medical providers that includes infectious disease specialists, radiologists, cardiologists, and cardiothoracic surgeons.
Collapse
Affiliation(s)
- Natalia E Castillo Almeida
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic College of Medicine and Science , Rochester, MN, USA
| | - Pooja Gurram
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic College of Medicine and Science , Rochester, MN, USA
| | - Zerelda Esquer Garrigos
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic College of Medicine and Science , Rochester, MN, USA
| | - Maryam Mahmood
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic College of Medicine and Science , Rochester, MN, USA
| | - Larry M Baddour
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic College of Medicine and Science , Rochester, MN, USA.,Department of Cardiovascular Diseases, Mayo Clinic College of Medicine and Science , Rochester, MN, USA
| | - M Rizwan Sohail
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic College of Medicine and Science , Rochester, MN, USA.,Department of Cardiovascular Diseases, Mayo Clinic College of Medicine and Science , Rochester, MN, USA
| |
Collapse
|
18
|
Miller RJ, Crosby HA, Schilcher K, Wang Y, Ortines RV, Mazhar M, Dikeman DA, Pinsker BL, Brown ID, Joyce DP, Zhang J, Archer NK, Liu H, Alphonse MP, Czupryna J, Anderson WR, Bernthal NM, Fortuno-Miranda L, Bulte JWM, Francis KP, Horswill AR, Miller LS. Development of a Staphylococcus aureus reporter strain with click beetle red luciferase for enhanced in vivo imaging of experimental bacteremia and mixed infections. Sci Rep 2019; 9:16663. [PMID: 31723175 PMCID: PMC6853927 DOI: 10.1038/s41598-019-52982-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/26/2019] [Indexed: 02/03/2023] Open
Abstract
In vivo bioluminescence imaging has been used to monitor Staphylococcus aureus infections in preclinical models by employing bacterial reporter strains possessing a modified lux operon from Photorhabdus luminescens. However, the relatively short emission wavelength of lux (peak 490 nm) has limited tissue penetration. To overcome this limitation, the gene for the click beetle (Pyrophorus plagiophtalamus) red luciferase (luc) (with a longer >600 emission wavelength), was introduced singly and in combination with the lux operon into a methicillin-resistant S. aureus strain. After administration of the substrate D-luciferin, the luc bioluminescent signal was substantially greater than the lux signal in vitro. The luc signal had enhanced tissue penetration and improved anatomical co-registration with infected internal organs compared with the lux signal in a mouse model of S. aureus bacteremia with a sensitivity of approximately 3 × 104 CFU from the kidneys. Finally, in an in vivo mixed bacterial wound infection mouse model, S. aureus luc signals could be spectrally unmixed from Pseudomonas aeruginosa lux signals to noninvasively monitor the bacterial burden of both strains. Therefore, the S. aureus luc reporter may provide a technological advance for monitoring invasive organ dissemination during S. aureus bacteremia and for studying bacterial dynamics during mixed infections.
Collapse
Affiliation(s)
- Robert J Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Heidi A Crosby
- Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045, USA
| | - Katrin Schilcher
- Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045, USA
| | - Yu Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Roger V Ortines
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Momina Mazhar
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dustin A Dikeman
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Bret L Pinsker
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Isabelle D Brown
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniel P Joyce
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeffrey Zhang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nathan K Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Haiyun Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Martin P Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | - Nicholas M Bernthal
- Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, Santa Monica, California, USA
| | - Lea Fortuno-Miranda
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Jeff W M Bulte
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA.,Department of Chemical & Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, Maryland, 21205, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Kevin P Francis
- PerkinElmer, Hopkinton, Massachusetts, USA.,Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, Santa Monica, California, USA
| | - Alexander R Horswill
- Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045, USA.,Denver VA Healthcare System, Denver, Colorado, USA
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA. .,Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA. .,Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA. .,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, 21218, USA.
| |
Collapse
|
19
|
Pratibha, Shukla M, Kaul G, Chopra S, Verma S. Nucleobase Soft Metallogel Composites with Antifouling Activities against ESKAPE Pathogens. ChemistrySelect 2019. [DOI: 10.1002/slct.201803693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Pratibha
- Department of ChemistryCenter for Environmental Science and EngineeringIndian Institute of Technology Kanpur Kanpur 208016 (UP) India
| | - Manjulika Shukla
- Division of MicrobiologyCSIR-Central Drug Research Institute, Sector 10, Janakipuram Extension Sitapur Road Lucknow 226031 India
| | - Grace Kaul
- Division of MicrobiologyCSIR-Central Drug Research Institute, Sector 10, Janakipuram Extension Sitapur Road Lucknow 226031 India
| | - Sidharth Chopra
- Division of MicrobiologyCSIR-Central Drug Research Institute, Sector 10, Janakipuram Extension Sitapur Road Lucknow 226031 India
| | - Sandeep Verma
- Department of ChemistryCenter for Environmental Science and EngineeringIndian Institute of Technology Kanpur Kanpur 208016 (UP) India
- DST Thematic Unit of Excellence on Soft NanofabricationIndian Institute of Technology Kanpur, Kanpur Uttar Pradesh 208016 India
| |
Collapse
|
20
|
Adjunctive Clavulanic Acid Abolishes the Cefazolin Inoculum Effect in an Experimental Rat Model of Methicillin-Sensitive Staphylococcus aureus Endocarditis. Antimicrob Agents Chemother 2018; 62:AAC.01158-18. [PMID: 30150459 DOI: 10.1128/aac.01158-18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/17/2018] [Indexed: 01/04/2023] Open
Abstract
We tested the ability of clavulanic acid to restore the efficacy of cefazolin against Staphylococcus aureus TX0117, which exhibits the cefazolin inoculum effect (CzIE). In the rat infective endocarditis model, the coadministration of cefazolin plus clavulanic acid resulted in a significant reduction of bacterial counts (7.1 ± 0.5 log10 CFU/g) compared to that with cefazolin alone (2 ± 0.6 log10 CFU/g; P < 0.0001). The addition of a β-lactamase inhibitor may be a viable strategy for overcoming the CzIE.
Collapse
|
21
|
Harraghy N, Seiler S, Jacobs K, Hannig M, Menger MD, Herrmann M. Advances in in Vitro and in Vivo Models for Studying the Staphylococcal Factors Involved in Implant Infections. Int J Artif Organs 2018; 29:368-78. [PMID: 16705605 DOI: 10.1177/039139880602900406] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Implant infections due to staphylococci are one of the greatest threats facing patients receiving implant devices. For many years researchers have sought to understand the mechanisms involved in the adherence of the bacterium to the implanted device and the formation of the unique structure, the biofilm, which protects the indwelling bacteria from the host defence and renders them resistant to antibiotic treatment. A major goal has been to develop in vitro and in vivo models that adequately reflect the real-life situation. From the simple microtiter plate assay and scanning electron microscopy, tools for studying adherence and biofilm formation have since evolved to include specialised equipment for studying adherence, flow cell systems, real-time analysis of biofilm formation using reporter gene assays both in vitro and in vivo, and a wide variety of animal models. In this article, we discuss advances in the last few years in selected in vitro and in vivo models as well as future developments in the study of adherence and biofilm formation by the staphylococci.
Collapse
Affiliation(s)
- N Harraghy
- Institute of Medical Microbiology and Hygiene, University of Saarland, Homburg/Saar, Germany.
| | | | | | | | | | | |
Collapse
|
22
|
In Vivo Bioluminescent Monitoring of Therapeutic Efficacy and Pharmacodynamic Target Assessment of Antofloxacin against Escherichia coli in a Neutropenic Murine Thigh Infection Model. Antimicrob Agents Chemother 2017; 62:AAC.01281-17. [PMID: 29038275 DOI: 10.1128/aac.01281-17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/16/2017] [Indexed: 12/14/2022] Open
Abstract
Antimicrobial resistance among uropathogens has increased the rates of infection-related morbidity and mortality. Antofloxacin is a novel fluoroquinolone with broad-spectrum antibacterial activity against urinary Gram-negative bacilli, such as Escherichia coli This study monitored the in vivo efficacy of antofloxacin using bioluminescent imaging and determined pharmacokinetic (PK)/pharmacodynamic (PD) targets against E. coli isolates in a neutropenic murine thigh infection model. The PK properties were determined after subcutaneous administration of antofloxacin at 2.5, 10, 40, and 160 mg/kg of body weight. Following thigh infection, the mice were treated with 2-fold-increasing doses of antofloxacin from 2.5 to 80 mg/kg administered every 12 h. Efficacy was assessed by quantitative determination of the bacterial burdens in thigh homogenates and was compared with the bioluminescent density. Antofloxacin demonstrated both static and killing endpoints in relation to the initial burden against all study strains. The PK/PD index area under the concentration-time curve (AUC)/MIC correlated well with efficacy (R2 = 0.92), and the dose-response relationship was relatively steep, as observed with escalating doses of antofloxacin. The mean free drug AUC/MIC targets necessary to produce net bacterial stasis and 1-log10 and 2-log10 kill for each isolate were 38.7, 66.1, and 147.0 h, respectively. In vivo bioluminescent imaging showed a rapid decrease in the bioluminescent density at free drug AUC/MIC exposures that exceeded the stasis targets. The integration of these PD targets combined with the results of PK studies with humans will be useful in setting optimal dosing regimens for the treatment of urinary tract infections due to E. coli.
Collapse
|
23
|
Avci P, Karimi M, Sadasivam M, Antunes-Melo WC, Carrasco E, Hamblin MR. In-vivo monitoring of infectious diseases in living animals using bioluminescence imaging. Virulence 2017; 9:28-63. [PMID: 28960132 PMCID: PMC6067836 DOI: 10.1080/21505594.2017.1371897] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Traditional methods of localizing and quantifying the presence of pathogenic microorganisms in living experimental animal models of infections have mostly relied on sacrificing the animals, dissociating the tissue and counting the number of colony forming units. However, the discovery of several varieties of the light producing enzyme, luciferase, and the genetic engineering of bacteria, fungi, parasites and mice to make them emit light, either after administration of the luciferase substrate, or in the case of the bacterial lux operon without any exogenous substrate, has provided a new alternative. Dedicated bioluminescence imaging (BLI) cameras can record the light emitted from living animals in real time allowing non-invasive, longitudinal monitoring of the anatomical location and growth of infectious microorganisms as measured by strength of the BLI signal. BLI technology has been used to follow bacterial infections in traumatic skin wounds and burns, osteomyelitis, infections in intestines, Mycobacterial infections, otitis media, lung infections, biofilm and endodontic infections and meningitis. Fungi that have been engineered to be bioluminescent have been used to study infections caused by yeasts (Candida) and by filamentous fungi. Parasitic infections caused by malaria, Leishmania, trypanosomes and toxoplasma have all been monitored by BLI. Viruses such as vaccinia, herpes simplex, hepatitis B and C and influenza, have been studied using BLI. This rapidly growing technology is expected to continue to provide much useful information, while drastically reducing the numbers of animals needed in experimental studies.
Collapse
Affiliation(s)
- Pinar Avci
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,b Department of Dermatology , Harvard Medical School , Boston , MA , USA
| | - Mahdi Karimi
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,c Department of Medical Nanotechnology , School of Advanced Technologies in Medicine, Iran University of Medical Sciences , Tehran , Iran.,d Cellular and Molecular Research Center, Iran University of Medical Sciences , Tehran , Iran
| | - Magesh Sadasivam
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,e Amity Institute of Nanotechnology, Amity University Uttar Pradesh , Noida , India
| | - Wanessa C Antunes-Melo
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,f University of Sao Paulo , Sao Carlos-SP , Brazil
| | - Elisa Carrasco
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,g Department of Biosciences , Durham University , Durham , United Kingdom
| | - Michael R Hamblin
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,b Department of Dermatology , Harvard Medical School , Boston , MA , USA.,h Harvard-MIT Division of Health Sciences and Technology , Cambridge , MA , USA
| |
Collapse
|
24
|
A Human Biofilm-Disrupting Monoclonal Antibody Potentiates Antibiotic Efficacy in Rodent Models of both Staphylococcus aureus and Acinetobacter baumannii Infections. Antimicrob Agents Chemother 2017; 61:AAC.00904-17. [PMID: 28717038 DOI: 10.1128/aac.00904-17] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/11/2017] [Indexed: 12/22/2022] Open
Abstract
Many serious bacterial infections are antibiotic refractory due to biofilm formation. A key structural component of biofilm is extracellular DNA, which is stabilized by bacterial proteins, including those from the DNABII family. TRL1068 is a high-affinity human monoclonal antibody against a DNABII epitope conserved across both Gram-positive and Gram-negative bacterial species. In the present study, the efficacy of TRL1068 for the disruption of biofilm was demonstrated in vitro in the absence of antibiotics by scanning electron microscopy. The in vivo efficacy of this antibody was investigated in a well-characterized catheter-induced aortic valve infective endocarditis model in rats infected with a methicillin-resistant Staphylococcus aureus (MRSA) strain with the ability to form thick biofilms, obtained from the blood of a patient with persistent clinical infection. Animals were treated with vancomycin alone or in combination with TRL1068. MRSA burdens in cardiac vegetations and within intracardiac catheters, kidneys, spleen, and liver showed significant reductions in the combination arm versus vancomycin alone (P < 0.001). A trend toward mortality reduction was also observed (P = 0.09). In parallel, the in vivo efficacy of TRL1068 against a multidrug-resistant clinical Acinetobacter baumannii isolate was explored by using an established mouse model of skin and soft tissue catheter-related biofilm infection. Catheter segments infected with A. baumannii were implanted subcutaneously into mice; animals were treated with imipenem alone or in combination with TRL1068. The combination showed a significant reduction of catheter-adherent bacteria versus the antibiotic alone (P < 0.001). TRL1068 shows excellent promise as an adjunct to standard-of-care antibiotics for a broad range of difficult-to-treat bacterial infections.
Collapse
|
25
|
Identification of Staphylococcus aureus Cellular Pathways Affected by the Stilbenoid Lead Drug SK-03-92 Using a Microarray. Antibiotics (Basel) 2017; 6:antibiotics6030017. [PMID: 28892020 PMCID: PMC5617981 DOI: 10.3390/antibiotics6030017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/25/2017] [Accepted: 09/07/2017] [Indexed: 12/16/2022] Open
Abstract
The mechanism of action for a new lead stilbene compound coded SK-03-92 with bactericidal activity against methicillin-resistant Staphylococcus aureus (MRSA) is unknown. To gain insight into the killing process, transcriptional profiling was performed on SK-03-92 treated vs. untreated S. aureus. Fourteen genes were upregulated and 38 genes downregulated by SK-03-92 treatment. Genes involved in sortase A production, protein metabolism, and transcriptional regulation were upregulated, whereas genes encoding transporters, purine synthesis proteins, and a putative two-component system (SACOL2360 (MW2284) and SACOL2361 (MW2285)) were downregulated by SK-03-92 treatment. Quantitative real-time polymerase chain reaction analyses validated upregulation of srtA and tdk as well as downregulation of the MW2284/MW2285 and purine biosynthesis genes in the drug-treated population. A quantitative real-time polymerase chain reaction analysis of MW2284 and MW2285 mutants compared to wild-type cells demonstrated that the srtA gene was upregulated by both putative two-component regulatory gene mutants compared to the wild-type strain. Using a transcription profiling technique, we have identified several cellular pathways regulated by SK-03-92 treatment, including a putative two-component system that may regulate srtA and other genes that could be tied to the SK-03-92 mechanism of action, biofilm formation, and drug persisters.
Collapse
|
26
|
Integrating printed microfluidics with silicon photomultipliers for miniaturised and highly sensitive ATP bioluminescence detection. Biosens Bioelectron 2017; 99:464-470. [PMID: 28820988 DOI: 10.1016/j.bios.2017.07.055] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/17/2017] [Accepted: 07/21/2017] [Indexed: 11/20/2022]
Abstract
Bioluminescence has been widely used for important biosensing applications such as the measurement of adenosine triphosphate (ATP), the energy unit in biological systems and an indicator of vital processes. The current technology for detection is mainly based on large equipment such as readers and imaging systems, which require intensive and time-consuming procedures. A miniaturised bioluminescence sensing system, which would allow sensitive and continuous monitoring of ATP, with an integrated and low-cost disposable microfluidic chamber for handling of biological samples, is highly desirable. Here, we report the design, fabrication and testing of 3D printed microfluidics chips coupled with silicon photomultipliers (SiPMs) for high sensitive real-time ATP detection. The 3D microfluidic chip reduces reactant consumption and facilitates solution delivery close to the SiPM to increase the detection efficiency. Our system detects ATP with a limit of detection (LoD) of 8nM and an analytical dynamic range between 15nM and 1µM, showing a stability error of 3%, and a reproducibility error below of 20%. We demonstrate the dynamic monitoring of ATP in a continuous-flow system exhibiting a fast response time, ~4s, and a full recovery to the baseline level within 17s. Moreover, the SiPM-based bioluminescence sensing system shows a similar analytical dynamic range for ATP detection to that of a full-size PerkinElmer laboratory luminescence reader.
Collapse
|
27
|
Singh KV, Tran TT, Nannini EC, Tam VH, Arias CA, Murray BE. Efficacy of Ceftaroline against Methicillin-Susceptible Staphylococcus aureus Exhibiting the Cefazolin High-Inoculum Effect in a Rat Model of Endocarditis. Antimicrob Agents Chemother 2017; 61:e00324-17. [PMID: 28483961 PMCID: PMC5487651 DOI: 10.1128/aac.00324-17] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/04/2017] [Indexed: 12/16/2022] Open
Abstract
Certain Staphylococcus aureus strains exhibit an inoculum effect (InE) with cefazolin (CFZ) that has been associated with therapeutic failures in high-inoculum infections. We assessed the in vitro activities of ceftaroline (CPT), CFZ, and nafcillin (NAF) against 17 type A β-lactamase (βla)-producing, methicillin-susceptible S. aureus (MSSA) strains, including the previously reported TX0117, which exhibits the CFZ InE, and its βla-cured derivative, TX0117c. Additionally, we determined the pharmacokinetics of CPT in rats after single intramuscular doses of 20 and 40 mg/kg of body weight and evaluated the activities of CPT (40 mg/kg every 8 h [q8h]), CFZ, and NAF against TX0117 and TX0117c in a rat model of infective endocarditis. No InE was observed for CPT or NAF, whereas a marked InE was detected for CFZ (MIC, 8 to ≥128 μg/ml). CPT and NAF treatment against TX0117 resulted in mean bacterial counts of 2.3 and 2.1 log10 CFU/g in vegetations, respectively, compared to a mean of 5.9 log10 CFU/g in the CFZ-treated group (CPT and NAF versus CFZ, P = 0.001; CPT versus NAF, P = 0.9830). Both CFZ and CPT were efficacious against the βla-cured derivative, TX0117c, compared to time zero (t0) (P = <0.0001 and 0.0015, respectively). Our data reiterate the in vivo consequences of the CFZ InE and show that CPT is not affected by this phenomenon. CPT might be considered for high-inoculum infections caused by MSSA exhibiting the CFZ InE.
Collapse
Affiliation(s)
- Kavindra V Singh
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
- Center for Antimicrobial Resistance and Microbial Genomics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Truc T Tran
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
- Center for Antimicrobial Resistance and Microbial Genomics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Esteban C Nannini
- Division of Infectious Diseases, School of Medicine, Universidad Nacional de Rosario,. Instituto de Inmunología Clínica y Experimental Rosario (IDICER), CONICET, Rosario, Argentina
| | - Vincent H Tam
- College of Pharmacy, University of Houston, Houston, Texas, USA
| | - Cesar A Arias
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
- Department of Microbiology and Molecular Genetics, University of Texas Medical School at Houston, Houston, Texas, USA
- Molecular Genetics and Antimicrobial Resistance Unit, Universidad El Bosque, Bogota, Colombia
- Center for Antimicrobial Resistance and Microbial Genomics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Barbara E Murray
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
- Department of Microbiology and Molecular Genetics, University of Texas Medical School at Houston, Houston, Texas, USA
- Center for Antimicrobial Resistance and Microbial Genomics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| |
Collapse
|
28
|
Suhardi VJ, Bichara DA, Kwok S, Freiberg AA, Rubash H, Malchau H, Yun SH, Muratoglu OK, Oral E. A Fully Functional Drug-Eluting Joint Implant. Nat Biomed Eng 2017; 1:0080. [PMID: 29354321 PMCID: PMC5773111 DOI: 10.1038/s41551-017-0080] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 05/02/2017] [Indexed: 12/13/2022]
Abstract
Despite advances in orthopedic materials, the development of drug-eluting bone and joint implants that can sustain the delivery of the drug and maintain the necessary mechanical strength in order to withstand loading has remained elusive. Here, we demonstrate that modifying the eccentricity of drug clusters and the percolation threshold in ultrahigh molecular weight polyethylene (UHMWPE) results in maximized drug elution and in the retention of mechanical strength. The optimized UHMWPE eluted antibiotic at a higher concentration for longer than the clinical gold standard antibiotic-eluting bone cement while retaining the mechanical and wear properties of clinically used UHMWPE joint prostheses. Treatment of lapine knees infected with Staphylococcus aureus with the antibiotic-eluting UHMWPE led to complete bacterial eradication and to the absence of detectable systemic effects. We argue that the antibiotic-eluting UHMWPE joint implant is a promising candidate for clinical trials.
Collapse
Affiliation(s)
- V J Suhardi
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA
- Department of Orthopaedic Surgery, Harvard Medical School
- Department of Medical Engineering and Medical Physics, Massachusetts Institute of Technology
| | - D A Bichara
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA
- Department of Orthopaedic Surgery, Harvard Medical School
| | - Sjj Kwok
- Department of Medical Engineering and Medical Physics, Massachusetts Institute of Technology
- Wellmann Center for Photomedicine, Massachusetts General Hospital, Boston, MA
| | - A A Freiberg
- Department of Orthopaedic Surgery, Harvard Medical School
| | - H Rubash
- Department of Orthopaedic Surgery, Harvard Medical School
| | - H Malchau
- Department of Orthopaedic Surgery, Harvard Medical School
| | - S H Yun
- Department of Medical Engineering and Medical Physics, Massachusetts Institute of Technology
- Wellmann Center for Photomedicine, Massachusetts General Hospital, Boston, MA
| | - O K Muratoglu
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA
- Department of Orthopaedic Surgery, Harvard Medical School
| | - E Oral
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA
- Department of Orthopaedic Surgery, Harvard Medical School
| |
Collapse
|
29
|
Efficacy of Telavancin Alone and in Combination with Ampicillin in a Rat Model of Enterococcus faecalis Endocarditis. Antimicrob Agents Chemother 2017; 61:AAC.02489-16. [PMID: 28320712 DOI: 10.1128/aac.02489-16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 03/06/2017] [Indexed: 11/20/2022] Open
Abstract
We first assessed telavancin (TLV) pharmacokinetics in rats after a single subcutaneous dose of 35 mg/kg of body weight. The pharmacokinetic data were used to predict a TLV dose that simulates human exposure, and the efficacy of TLV was then evaluated using a TLV dose of 21 mg/kg every 12 h against Enterococcus faecalis OG1RF (TLV MIC of 0.06 μg/ml) in a rat endocarditis model with an indwelling catheter. Therapy was given for 3 days with TLV, daptomycin (DAP), or ampicillin (AMP) monotherapy and with combinations of TLV plus AMP, AMP plus gentamicin (GEN), and AMP plus ceftriaxone (CRO); rats were sacrificed 24 h after the last dose. Antibiotics were given to simulate clinically relevant concentrations or as used in other studies. TLV treatment resulted in a significant decrease in bacterial burden (CFU per gram) in vegetations from 6.0 log10 at time 0 to 3.1 log10 after 3 days of therapy. Bacterial burdens in vegetations were also significantly lower in the TLV-treated rats than in the AMP (P = 0.0009)- and AMP-plus-GEN (P = 0.035)-treated rats but were not significantly different from that of the AMP-plus-CRO-treated rats. Bacterial burdens from vegetations in TLV monotherapy and TLV-plus-AMP-and-DAP groups were similar to each other (P ≥ 0.05). Our data suggest that further study of TLV as a therapeutic alternative for deep-seated infections caused by vancomycin-susceptible E. faecalis is warranted.
Collapse
|
30
|
Bielen K, 's Jongers B, Malhotra-Kumar S, Jorens PG, Goossens H, Kumar-Singh S. Animal models of hospital-acquired pneumonia: current practices and future perspectives. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:132. [PMID: 28462212 DOI: 10.21037/atm.2017.03.72] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Lower respiratory tract infections are amongst the leading causes of mortality and morbidity worldwide. Especially in hospital settings and more particularly in critically ill ventilated patients, nosocomial pneumonia is one of the most serious infectious complications frequently caused by opportunistic pathogens. Pseudomonas aeruginosa is one of the most important causes of ventilator-associated pneumonia as well as the major cause of chronic pneumonia in cystic fibrosis patients. Animal models of pneumonia allow us to investigate distinct types of pneumonia at various disease stages, studies that are not possible in patients. Different animal models of pneumonia such as one-hit acute pneumonia models, ventilator-associated pneumonia models and biofilm pneumonia models associated with cystic fibrosis have been extensively studied and have considerably aided our understanding of disease pathogenesis and testing and developing new treatment strategies. The present review aims to guide investigators in choosing appropriate animal pneumonia models by describing and comparing the relevant characteristics of each model using P. aeruginosa as a model etiology for hospital-acquired pneumonia. Key to establishing and studying these animal models of infection are well-defined end-points that allow precise monitoring and characterization of disease development that could ultimately aid in translating these findings to patient populations in order to guide therapy. In this respect, and discussed here, is the development of humanized animal models of bacterial pneumonia that could offer unique advantages to study bacterial virulence factor expression and host cytokine production for translational purposes.
Collapse
Affiliation(s)
- Kenny Bielen
- Molecular Pathology Group, Faculty of Medicine and Health Sciences, Laboratory of Cell Biology and Histology, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium.,Laboratory of Medical Microbiology - Vaccine and Infectious Disease Institute, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Bart 's Jongers
- Molecular Pathology Group, Faculty of Medicine and Health Sciences, Laboratory of Cell Biology and Histology, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium.,Laboratory of Medical Microbiology - Vaccine and Infectious Disease Institute, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Surbhi Malhotra-Kumar
- Laboratory of Medical Microbiology - Vaccine and Infectious Disease Institute, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Philippe G Jorens
- Department of Critical Care Medicine, Antwerp University Hospital and University of Antwerp, LEMP, Wilrijkstraat 10, B-2650 Edegem, Belgium
| | - Herman Goossens
- Laboratory of Medical Microbiology - Vaccine and Infectious Disease Institute, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Samir Kumar-Singh
- Molecular Pathology Group, Faculty of Medicine and Health Sciences, Laboratory of Cell Biology and Histology, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium.,Laboratory of Medical Microbiology - Vaccine and Infectious Disease Institute, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| |
Collapse
|
31
|
Comparative Efficacies of Tedizolid Phosphate, Linezolid, and Vancomycin in a Murine Model of Subcutaneous Catheter-Related Biofilm Infection Due to Methicillin-Susceptible and -Resistant Staphylococcus aureus. Antimicrob Agents Chemother 2016; 60:5092-6. [PMID: 27297485 DOI: 10.1128/aac.00880-16] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 06/07/2016] [Indexed: 12/17/2022] Open
Abstract
Tedizolid, a novel oxazolidinone, exhibits bacteriostatic activity through inhibition of protein synthesis. The efficacies of tedizolid, linezolid, and vancomycin were compared in a murine catheter-related biofilm infection caused by methicillin-susceptible and -resistant Staphylococcus aureus (MSSA and MRSA, respectively) strains engineered for bioluminescence. We observed significantly improved efficacy in terms of decreased S. aureus densities and bioluminescent signals in the tedizolid-treated group versus the linezolid- and vancomycin-treated groups in the model of infection caused by the MSSA and MRSA strains.
Collapse
|
32
|
Stevens CS, Rosado H, Harvey RJ, Taylor PW. Epicatechin gallate, a naturally occurring polyphenol, alters the course of infection with β-lactam-resistant Staphylococcus aureus in the zebrafish embryo. Front Microbiol 2015; 6:1043. [PMID: 26441953 PMCID: PMC4585009 DOI: 10.3389/fmicb.2015.01043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 09/14/2015] [Indexed: 11/17/2022] Open
Abstract
(-)-epicatechin gallate (ECg) substantially modifies the properties of Staphylococcus aureus and reversibly abrogates β-lactam resistance in methicillin/oxacillin resistant (MRSA) isolates. We have determined the capacity of ECg to alter the course of infection in zebrafish embryos challenged with epidemic clinical isolate EMRSA-16. At 30 h post fertilization (hpf), embryos were infected by injection of 1–5 × 103 colony forming units (CFU) of EMRSA-16 into the circulation valley or yolk sac. Infection by yolk sac injection was lethal with a challenge dose above 3 × 103 CFU, with no survivors at 70 hpf. In contrast, survival at 70 hpf after injection into the circulation was 83 and 44% following challenge with 3 × 103 and 1–5 × 103 CFU, respectively. No significant increases in survival were noted when infected embryos were maintained in medium containing 12.5–100 μg/mL ECg with or without 4 or 16 μg/mL oxacillin. However, when EMRSA-16 was grown in medium containing 12.5 μg/mL ECg and the bacteria used to infect embryos by either the circulation valley or yolk sac, there were significant increases in embryo survival in both the presence and absence of oxacillin. ECg-modified and unmodified, GFP-transformed EMRSA-16 bacteria were visualized within phagocytic cells in the circulation and yolk sac; pre-treatment with ECg also significantly increased induction of the respiratory burst and suppressed increases in IL-1β expression typical of infection with untreated EMRSA-16. We conclude that exposure to ECg prior to infection reduces the lethality of EMRSA-16, renders cells more susceptible to elimination by immune processes and compromises their capacity to establish an inflammatory response in comparison to non-exposed bacteria.
Collapse
Affiliation(s)
| | - Helena Rosado
- UCL School of Pharmacy, University College London London, UK
| | - Robert J Harvey
- UCL School of Pharmacy, University College London London, UK
| | - Peter W Taylor
- UCL School of Pharmacy, University College London London, UK
| |
Collapse
|
33
|
Terai T, Okumura T, Imai S, Nakao M, Yamaji K, Ito M, Nagata T, Kaneko K, Miyazaki K, Okada A, Nomura Y, Hanada N. Screening of Probiotic Candidates in Human Oral Bacteria for the Prevention of Dental Disease. PLoS One 2015; 10:e0128657. [PMID: 26053410 PMCID: PMC4459870 DOI: 10.1371/journal.pone.0128657] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 04/29/2015] [Indexed: 11/18/2022] Open
Abstract
The oral cavity in healthy subjects has a well-balanced microbiota that consists of more than 700 species. However, a disturbance of this balance, with an increase of harmful microbes and a decrease of beneficial microbes, causes oral disorders such as periodontal disease or dental caries. Nowadays, probiotics are expected to confer oral health benefits by modulating the oral microbiota. This study screened new probiotic candidates with potential oral health benefits and no harmful effects on the oral cavity. We screened 14 lactobacillus strains and 36 streptococcus strains out of 896 oral isolates derived from healthy subjects. These bacteria did not produce volatile sulfur compounds or water-insoluble glucan, had higher antibacterial activity against periodontal bacteria, and had higher adherence activity to oral epithelial cells or salivary-coated hydroxyapatite in vitro. We then evaluated the risk of primary cariogenicity and infective endocarditis of the selected oral isolates. As a result, Lactobacillus crispatus YIT 12319, Lactobacillus fermentum YIT 12320, Lactobacillus gasseri YIT 12321, and Streptococcus mitis YIT 12322 were selected because they showed no cariogenic potential in an artificial mouth system and a lower risk of experimental infective endocarditis in a rat model. These candidates are expected as new probiotics with potential oral health benefits and no adverse effects on general health.
Collapse
Affiliation(s)
- Tomohiko Terai
- Yakult Central Institute, Kunitachi, Tokyo, Japan
- * E-mail:
| | | | - Susumu Imai
- Department of Translational Research, Tsurumi University School of Dental Medicine, Yokohama, Kanagawa, Japan
| | - Masumi Nakao
- Yakult Central Institute, Kunitachi, Tokyo, Japan
| | | | - Masahiko Ito
- Yakult Central Institute, Kunitachi, Tokyo, Japan
| | | | | | | | - Ayako Okada
- Department of Translational Research, Tsurumi University School of Dental Medicine, Yokohama, Kanagawa, Japan
| | - Yoshiaki Nomura
- Department of Translational Research, Tsurumi University School of Dental Medicine, Yokohama, Kanagawa, Japan
| | - Nobuhiro Hanada
- Department of Translational Research, Tsurumi University School of Dental Medicine, Yokohama, Kanagawa, Japan
| |
Collapse
|
34
|
Bruckbauer ST, Kvitko BH, Karkhoff-Schweizer RR, Schweizer HP. Tn5/7-lux: a versatile tool for the identification and capture of promoters in gram-negative bacteria. BMC Microbiol 2015; 15:17. [PMID: 25648327 PMCID: PMC4328036 DOI: 10.1186/s12866-015-0354-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 01/19/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The combination of imaging technologies and luciferase-based bioluminescent bacterial reporter strains provide a sensitive and simple non-invasive detection method (photonic bioimaging) for the study of diverse biological processes, as well as efficacy of therapeutic interventions, in live animal models of disease. The engineering of bioluminescent bacteria required for photonic bioimaging is frequently hampered by lack of promoters suitable for strong, yet stable luciferase gene expression. RESULTS We devised a novel method for identification of constitutive native promoters in Gram-negative bacteria. The method is based on a Tn5/7 transposon that exploits the unique features of Tn5 (random transposition) and Tn7 (site-specific transposition). The transposons are designed such that Tn5 transposition will allow insertion of a promoter-less bacterial luxCDABE operon downstream of a bacterial gene promoter. Cloning of DNA fragments from luminescent isolates results in a plasmid that replicates in pir (+) hosts. Sequencing of the lux-chromosomal DNA junctions on the plasmid reveals transposon insertion sites within genes or operons. The plasmid is also a mini-Tn7-lux delivery vector that can be used to introduce the promoter-lux operon fusion into other derivatives of the bacterium of interest in an isogenic fashion. Alternatively, promoter-containing sequences can be PCR-amplified from plasmid or chromosomal DNA and cloned into a series of accompanying mini-Tn7-lux vectors. The mini-Tn5/7-lux and mini-Tn7-lux vectors are equipped with diverse selection markers and thus applicable in numerous Gram-negative bacteria. Various mini-Tn5/7-lux vectors were successfully tested for transposition and promoter identification by imaging in Acinetobacter baumannii, Escherichia coli, and Burkholderia pseudomallei. Strong promoters were captured for lux expression in E. coli and A. baumannii. Some mini-Tn7-lux vectors are also equipped with attB sites for swapping of the lux operon with other reporter genes using Gateway technology. CONCLUSIONS Although mini-Tn5-lux and mini-Tn7-lux elements have previously been developed and used for bacterial promoter identification and chromosomal insertion of promoter-lux gene fusions, respectively, the newly developed mini-Tn5/7-lux and accompanying accessory plasmids streamline and accelerate the promoter discovery and bioluminescent strain engineering processes. Availability of vectors with diverse selection markers greatly extend the host-range of promoter probe and lux gene fusion vectors.
Collapse
Affiliation(s)
- Steven T Bruckbauer
- Department of Microbiology, Immunology and Pathology, and Rocky Mountain Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, Colorado State University, Fort Collins, 80523, CO, USA. .,Present Address: Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, 53706, WI, USA.
| | - Brian H Kvitko
- Department of Microbiology, Immunology and Pathology, and Rocky Mountain Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, Colorado State University, Fort Collins, 80523, CO, USA. .,Present Address: MSU-DOE Plant Research Laboratory, Michigan State University, East Lansing, 48824, MI, USA.
| | - RoxAnn R Karkhoff-Schweizer
- Department of Microbiology, Immunology and Pathology, and Rocky Mountain Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, Colorado State University, Fort Collins, 80523, CO, USA.
| | - Herbert P Schweizer
- Department of Microbiology, Immunology and Pathology, and Rocky Mountain Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, Colorado State University, Fort Collins, 80523, CO, USA. .,Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Emerging Pathogens Institute, PO Box 100266, Gainesville, 32610-0266, FL, USA.
| |
Collapse
|
35
|
Yoshioka K, Ishii K, Kuramoto T, Nagai S, Funao H, Ishihama H, Shiono Y, Sasaki A, Aizawa M, Okada Y, Koyasu S, Toyama Y, Matsumoto M. A novel mouse model of soft-tissue infection using bioluminescence imaging allows noninvasive, real-time monitoring of bacterial growth. PLoS One 2014; 9:e106367. [PMID: 25184249 PMCID: PMC4153648 DOI: 10.1371/journal.pone.0106367] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 07/28/2014] [Indexed: 12/18/2022] Open
Abstract
Musculoskeletal infections, including surgical-site and implant-associated infections, often cause progressive inflammation and destroy areas of the soft tissue. Treating infections, especially those caused by multi-antibiotic resistant bacteria such as methicillin-resistant Staphylococcus aureus (MRSA) remains a challenge. Although there are a few animal models that enable the quantitative evaluation of infection in soft tissues, these models are not always reproducible or sustainable. Here, we successfully established a real-time, in vivo, quantitative mouse model of soft-tissue infection in the superficial gluteus muscle (SGM) using bioluminescence imaging. A bioluminescent strain of MRSA was inoculated into the SGM of BALB/c adult male mice, followed by sequential measurement of bacterial photon intensity and serological and histological analyses of the mice. The mean photon intensity in the mice peaked immediately after inoculation and remained stable until day 28. The serum levels of interleukin-6, interleukin-1 and C-reactive protein at 12 hours after inoculation were significantly higher than those prior to inoculation, and the C-reactive protein remained significantly elevated until day 21. Histological analyses showed marked neutrophil infiltration and abscesses containing necrotic and fibrous tissues in the SGM. With this SGM mouse model, we successfully visualized and quantified stable bacterial growth over an extended period of time with bioluminescence imaging, which allowed us to monitor the process of infection without euthanizing the experimental animals. This model is applicable to in vivo evaluations of the long-term efficacy of novel antibiotics or antibacterial implants.
Collapse
Affiliation(s)
- Kenji Yoshioka
- Department of Orthopedic Surgery, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Ken Ishii
- Department of Orthopedic Surgery, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
- Kanagawa Academy of Science and Technology (KAST), Kawasaki, Kanagawa, Japan
- * E-mail:
| | - Tetsuya Kuramoto
- Department of Orthopedic Surgery, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Shigenori Nagai
- Department of Microbiology and Immunology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo, Japan
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Haruki Funao
- Department of Orthopedic Surgery, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Hiroko Ishihama
- Department of Orthopedic Surgery, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Yuta Shiono
- Department of Orthopedic Surgery, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Aya Sasaki
- Department of Pathology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Mamoru Aizawa
- Department of Applied Chemistry, School of Science and Technology, Meiji University, Kawasaki, Kanagawa, Japan
- Kanagawa Academy of Science and Technology (KAST), Kawasaki, Kanagawa, Japan
| | - Yasunori Okada
- Department of Pathology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Shigeo Koyasu
- Department of Microbiology and Immunology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
- Laboratory for Immune Cell System, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa, Japan
| | - Yoshiaki Toyama
- Department of Orthopedic Surgery, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Morio Matsumoto
- Department of Orthopedic Surgery, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| |
Collapse
|
36
|
Jiang H, Feng J, Du Z, Zhen H, Lin M, Jia S, Li T, Huang X, Ostenson CG, Chen Z. Oral administration of soybean peptide Vglycin normalizes fasting glucose and restores impaired pancreatic function in Type 2 diabetic Wistar rats. J Nutr Biochem 2014; 25:954-63. [PMID: 24985367 DOI: 10.1016/j.jnutbio.2014.04.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 03/31/2014] [Accepted: 04/17/2014] [Indexed: 12/18/2022]
Abstract
Vglycin, a natural 37-residue polypeptide isolated from pea seeds in which six half-cysteine residues are embedded in three pairs of disulfide bonds, is resistant to digestive enzymes and has antidiabetic potential. To investigate the pharmacological activity of Vglycin in vivo and to examine the mechanisms involved, the therapeutic effect of Vglycin in diabetic rats was examined. Diabetes was induced in Wistar rats by high-fat diet and multiple streptozotocin intraperitoneal injections. Diabetic rats were treated daily with Vglycin for 4 weeks. Body weight, food intake, fasting plasma glucose and insulin levels were assayed weekly. Glucose and insulin tolerance tests were conducted on Day 29. Subsequently, levels of p-Akt in the liver and pancreas and cleaved PARP, Pdx-1 and insulin in the pancreas were detected by immunoblotting. The morphology of the pancreas and the insulin expression in the pancreas were analyzed by hematoxylin-eosin staining and immunohistochemistry, respectively. Furthermore, human liver-derived cell lines were used to explore the in vitro effects of Vglycin on insulin sensitivity and glucose uptake. Chronic treatment with Vglycin normalized fasting glucose levels in diabetic rats. The improvement in glucose homeostasis and the increased insulin sensitivity mediated by restored insulin signaling likely contributed to decreased food intake and reduced body weight. Vglycin protected pancreatic cells from damage by streptozotocin. Although insulin synthesis and secretion in impaired β-cell were not significantly elevated, islets morphology was improved in the Vglycin-treated groups. These results suggest that Vglycin could be useful in Type 2 diabetes for restoring impaired insulin signaling, glucose tolerance and pancreatic function.
Collapse
Affiliation(s)
- Hua Jiang
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Wuhan, P.R. China
| | - Jueping Feng
- Department of Oncology, Wuhan Pu-Ai Hospital, Tong ji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Zhongxia Du
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Wuhan, P.R. China
| | - Hui Zhen
- Department of Clinical Laboratory, the Second Staff Hospital of Wuhan Iron and Steel (Group) Corporation, Wuhan, P.R. China
| | - Mei Lin
- Department of Oncology, Wuhan Pu-Ai Hospital, Tong ji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Shaohui Jia
- College of Health Science, Wuhan Sports University, Wuhan, P.R. China
| | - Tao Li
- Department of narcotics control, Chongqing Police College, Chongqing City, P.R. China
| | - Xinyuan Huang
- College of Life Science and Technology, Hubei Engineering University, Xiaogan, P.R. China
| | | | - Zhengwang Chen
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Wuhan, P.R. China.
| |
Collapse
|
37
|
Slate AR, Bandyopadhyay S, Francis KP, Papich MG, Karolewski B, Hod EA, Prestia KA. Efficacy of enrofloxacin in a mouse model of sepsis. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2014; 53:381-386. [PMID: 25199094 PMCID: PMC4113238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 11/06/2013] [Accepted: 12/06/2013] [Indexed: 06/03/2023]
Abstract
We examined the efficacy of enrofloxacin administered by 2 different routes in a mouse model of sepsis. Male CD1 mice were infected with a bioluminescent strain of enteropathogenic Escherichia coli and treated with enrofloxacin either by injection or in drinking water. Peak serum levels were evaluated by using HPLC. Mice were monitored for signs of clinical disease, and infections were monitored by using bioluminescence imaging. Serum levels of enrofloxacin and the active metabolite ciprofloxacin were greater in the group treated by injection than in controls or the groups treated by administration in drinking water. Survival of the group treated with enrofloxacin injection was greater than that of controls and groups treated with enrofloxacin in the drinking water. Bioluminescence in the group treated with enrofloxacin injection was less than that in the groups treated with oral administration at 12 h and in the groups treated orally and the control group at 16 h. According to these findings, we recommend the use of injectable enrofloxacin at 5 mg/kg SC for mice with systemic infections.
Collapse
Affiliation(s)
- Andrea R Slate
- Division of Comparative Medicine, University of South Florida, Tampa, Florida, USA
| | - Sheila Bandyopadhyay
- Laboratory of Transfusion Biology, Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | | | - Mark G Papich
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
| | - Brian Karolewski
- Institute of Comparative Medicine, Columbia University, New York, New York, USA
| | - Eldad A Hod
- Laboratory of Transfusion Biology, Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Kevin A Prestia
- Laboratory of Transfusion Biology, Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York, USA; Institute of Comparative Medicine, Columbia University, New York, New York, USA
| |
Collapse
|
38
|
Panizzi P, Stone JR, Nahrendorf M. Endocarditis and molecular imaging. J Nucl Cardiol 2014; 21:486-95. [PMID: 24797384 PMCID: PMC4106242 DOI: 10.1007/s12350-014-9902-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 04/03/2014] [Indexed: 10/25/2022]
Affiliation(s)
- Peter Panizzi
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | - James R. Stone
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge St., Boston, MA 02114, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge St., Boston, MA 02114, USA
| |
Collapse
|
39
|
Gong J, Li D, Yan J, Liu Y, Li D, Dong J, Gao Y, Sun T, Yang G. The accessory gene regulator (agr) controls Staphylococcus aureus virulence in a murine intracranial abscesses model. Braz J Infect Dis 2014; 18:501-6. [PMID: 24833200 PMCID: PMC9428208 DOI: 10.1016/j.bjid.2014.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 02/20/2014] [Accepted: 03/18/2014] [Indexed: 01/14/2023] Open
Abstract
Background Intracranial abscesses are associated with high mortality. Staphylococcus aureus is one of the main pathogens that cause intracranial infection. Until now, there is no report to identify the key effectors of S. aureus during the intracranial infection. Methods The murine intracranial abscesses model induced by S. aureus was constructed. The vital sign and survival rate of mice were observed to evaluate the infection. Histological examination was used to diagnose the pathological alterations of mouse tissues. The sensitivity of S. aureus to whole blood was evaluated by whole-blood killing assay. Results In murine intracranial abscesses model, it was shown that the mortality caused by the accessory gene regulator (agr) locus deficient strain was significant decreased compared with its parent strain. Moreover, we found that RNAIII, the effector of agr system, was essential for the intracranial infection caused by S. aureus. In the further investigation, it was shown that restoration the expression of α-toxin in agr deficient strain could partially recover the mortality in the murine intracranial abscesses model. Conclusion Our data suggested that the agr system of S. aureus is an important virulence determinant in the induction and mortality of intracranial abscesses in mice.
Collapse
Affiliation(s)
- Jian Gong
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing Neurosurgical Institute, Beijing, China
| | - Dongzhi Li
- Ningxia Medical University, Incubation Base of National Key Laboratory for Cerebrocranial Diseases, Yinchuan, China
| | - Jun Yan
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yu Liu
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Di Li
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jie Dong
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yaping Gao
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Tao Sun
- Ningxia Medical University, Incubation Base of National Key Laboratory for Cerebrocranial Diseases, Yinchuan, China.
| | - Guang Yang
- Beijing Institute of Basic Medical Sciences, Beijing, China.
| |
Collapse
|
40
|
Combination prophylactic therapy with rifampin increases efficacy against an experimental Staphylococcus epidermidis subcutaneous implant-related infection. Antimicrob Agents Chemother 2014; 58:2377-86. [PMID: 24514089 DOI: 10.1128/aac.01943-13] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The incidence of infections related to cardiac devices (such as permanent pacemakers) has been increasing out of proportion to implantation rates. As management of device infections typically requires explantation of the device, optimal prophylactic strategies are needed. Cefazolin and vancomycin are widely used as single agents for surgical prophylaxis against cardiac device-related infections. However, combination antibiotic prophylaxis may further reduce infectious complications. To model a localized subcutaneous implant-related infection, a bioluminescent strain of Staphylococcus epidermidis was inoculated onto a medical-procedure-grade titanium disc, which was placed into a subcutaneous pocket in the backs of mice. In vivo bioluminescence imaging, quantification of ex vivo CFU from the capsules and implants, variable-pressure scanning electron microscopy (VP-SEM), and neutrophil enhanced green fluorescent protein (EGFP) fluorescence in LysEGFP mice were employed to monitor the infection. This model was used to evaluate the efficacies of low- and high-dose cefazolin (50 and 200 mg/kg of body weight) and vancomycin (10 and 110 mg/kg) intravenous prophylaxis with or without rifampin (25 mg/kg). High-dose cefazolin and high-dose vancomycin treatment resulted in almost complete bacterial clearance, whereas both low-dose cefazolin and low-dose vancomycin reduced the in vivo and ex vivo bacterial burden only moderately. The addition of rifampin to low-dose cefazolin and vancomycin was highly effective in further reducing the CFU harvested from the implants. However, vancomycin-rifampin was more effective than cefazolin-rifampin in further reducing the CFU harvested from the surrounding tissue capsules. Future studies in humans will be required to determine whether the addition of rifampin has improved efficacy in preventing device-related infections in clinical practice.
Collapse
|
41
|
Noninvasive imaging of Staphylococcus aureus infections with a nuclease-activated probe. Nat Med 2014; 20:301-6. [PMID: 24487433 PMCID: PMC3949172 DOI: 10.1038/nm.3460] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 04/26/2013] [Indexed: 12/21/2022]
Abstract
Technologies that enable the rapid detection and localization of bacterial infections in living animals could address an unmet need for infectious disease diagnostics. We describe a molecular imaging approach for the specific, non-invasive detection of S. aureus based on the activity of its secreted nuclease, micrococcal nuclease (MN). Several short, synthetic oligonucleotides, rendered resistant to mammalian serum nucleases by various chemical modifications, flanked with a fluorophore and quencher, were activated upon degradation by recombinant MN and in S. aureus culture supernatants. A probe consisting of a pair of deoxythymidines flanked by several 2′-O-methyl-modified nucleotides was activated in culture supernatants of S. aureus but not in culture supernatants of several other pathogenic bacteria. Systemic administration of this probe to mice bearing bioluminescent S. aureus muscle infections resulted in probe activation at the infection sites in an MN-dependent manner. This novel bacterial imaging approach has potential clinical applicability for S. aureus and several other medically significant pathogens.
Collapse
|
42
|
In vitro and in vivo bioluminescent imaging to evaluate anti-Escherichia coli activity of Galla Chinensis. Biomedicine (Taipei) 2013. [DOI: 10.1016/j.biomed.2013.04.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
43
|
Gupta RK, Alba J, Xiong YQ, Bayer AS, Lee CY. MgrA activates expression of capsule genes, but not the α-toxin gene in experimental Staphylococcus aureus endocarditis. J Infect Dis 2013; 208:1841-8. [PMID: 23901087 DOI: 10.1093/infdis/jit367] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Staphylococcus aureus produces numerous virulence factors but little is known about their in vivo regulation during an infection. METHODS The production of capsule and α-toxin, and the expression of their respective genes, cap5 and hla, were analyzed by comparing CYL11481 (derivative of Newman) and its isogenic regulatory mutants in vitro. The temporal expression of cap5 and hla and the regulatory genes in vivo was carried out using a rat infective endocarditis model. RESULTS In vitro analyses showed that capsule was positively regulated by MgrA, Agr, Sae, ArlR, and ClpC, and negatively by CodY and SbcDC. The α-toxin was positively regulated by MgrA, Agr, Sae, ArlR, and SbcDC but negatively by ClpC and CodY. In vivo analyses showed that cap5 expression correlated best with mgrA expression, whereas hla expression correlated best with sae expression. Mutation in mgrA drastically reduced cap5 expression in vivo. CONCLUSIONS Our results suggest that, in vitro, Agr is the most important regulator for capsule and α-toxin production, as well as for cap5 transcription, but SaeR is the most critical for hla transcription. However, in vivo, MgrA is the major transcriptional regulator of capsule, but not α-toxin, whereas saeR expression correlates best with hla expression.
Collapse
Affiliation(s)
- Ravi Kr Gupta
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock
| | | | | | | | | |
Collapse
|
44
|
Velkov T, Bergen PJ, Lora-Tamayo J, Landersdorfer CB, Li J. PK/PD models in antibacterial development. Curr Opin Microbiol 2013; 16:573-9. [PMID: 23871724 DOI: 10.1016/j.mib.2013.06.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 05/09/2013] [Accepted: 06/25/2013] [Indexed: 01/08/2023]
Abstract
There is an urgent need for novel antibiotics to treat life-threatening infections caused by bacterial 'superbugs'. Validated in vitro pharmacokinetic/pharmacodynamic (PK/PD) and animal infection models have been employed to identify the most predictive PK/PD indices and serve as key tools in the antibiotic development process. The results obtained can be utilized for optimizing study designs in order to minimize the cost and duration of clinical trials. This review outlines the key in vitro PK/PD and animal infection models which have been extensively used in antibiotic discovery and development. These models have shown great potential in accelerating drug development programs and will continue to make significant contributions to antibiotic development.
Collapse
Affiliation(s)
- Tony Velkov
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | | | | | | | | |
Collapse
|
45
|
In Vivo Effects of Cefazolin, Daptomycin, and Nafcillin in Experimental Endocarditis with a Methicillin-Susceptible Staphylococcus aureus Strain Showing an Inoculum Effect against Cefazolin. Antimicrob Agents Chemother 2013; 57:4276-4281. [PMID: 23796934 DOI: 10.1128/aac.00856-13] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 06/14/2013] [Indexed: 12/11/2022] Open
Abstract
Several reports have implicated the inoculum effect that some strains of type A beta-lactamase (Bla)-producing, methicillin-susceptible Staphylococcus aureus (MSSA) show against cefazolin as the cause for clinical failures in certain serious deep-seated infections. Here, using a previously reported MSSA strain displaying this phenotype (TX0117), we obtained a Bla-cured derivative (TX0117c) with a combination of novobiocin and high temperature. Both isolates were then used in a rat endocarditis model and treated with cefazolin, nafcillin, and daptomycin, given to simulate human dosing. Animals were treated for 3 days and either sacrificed at 24 h after the last antibiotic dose (standard group) or left untreated for an additional 3 days (relapse group). With TX0117 in the standard treatment group, daptomycin and nafcillin were both significantly better than cefazolin in reducing CFU/g of vegetations, achieving mean log10 reductions compared to levels in untreated rats of 7.1, 5.3, and 1.8, respectively (cefazolin versus daptomycin, P < 0.0001; cefazolin versus nafcillin, P = 0.005; daptomycin versus nafcillin, P = 0.053). In addition, cefazolin was significantly more effective in reducing vegetation titers of TX0117c than of TX0117 (mean log10 reduction of 1.4 versus 5.5, respectively; P = 0.0001). Similar results were observed with animals in the relapse group. Thus, these data show that there can be an in vivo consequence of the in vitro inoculum effect that some MSSA strains display against cefazolin and indicate a specific role for Bla production using a Bla-cured derivative strain against which cefazolin regained both in vitro and in vivo activity.
Collapse
|
46
|
Gabriel EM, Coffey A, O'Mahony JM. Investigation into the prevalence, persistence and antibiotic resistance profiles of staphylococci isolated from euro currency. J Appl Microbiol 2013; 115:565-71. [PMID: 23679680 DOI: 10.1111/jam.12247] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 04/12/2013] [Accepted: 04/29/2013] [Indexed: 12/21/2022]
Abstract
AIMS The study set out to sample € 10 banknotes for the presence of coagulase-positive staphylococci (CPS) such as Staphylococcus aureus (S. aureus) and coagulase-negative staphylococci (CoNS) in Southern Ireland, to assess the levels of antibiotic resistance among those isolated, and determine the persistence of S. aureus on € 10 banknotes and € 2 coins. METHODS AND RESULTS We report that 97% of € 10 banknotes screened (n = 155) harboured multiple species of staphylococci. From the generated bank of strains, a total of 150 representative staphylococci isolates were used for further study, 71 were CPS and 79 were CoNS. Of these, we found that 62% of the staphylococci demonstrated resistance to at least one of the first-line antibiotics (52.11% of CPS isolates and 76.71% of the CoNS isolates). Resistance to multiple antibiotics was seen in 31.18% of the resistant isolates. In relation to persistence studies, S. aureus was shown to remain viable on euro banknotes and coins for significant periods (on average, 19.33 days on € 10 banknotes and 16.67 days on € 2 coins) as determined using bioluminescence. CONCLUSIONS We advocate the expansion of antibiotic surveillance programs, with a view to tracking/monitoring antibiotic resistance dissemination among environmental contaminants. Additionally, we propose that 'cashless transactions' should be encouraged in high-risk environments such as hospitals and healthcare settings, as well as stricter infection controls. SIGNIFICANCE AND IMPACT OF THE STUDY Although it is accepted that circulating currency has the potential to harbour disease-causing pathogens, studies investigating prevalence and persistence of such pathogens on euro currency are virtually nonexistent. In an attempt to rectify this, we examined the prevalence of staphylococci on € 10 banknotes in Ireland and reported relatively high levels of antibiotic resistance among the isolates. Furthermore, we have established the persistence of S. aureus on euro currency for the first time.
Collapse
|
47
|
Molecular basis of in vivo biofilm formation by bacterial pathogens. ACTA ACUST UNITED AC 2013; 19:1503-13. [PMID: 23261595 DOI: 10.1016/j.chembiol.2012.10.022] [Citation(s) in RCA: 253] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 10/26/2012] [Accepted: 10/30/2012] [Indexed: 12/11/2022]
Abstract
Bacterial biofilms are involved in a multitude of serious chronic infections. In recent years, modeling of biofilm infection in vitro has led to the identification of microbial determinants that govern biofilm development. However, we lack information as to whether the biofilm formation mechanisms identified in vitro have relevance for biofilm-associated infection. Here, we discuss the molecular basis of biofilm formation. Staphylococci and Pseudomonas aeruginosa are used to illustrate key points because their biofilm development process has been well studied. We focus on in vivo findings, such as obtained in animal infection models, and critically evaluate the in vivo relevance of in vitro findings. Although conflicting results about the role of quorum sensing in biofilm formation have been obtained, we argue that integration of in vitro and in vivo studies allows a differentiated view of this mechanism as it relates to biofilm infection.
Collapse
|
48
|
Prajsnar TK, Hamilton R, Garcia-Lara J, McVicker G, Williams A, Boots M, Foster SJ, Renshaw SA. A privileged intraphagocyte niche is responsible for disseminated infection of Staphylococcus aureus in a zebrafish model. Cell Microbiol 2012; 14:1600-19. [PMID: 22694745 PMCID: PMC3470706 DOI: 10.1111/j.1462-5822.2012.01826.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 06/04/2012] [Accepted: 06/07/2012] [Indexed: 01/30/2023]
Abstract
The innate immune system is the primary defence against the versatile pathogen, Staphylococcus aureus. How this organism is able to avoid immune killing and cause infections is poorly understood. Using an established larval zebrafish infection model, we have shown that overwhelming infection is due to subversion of phagocytes by staphylococci, allowing bacteria to evade killing and found foci of disease. Larval zebrafish coinfected with two S. aureus strains carrying different fluorescent reporter gene fusions (but otherwise isogenic) had bacterial lesions, at the time of host death, containing predominantly one strain. Quantitative data using two marked strains revealed that the strain ratios, during overwhelming infection, were often skewed towards the extremes, with one strain predominating. Infection with passaged bacterial clones revealed the phenomenon not to bedue to adventitious mutations acquired by the pathogen. After infection of the host, all bacteria are internalized by phagocytes and the skewing of population ratios is absolutely dependent on the presence of phagocytes. Mathematical modelling of pathogen population dynamics revealed the data patterns are consistent with the hypothesis that a small number of infected phagocytes serve as an intracellular reservoir for S. aureus, which upon release leads to disseminated infection. Strategies to specifically alter neutrophil/macrophage numbers were used to map the potential subpopulation of phagocytes acting as a pathogen reservoir, revealing neutrophils as the likely ‘niche’. Subsequently in a murine sepsis model, S. aureus abscesses in kidneys were also found to be predominantly clonal, therefore likely founded by an individual cell, suggesting a potential mechanism analogous to the zebrafish model with few protected niches. These findings add credence to the argument that S. aureus control regimes should recognize both the intracellular as well as extracellular facets of the S. aureus life cycle.
Collapse
Affiliation(s)
- Tomasz K Prajsnar
- Krebs Institute, University of Sheffield, Western Bank, Sheffield, UK
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Barman TK, Rao M, Bhati A, Kishore K, Shukla G, Kumar M, Mathur T, Pandya M, Upadhyay DJ. Non invasive real-time monitoring of bacterial infection & therapeutic effect of anti-microbials in five mouse models. Indian J Med Res 2012; 134:688-95. [PMID: 22199109 PMCID: PMC3249968 DOI: 10.4103/0971-5916.90997] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Background & objectives: In vivo imaging system has contributed significantly to the understanding of bacterial infection and efficacy of drugs in animal model. We report five rapid, reproducible, and non invasive murine pulmonary infection, skin and soft tissue infection, sepsis, and meningitis models using Xenogen bioluminescent strains and specialized in vivo imaging system (IVIS). Methods: The progression of bacterial infection in different target organs was evaluated by the photon intensity and target organ bacterial counts. Genetically engineered bioluminescent bacterial strains viz. Staphylococcus aureus Xen 8.1, 29 and 31; Streptococcus pneumoniae Xen 9 and 10 and Pseudomonas aeruginosa Xen-5 were used to induce different target organs infection and were validated with commercially available antibiotics. Results: The lower limit of detection of colony forming unit (cfu) was 1.7-log10 whereas the lower limit of detection of relative light unit (RLU) was 4.2-log10. Recovery of live bacteria from different target organs showed that the bioluminescent signal correlated to the live bacterial count. Interpretation & conclusions: This study demonstrated the real time monitoring and non-invasive analysis of progression of infection and pharmacological efficacy of drugs. These models may be useful for pre-clinical discovery of new antibiotics.
Collapse
Affiliation(s)
- Tarani Kanta Barman
- Department of Infectious Diseases, New Drug Discovery Research, Ranbaxy Research Laboratories, Gurgaon, India.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Hady WA, Bayer AS, Xiong YQ. Experimental endocarditis model of methicillin resistant Staphylococcus aureus (MRSA) in rat. J Vis Exp 2012:e3863. [PMID: 22711072 DOI: 10.3791/3863] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Endovascular infections, including endocarditis, are life-threatening infectious syndromes. Staphylococcus aureus is the most common world-wide cause of such syndromes with unacceptably high morbidity and mortality even with appropriate antimicrobial agent treatments. The increase in infections due to methicillin-resistant S. aureus (MRSA), the high rates of vancomycin clinical treatment failures and growing problems of linezolid and daptomycin resistance have all further complicated the management of patients with such infections, and led to high healthcare costs. In addition, it should be emphasized that most recent studies with antibiotic treatment outcomes have been based in clinical settings, and thus might well be influenced by host factors varying from patient-to-patient. Therefore, a relevant animal model of endovascular infection in which host factors are similar from animal-to-animal is more crucial to investigate microbial pathogenesis, as well as the efficacy of novel antimicrobial agents. Endocarditis in rat is a well-established experimental animal model that closely approximates human native valve endocarditis. This model has been used to examine the role of particular staphylococcal virulence factors and the efficacy of antibiotic treatment regimens for staphylococcal endocarditis. In this report, we describe the experimental endocarditis model due to MRSA that could be used to investigate bacterial pathogenesis and response to antibiotic treatment.
Collapse
Affiliation(s)
- Wessam Abdel Hady
- Department of Medicine, Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, USA
| | | | | |
Collapse
|