51
|
Visualization of the role of host heme on the virulence of the heme auxotroph Streptococcus agalactiae. Sci Rep 2017; 7:40435. [PMID: 28091535 PMCID: PMC5238366 DOI: 10.1038/srep40435] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 12/06/2016] [Indexed: 12/21/2022] Open
Abstract
Heme is essential for several cellular key functions but is also toxic. Whereas most bacterial pathogens utilize heme as a metabolic cofactor and iron source, the impact of host heme during bacterial infection remains elusive. The opportunist pathogen Streptococcus agalactiae does not synthesize heme but still uses it to activate a respiration metabolism. Concomitantly, heme toxicity is mainly controlled by the HrtBA efflux transporter. Here we investigate how S. agalactiae manages heme toxicity versus benefits in the living host. Using bioluminescent bacteria and heme-responsive reporters for in vivo imaging, we show that the capacity of S. agalactiae to overcome heme toxicity is required for successful infection, particularly in blood-rich organs. Host heme is simultaneously required, as visualized by a generalized infection defect of a respiration-negative mutant. In S. agalactiae, HrtBA expression responds to an intracellular heme signal via activation of the two-component system HssRS. A hssRS promoter-driven intracellular luminescent heme sensor was designed to identify host compartments that supply S. agalactiae with heme. S. agalactiae acquires heme in heart, kidneys, and liver, but not in the brain. We conclude that S. agalactiae response to heme is organ-dependent, and its efflux may be particularly relevant in late stages of infection.
Collapse
|
52
|
Robertson J, Dalton J, Wiles S, Gizdavic-Nikolaidis M, Swift S. The tuberculocidal activity of polyaniline and functionalised polyanilines. PeerJ 2016; 4:e2795. [PMID: 28028468 PMCID: PMC5178338 DOI: 10.7717/peerj.2795] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/15/2016] [Indexed: 12/17/2022] Open
Abstract
Tuberculosis is considered a leading cause of death worldwide. More than 95% of cases and deaths occur in low- and middle-income countries. In resource-limited countries, hospitals often lack adequate facilities to manage and isolate patients with infectious tuberculosis (TB), relying instead on personal protective equipment, such as facemasks, to reduce nosocomial transmission of the disease. Facemasks impregnated with an antimicrobial agent may be a cost-effective way of adding an extra level of protection against the spread of TB by reducing the risk of disease transmission. Conducting polymers, such as polyaniline (PANI), and their functionalised derivatives are a novel class of antimicrobial agents with potential as non-leaching additives to provide contamination resistant surfaces. We have investigated the antimicrobial action of PANI and a functionalised derivative, poly-3-aminobenzoic acid (P3ABA), against mycobacteria and have determined the optimal treatment time and concentration to achieve significant knockdown of Mycobacterium smegmatis and Mycobacterium tuberculosis on an agar surface. Results indicated that P3ABA is a potential candidate for use as an anti-tuberculoid agent in facemasks to reduce TB transmission.
Collapse
Affiliation(s)
- Julia Robertson
- Department of Molecular Medicine and Pathology, University of Auckland , Auckland , New Zealand
| | - James Dalton
- Department of Molecular Medicine and Pathology, University of Auckland , Auckland , New Zealand
| | - Siouxsie Wiles
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; Bioluminescent Superbugs Lab, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | | | - Simon Swift
- Department of Molecular Medicine and Pathology, University of Auckland , Auckland , New Zealand
| |
Collapse
|
53
|
Davis RW, Eggleston H, Johnson F, Nahrendorf M, Bock PE, Peterson T, Panizzi P. In Vivo Tracking of Streptococcal Infections of Subcutaneous Origin in a Murine Model. Mol Imaging Biol 2016; 17:793-801. [PMID: 25921659 DOI: 10.1007/s11307-015-0856-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
PURPOSE Generation of plasmin in vivo by Streptococcus pyogenes is thought to localize the active protease complexes to the pathogen surface to aid in tissue dissemination. Here, we chose to follow cutaneous streptococcal infections by the use of non-invasive bioluminescence imaging to determine if this pathogen can be followed by this approach and the extent of bacterial spread in the absence of canonical plasminogen activation by streptokinase. PROCEDURES Mice were injected subcutaneously with either bioluminescent strains of streptococci, namely Xen20 and Xen10 or S. pyogenes ALAB49. Bioluminescence imaging was performed daily and results were correlated with microbiological and histological analyses. RESULTS Comparative analysis of chronologic non-invasive datasets indicated that Xen20 did not disseminate from the initial infection site. Contrary to this, microbiological and histological analyses of Xen20 mice for total bacterial burden indicated sepsis and widespread pathogen involvement. CONCLUSIONS The use of bioluminescence in microbe-based studies requires genomic and pathologic characterization to correlate imaging results with underlying pathology.
Collapse
Affiliation(s)
- Richard W Davis
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, 4306 Walker Building, Auburn, AL, 36849, USA
| | - Heather Eggleston
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, 4306 Walker Building, Auburn, AL, 36849, USA
| | - Frances Johnson
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, 4306 Walker Building, Auburn, AL, 36849, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge St., Boston, MA, 02114, USA
| | - Paul E Bock
- Department of Pathology, Microbiology, & Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Tiffany Peterson
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, 4306 Walker Building, Auburn, AL, 36849, USA
| | - Peter Panizzi
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, 4306 Walker Building, Auburn, AL, 36849, USA.
| |
Collapse
|
54
|
Abstract
Immunomics is a relatively new field of research which integrates the disciplines of immunology, genomics, proteomics, transcriptomics and bioinformatics to characterize the host-pathogen interface. Herein, we discuss how rapid advances in molecular immunology, sophisticated tools and molecular databases are facilitating in-depth exploration of the immunome. In our opinion, an immunomics-based approach presides over traditional antigen and vaccine discovery methods that have proved ineffective for highly complex pathogens such as the causative agents of malaria, tuberculosis and schistosomiasis that have evolved genetic and immunological host-parasite adaptations over time. By using an integrative multidisciplinary approach, immunomics offers enormous potential to advance 21st century antigen discovery and rational vaccine design against complex pathogens such as the Plasmodium parasite.
Collapse
|
55
|
Read HM, Mills G, Johnson S, Tsai P, Dalton J, Barquist L, Print CG, Patrick WM, Wiles S. The in vitro and in vivo effects of constitutive light expression on a bioluminescent strain of the mouse enteropathogen Citrobacter rodentium. PeerJ 2016; 4:e2130. [PMID: 27366640 PMCID: PMC4924136 DOI: 10.7717/peerj.2130] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 05/24/2016] [Indexed: 11/30/2022] Open
Abstract
Bioluminescent reporter genes, such as those from fireflies and bacteria, let researchers use light production as a non-invasive and non-destructive surrogate measure of microbial numbers in a wide variety of environments. As bioluminescence needs microbial metabolites, tagging microorganisms with luciferases means only live metabolically active cells are detected. Despite the wide use of bioluminescent reporter genes, very little is known about the impact of continuous (also called constitutive) light expression on tagged bacteria. We have previously made a bioluminescent strain of Citrobacter rodentium, a bacterium which infects laboratory mice in a similar way to how enteropathogenic Escherichia coli (EPEC) and enterohaemorrhagic E. coli (EHEC) infect humans. In this study, we compared the growth of the bioluminescent C. rodentium strain ICC180 with its non-bioluminescent parent (strain ICC169) in a wide variety of environments. To understand more about the metabolic burden of expressing light, we also compared the growth profiles of the two strains under approximately 2,000 different conditions. We found that constitutive light expression in ICC180 was near-neutral in almost every non-toxic environment tested. However, we also found that the non-bioluminescent parent strain has a competitive advantage over ICC180 during infection of adult mice, although this was not enough for ICC180 to be completely outcompeted. In conclusion, our data suggest that constitutive light expression is not metabolically costly to C. rodentium and supports the view that bioluminescent versions of microbes can be used as a substitute for their non-bioluminescent parents to study bacterial behaviour in a wide variety of environments.
Collapse
Affiliation(s)
- Hannah M Read
- Bioluminescent Superbugs Lab, University of Auckland, Auckland, New Zealand; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Grant Mills
- Bioluminescent Superbugs Lab, University of Auckland, Auckland, New Zealand; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Sarah Johnson
- Bioluminescent Superbugs Lab, University of Auckland, Auckland, New Zealand; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Peter Tsai
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; Bioinformatics Institute, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - James Dalton
- Bioluminescent Superbugs Lab, University of Auckland, Auckland, New Zealand; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand
| | - Lars Barquist
- Institute for Molecular Infection Biology, University of Würzburg , Würzburg , Germany
| | - Cristin G Print
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; Bioinformatics Institute, School of Biological Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand
| | - Wayne M Patrick
- Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand; Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Siouxsie Wiles
- Bioluminescent Superbugs Lab, University of Auckland, Auckland, New Zealand; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand
| |
Collapse
|
56
|
Burrell-Saward H, Ward TH. Bioluminescence Imaging to Detect Late Stage Infection of African Trypanosomiasis. JOURNAL OF VISUALIZED EXPERIMENTS : JOVE 2016. [PMID: 27284970 DOI: 10.3791/54032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Human African trypanosomiasis (HAT) is a multi-stage disease that manifests in two stages; an early blood stage and a late stage when the parasite invades the central nervous system (CNS). In vivo study of the late stage has been limited as traditional methodologies require the removal of the brain to determine the presence of the parasites. Bioluminescence imaging is a non-invasive, highly sensitive form of optical imaging that enables the visualization of a luciferase-transfected pathogen in real-time. By using a transfected trypanosome strain that has the ability to produce late stage disease in mice we are able to study the kinetics of a CNS infection in a single animal throughout the course of infection, as well as observe the movement and dissemination of a systemic infection. Here we describe a robust protocol to study CNS infections using a bioluminescence model of African trypanosomiasis, providing real time non-invasive observations which can be further analyzed with optional downstream approaches.
Collapse
Affiliation(s)
| | - Theresa H Ward
- Department of Infection and Immunity, London School of Hygiene & Tropical Diseases;
| |
Collapse
|
57
|
Kong Y, Yang D, Cirillo SLG, Li S, Akin A, Francis KP, Maloney T, Cirillo JD. Application of Fluorescent Protein Expressing Strains to Evaluation of Anti-Tuberculosis Therapeutic Efficacy In Vitro and In Vivo. PLoS One 2016; 11:e0149972. [PMID: 26934495 PMCID: PMC4774912 DOI: 10.1371/journal.pone.0149972] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/08/2016] [Indexed: 12/11/2022] Open
Abstract
The slow growth of Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), hinders development of new diagnostics, therapeutics and vaccines. Using non-invasive real-time imaging technologies to monitor the disease process in live animals would facilitate TB research in all areas. We developed fluorescent protein (FP) expressing Mycobacterium bovis BCG strains for in vivo imaging, which can be used to track bacterial location, and to quantify bacterial load in live animals. We selected an optimal FP for in vivo imaging, by first cloning six FPs: tdTomato, mCherry, mPlum, mKate, Katushka and mKeima, into mycobacteria under either a mycobacterial Hsp60 or L5 promoter, and compared their fluorescent signals in vitro and in vivo. Fluorescence from each FP-expressing strain was measured with a multimode reader using the optimal excitation and emission wavelengths for the FP. After normalizing bacterial numbers with optical density, the strain expressing L5-tdTomato displayed the highest fluorescence. We used the tdTomato-labeled M. bovis BCG to obtain real-time images of pulmonary infections in living mice and rapidly determined the number of bacteria present. Further comparison between L5-tdTomato and Hsp60-tdTomato revealed that L5-tdTomato carried four-fold more tdTomato gene copies than Hsp60-tdTomato, which eventually led to higher protein expression of tdTomato. Evaluating anti-TB efficacy of rifampicin and isoniazid therapy in vitro and in vivo using the L5-tdTomato strain demonstrated that this strain can be used to identify anti-TB therapeutic efficacy as quickly as 24 h post-treatment. These M. bovis BCG reporter strains represent a valuable new tool for evaluation of therapeutics, vaccines and virulence.
Collapse
Affiliation(s)
- Ying Kong
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Dong Yang
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Suat L. G. Cirillo
- Department of Microbial Pathogenesis and Immunology, Texas A & M Health Science Center, Bryan, Texas, United States of America
| | - Shaoji Li
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Ali Akin
- Caliper Life Sciences, PerkinElmer, Waltham, Massachusetts, United States of America
| | - Kevin P. Francis
- Caliper Life Sciences, PerkinElmer, Waltham, Massachusetts, United States of America
| | - Taylor Maloney
- Department of Microbial Pathogenesis and Immunology, Texas A & M Health Science Center, Bryan, Texas, United States of America
| | - Jeffrey D. Cirillo
- Department of Microbial Pathogenesis and Immunology, Texas A & M Health Science Center, Bryan, Texas, United States of America
| |
Collapse
|
58
|
Nooshabadi F, Yang HJ, Bixler JN, Kong Y, Cirillo JD, Maitland KC. Intravital Fluorescence Excitation in Whole-Animal Optical Imaging. PLoS One 2016; 11:e0149932. [PMID: 26901051 PMCID: PMC4762773 DOI: 10.1371/journal.pone.0149932] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 02/06/2016] [Indexed: 12/22/2022] Open
Abstract
Whole-animal fluorescence imaging with recombinant or fluorescently-tagged pathogens or cells enables real-time analysis of disease progression and treatment response in live animals. Tissue absorption limits penetration of fluorescence excitation light, particularly in the visible wavelength range, resulting in reduced sensitivity to deep targets. Here, we demonstrate the use of an optical fiber bundle to deliver light into the mouse lung to excite fluorescent bacteria, circumventing tissue absorption of excitation light in whole-animal imaging. We present the use of this technology to improve detection of recombinant reporter strains of tdTomato-expressing Mycobacterium bovis BCG (Bacillus Calmette Guerin) bacteria in the mouse lung. A microendoscope was integrated into a whole-animal fluorescence imager to enable intravital excitation in the mouse lung with whole-animal detection. Using this technique, the threshold of detection was measured as 103 colony forming units (CFU) during pulmonary infection. In comparison, the threshold of detection for whole-animal fluorescence imaging using standard epi-illumination was greater than 106 CFU.
Collapse
Affiliation(s)
- Fatemeh Nooshabadi
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, United States of America
| | - Hee-Jeong Yang
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, United States of America
| | - Joel N. Bixler
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, United States of America
| | - Ying Kong
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, United States of America
| | - Jeffrey D. Cirillo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, United States of America
| | - Kristen C. Maitland
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, United States of America
- * E-mail:
| |
Collapse
|
59
|
Merritt J, Senpuku H, Kreth J. Let there be bioluminescence: development of a biophotonic imaging platform for in situ analyses of oral biofilms in animal models. Environ Microbiol 2016; 18:174-90. [PMID: 26119252 PMCID: PMC5050008 DOI: 10.1111/1462-2920.12953] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 06/09/2015] [Accepted: 06/10/2015] [Indexed: 12/23/2022]
Abstract
In the current study, we describe a novel biophotonic imaging-based reporter system that is particularly useful for the study of virulence in polymicrobial infections and interspecies interactions within animal models. A suite of luciferase enzymes was compared using three early colonizing species of the human oral flora (Streptococcus mutans, Streptococcus gordonii and Streptococcus sanguinis) to determine the utility of the different reporters for multiplexed imaging studies in vivo. Using the multiplex approach, we were able to track individual species within a dual-species oral infection model in mice with both temporal and spatial resolution. We also demonstrate how biophotonic imaging of multiplexed luciferase reporters could be adapted for real-time quantification of bacterial gene expression in situ. By creating an inducible dual-luciferase expressing reporter strain of S. mutans, we were able to exogenously control and measure expression of nlmAB (encoding the bacteriocin mutacin IV) within mice to assess its importance for the persistence ability of S. mutans in the oral cavity. The imaging system described in the current study circumvents many of the inherent limitations of current animal model systems, which should now make it feasible to test hypotheses that were previously impractical to model.
Collapse
Affiliation(s)
- Justin Merritt
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- College of Dentistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Hidenobu Senpuku
- Department of Bacteriology I, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Jens Kreth
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- College of Dentistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
60
|
Lin Q, Huang H, Chen J, Zheng G. Using Fluorescence Imaging to Track Drug Delivery and Guide Treatment Planning In Vivo. Methods Mol Biol 2016; 1444:153-166. [PMID: 27283425 DOI: 10.1007/978-1-4939-3721-9_14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Imaging has become an indispensable tool in both clinical medicine and preclinical sciences. It enables doctors to locate sites of cancer/disease, track drug delivery, and guide operative planning, thus enhancing the treatment efficacy. Recently, we developed a multimodal theranostic lipid nanoparticles, named HPPS(NIR)-chol-siRNA with its built-in near-infrared (NIR) fluorescent probe core as a useful surrogate for tracking small interfering RNA (siRNA) delivery. By using the image co-registration of computed tomography (CT) and fluorescence molecular tomography (FMT), we achieved noninvasive assessment and treatment planning of siRNA delivery into the orthotopic tumor, thus enabling efficacious RNA interference (RNAi) therapy. In this chapter, we introduce this method to illustrate the use of CT-FMT co-registration for tracking drug delivery and guiding treatment planning in vivo.
Collapse
Affiliation(s)
- Qiaoya Lin
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China
| | - Huang Huang
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
- DLVR Therapeutics Inc., Toronto, ON, Canada
| | - Juan Chen
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada.
| | - Gang Zheng
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
61
|
A stable luciferase reporter plasmid for in vivo imaging in murine models of Staphylococcus aureus infections. Appl Microbiol Biotechnol 2015; 100:3197-206. [PMID: 26685857 DOI: 10.1007/s00253-015-7229-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 12/03/2015] [Accepted: 12/05/2015] [Indexed: 11/27/2022]
Abstract
In vivo imaging of bioluminescent bacteria permits their visualization in infected mice, allowing spatial and temporal evaluation of infection progression. Most available bioluminescent strains were obtained by integration of the luciferase genes into the bacterial chromosome, a challenging and time-consuming approach. Recently, episomal plasmids were used, which were introduced in bacteria and expressed all genes required for bioluminescence emission. However, the plasmid was progressively lost in vitro and in vivo, if bacteria were not maintained under antibiotic selective pressure. Increased stability could be obtained inserting into the plasmid backbone sequences that assured plasmid partition between daughter bacterial cells, or caused death of bacteria that had lost the plasmid. So far, no detailed analysis was performed of either plasmid stability in vivo or contribution of different stabilizing sequence types. Here we report the construction of a plasmid, which includes the Photorhabdus luminescens lux cassette expressed under the control of a Staphylococcus aureus specific gene promoter, and toxin/antitoxin (T/A) and partition sequences (Par) conferring stability and transmissibility of the plasmid. Following infection of mice with S. aureus carrying this plasmid, we demonstrated that the promoter-lux fusion was functional in vivo, that the plasmid was retained by 70-100% of bacterial cells 7 days post-infection, and that both stabilizing sequence types were required to maximize plasmid retention. These data suggest that the plasmid can be a valuable tool to study gene expression and bacterial spread in small laboratory animals infected with S. aureus or possibly other Gram-positive human pathogens.
Collapse
|
62
|
van Zyl WF, Deane SM, Dicks LMT. Reporter systems for in vivo tracking of lactic acid bacteria in animal model studies. Gut Microbes 2015; 6:291-9. [PMID: 26516656 PMCID: PMC4826117 DOI: 10.1080/19490976.2015.1086058] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Bioluminescence (BLI) and fluorescence imaging (FI) allow for non-invasive detection of viable microorganisms from within living tissue and are thus ideally suited for in vivo probiotic studies. Highly sensitive optical imaging techniques detect signals from the excitation of fluorescent proteins, or luciferase-catalyzed oxidation reactions. The excellent relation between microbial numbers and photon emission allow for quantification of tagged bacteria in vivo with extreme accuracy. More information is gained over a shorter period compared to traditional pre-clinical animal studies. The review summarizes the latest advances in in vivo bioluminescence and fluorescence imaging and points out the advantages and limitations of different techniques. The practical application of BLI and FI in the tracking of lactic acid bacteria in animal models is addressed.
Collapse
Affiliation(s)
- Winschau F van Zyl
- Department of Microbiology; Stellenbosch University; Matieland, Stellenbosch, South Africa
| | - Shelly M Deane
- Department of Microbiology; Stellenbosch University; Matieland, Stellenbosch, South Africa
| | - Leon M T Dicks
- Department of Microbiology; Stellenbosch University; Matieland, Stellenbosch, South Africa,Correspondence to: Leon M T Dicks;
| |
Collapse
|
63
|
van Zyl WF, Deane SM, Dicks LMT. Use of the mCherry Fluorescent Protein To Study Intestinal Colonization by Enterococcus mundtii ST4SA and Lactobacillus plantarum 423 in Mice. Appl Environ Microbiol 2015; 81:5993-6002. [PMID: 26116681 PMCID: PMC4551250 DOI: 10.1128/aem.01247-15] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 05/22/2015] [Indexed: 12/19/2022] Open
Abstract
Lactic acid bacteria (LAB) are natural inhabitants of the gastrointestinal tract (GIT) of humans and animals, and some LAB species receive considerable attention due to their health benefits. Although many papers have been published on probiotic LAB, only a few reports have been published on the migration and colonization of the cells in the GIT. This is due mostly to the lack of efficient reporter systems. In this study, we report on the application of the fluorescent mCherry protein in the in vivo tagging of the probiotic strains Enterococcus mundtii ST4SA and Lactobacillus plantarum 423. The mCherry gene, encoding a red fluorescent protein (RFP), was integrated into a nonfunctional region on the genome of L. plantarum 423 by homologous recombination. In the case of E. mundtii ST4SA, the mCherry gene was cloned into the pGKV223D LAB/Escherichia coli expression vector. Expression of the mCherry gene did not alter the growth rate of the two strains and had no effect on bacteriocin production. Both strains colonized the cecum and colon of mice.
Collapse
Affiliation(s)
- Winschau F van Zyl
- Department of Microbiology, University of Stellenbosch, Stellenbosch, South Africa
| | - Shelly M Deane
- Department of Microbiology, University of Stellenbosch, Stellenbosch, South Africa
| | - Leon M T Dicks
- Department of Microbiology, University of Stellenbosch, Stellenbosch, South Africa
| |
Collapse
|
64
|
Abstract
Infectious murine models greatly benefit from optical imaging using bioluminescent bacteria to non-invasively and repeatedly follow in vivo bacterial infection. In this context, one of the most critical parameters is the bioluminescence sensitivity to reliably detect the smallest number of bacteria. Another critical point is the anesthetic approaches that have been demonstrated to impact the bioluminescence flux emission in studies with luciferase-transfected tumor cells. However, this impact has never been assessed on bacteria bioluminescent models. To this end, we investigated the effects of four anesthesia protocols on the bioluminescence flux in a central venous catheter murine model (SKH1-hr(hr) mice) infected by a bioluminescent S. aureus Xen36 strain. Bioluminescence imaging was performed on mice anesthetized by either ketamine/xylazine (with or without oxygen supplementation), or isoflurane carried with air or oxygen. Total flux emission was determined in vivo daily for 3 days and ex vivo at the end of the study together with a CFU counting of the biofilm in the catheter. Bioluminescence flux differences appear between the different anesthetic protocols. Using a ketamine/xylazine anesthesia (with air), bacteria detection was impossible since the bioluminescence signal remains in the background signal. Mice anesthetized with isoflurane and oxygen led to a signal significantly higher to the background all along the kinetics. The use of isoflurane in air presents a bioluminescence signal similar to the use of ketamine/xylazine with oxygen. These data highlight the importance of oxygen to improve bioluminescence flux by bacteria with isoflurane as well as with ketamine/xylazine anesthetics. As a conclusion, we recommend the use of isoflurane anesthetic with oxygen to increase the bioluminescence sensitivity in this kind of study.
Collapse
|
65
|
Dual-Color Bioluminescence Imaging for Simultaneous Monitoring of the Intestinal Persistence of Lactobacillus plantarum and Lactococcus lactis in Living Mice. Appl Environ Microbiol 2015; 81:5344-9. [PMID: 26025906 DOI: 10.1128/aem.01042-15] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/22/2015] [Indexed: 12/17/2022] Open
Abstract
Lactic acid bacteria are found in the gastrointestinal tract of mammals and have received tremendous attention due to their health-promoting properties. We report the development of two dual-color luciferase-producing Lactobacillus (Lb.) plantarum and Lactococcus (Lc.) lactis strains for noninvasive simultaneous tracking in the mouse gastrointestinal tract. We previously described the functional expression of the red luciferase mutant (CBRluc) from Pyrophorus plagiophthalamus in Lb. plantarum NCIMB8826 and Lc. lactis MG1363 (C. Daniel, S. Poiret, V. Dennin, D. Boutillier, and B. Pot, Appl Environ Microbiol 79:1086-1094, 2013, http://dx.doi.org/10.1128/AEM.03221-12). In this study, we determined that CBRluc is a better-performing luciferase for in vivo localization of both lactic acid bacteria after oral administration than the green click beetle luciferase mutant construct developed in this study. We further established the possibility to simultaneously detect red- and green-emitting lactic acid bacteria by dual-wavelength bioluminescence imaging in combination with spectral unmixing. The difference in spectra of light emission by the red and green click beetle luciferase mutants and dual bioluminescence detection allowed in vitro and in vivo quantification of the red and green emitted signals; thus, it allowed us to monitor the dynamics and fate of the two bacterial populations simultaneously. Persistence and viability of both strains simultaneously administered to mice in different ratios was studied in vivo in anesthetized mice and ex vivo in mouse feces. The application of dual-luciferase-labeled bacteria has considerable potential to simultaneously study the interactions and potential competitions of different targeted bacteria and their hosts.
Collapse
|
66
|
Anderson E, Salisbury V. Rapid in-vitro testing for chemotherapy sensitivity in leukaemia patients. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2015; 145:189-214. [PMID: 25216956 DOI: 10.1007/978-3-662-43619-6_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Bioluminescent bacterial biosensors can be used in a rapid in vitro assay to predict sensitivity to commonly used chemotherapy drugs in acute myeloid leukemia (AML). The nucleoside analog cytarabine (ara-C) is the key agent for treating AML; however, up to 30 % of patients fail to respond to treatment. Screening of patient blood samples to determine drug response before commencement of treatment is needed. To achieve this aim, a self-bioluminescent reporter strain of Escherichia coli has been constructed and evaluated for use as an ara-C biosensor and an in vitro assay has been designed to predict ara-C response in clinical samples. Transposition mutagenesis was used to create a cytidine deaminase (cdd)-deficient mutant of E. coli MG1655 that responded to ara-C. The strain was transformed with the luxCDABE operon and used as a whole-cell biosensor for development an 8-h assay to determine ara-C uptake and phosphorylation by leukemic cells. Intracellular concentrations of 0.025 μmol/L phosphorylated ara-C were detected by significantly increased light output (P < 0.05) from the bacterial biosensor. Results using AML cell lines with known response to ara-C showed close correlation between the 8-h assay and a 3-day cytotoxicity test for ara-C cell killing. In retrospective tests with 24 clinical samples of bone marrow or peripheral blood, the biosensor-based assay predicted leukemic cell response to ara-C within 8 h. The biosensor-based assay may offer a predictor for evaluating the sensitivity of leukemic cells to ara-C before patients undergo chemotherapy and allow customized treatment of drug-sensitive patients with reduced ara-C dose levels. The 8-h assay monitors intracellular ara-CTP (cytosine arabinoside triphosphate) levels and, if fully validated, may be suitable for use in clinical settings.
Collapse
MESH Headings
- Antimetabolites, Antineoplastic/metabolism
- Antimetabolites, Antineoplastic/pharmacology
- Arabinofuranosylcytosine Triphosphate/analysis
- Arabinofuranosylcytosine Triphosphate/metabolism
- Biological Assay
- Biosensing Techniques
- Cell Line, Tumor
- Cytarabine/metabolism
- Cytarabine/pharmacology
- Drug Screening Assays, Antitumor
- Escherichia coli/genetics
- Escherichia coli/metabolism
- Genes, Reporter
- Genetic Engineering
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Limit of Detection
- Luciferases, Bacterial/genetics
- Luciferases, Bacterial/metabolism
- Luminescent Measurements
- Operon
- Phosphorylation
Collapse
Affiliation(s)
- Elizabeth Anderson
- Institute of Bio-Sensing Technology, University of the West of England, Bristol, UK
| | | |
Collapse
|
67
|
Forestier CL, Späth GF, Prina E, Dasari S. Simultaneous multi-parametric analysis of Leishmania and of its hosting mammal cells: A high content imaging-based method enabling sound drug discovery process. Microb Pathog 2014; 88:103-8. [PMID: 25448129 DOI: 10.1016/j.micpath.2014.10.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 08/18/2014] [Accepted: 10/21/2014] [Indexed: 11/18/2022]
Abstract
Leishmaniasis is a vector-borne disease for which only limited therapeutic options are available. The disease is ranked among the six most important tropical infectious diseases and represents the second-largest parasitic killer in the world. The development of new therapies has been hampered by the lack of technologies and methodologies that can be integrated into the complex physiological environment of a cell or organism and adapted to suitable in vitro and in vivo Leishmania models. Recent advances in microscopy imaging offer the possibility to assess the efficacy of potential drug candidates against Leishmania within host cells. This technology allows the simultaneous visualization of relevant phenotypes in parasite and host cells and the quantification of a variety of cellular events. In this review, we present the powerful cellular imaging methodologies that have been developed for drug screening in a biologically relevant context, addressing both high-content and high-throughput needs. Furthermore, we discuss the potential of intra-vital microscopy imaging in the context of the anti-leishmanial drug discovery process.
Collapse
Affiliation(s)
- Claire-Lise Forestier
- INSERM U1095, URMITE-UMR CNRS 7278, Infectiopole Sud, University of Aix-Marseille, Marseille, France.
| | - Gerald Frank Späth
- Institut Pasteur and CNRS URA2581, Unité de Parasitologie moléculaire et Signalisation, Paris, France
| | - Eric Prina
- Institut Pasteur and CNRS URA2581, Unité de Parasitologie moléculaire et Signalisation, Paris, France
| | - Sreekanth Dasari
- INSERM U1095, URMITE-UMR CNRS 7278, Infectiopole Sud, University of Aix-Marseille, Marseille, France
| |
Collapse
|
68
|
Jouvion G, Brock M, Droin-Bergere S, Ibrahim-Granet O. Duality of liver and kidney lesions after systemic infection of immunosuppressed and immunocompetent mice withAspergillus fumigatus. Virulence 2014; 3:43-50. [DOI: 10.4161/viru.3.1.18654] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
69
|
Lewis MD, Francisco AF, Taylor MC, Kelly JM. A new experimental model for assessing drug efficacy against Trypanosoma cruzi infection based on highly sensitive in vivo imaging. ACTA ACUST UNITED AC 2014; 20:36-43. [PMID: 25296657 PMCID: PMC4361455 DOI: 10.1177/1087057114552623] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The protozoan Trypanosoma cruzi is the causative agent of Chagas disease, one of the world’s major neglected infections. Although development of improved antiparasitic drugs is considered a priority, there have been no significant treatment advances in the past 40 years. Factors that have limited progress include an incomplete understanding of pathogenesis, tissue tropism, and disease progression. In addition, in vivo models, which allow parasite burdens to be tracked throughout the chronic stage of infection, have been lacking. To address these issues, we have developed a highly sensitive in vivo imaging system based on bioluminescent T. cruzi, which express a red-shifted luciferase that emits light in the tissue-penetrating orange-red region of the spectrum. The exquisite sensitivity of this noninvasive murine model has been exploited to monitor parasite burden in real time throughout the chronic stage, has allowed the identification of the gastrointestinal tract as the major niche of long-term infection, and has demonstrated that chagasic heart disease can develop in the absence of locally persistent parasites. Here, we review the parameters of the imaging system and describe how this experimental model can be incorporated into drug development programs as a valuable tool for assessing efficacy against both acute and chronic T. cruzi infections.
Collapse
Affiliation(s)
- Michael D Lewis
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Amanda Fortes Francisco
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Martin C Taylor
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - John M Kelly
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| |
Collapse
|
70
|
Real-time bioluminescence imaging of mixed mycobacterial infections. PLoS One 2014; 9:e108341. [PMID: 25265287 PMCID: PMC4180448 DOI: 10.1371/journal.pone.0108341] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 08/28/2014] [Indexed: 01/18/2023] Open
Abstract
Molecular analysis of infectious processes in bacteria normally involves construction of isogenic mutants that can then be compared to wild type in an animal model. Pathogenesis and antimicrobial studies are complicated by variability between animals and the need to sacrifice individual animals at specific time points. Live animal imaging allows real-time analysis of infections without the need to sacrifice animals, allowing quantitative data to be collected at multiple time points in all organs simultaneously. However, imaging has not previously allowed simultaneous imaging of both mutant and wild type strains of mycobacteria in the same animal. We address this problem by using both firefly (Photinus pyralis) and click beetle (Pyrophorus plagiophthalamus) red luciferases, which emit distinct bioluminescent spectra, allowing simultaneous imaging of two different mycobacterial strains during infection. We also demonstrate that these same bioluminescence reporters can be used to evaluate therapeutic efficacy in real-time, greatly facilitating our ability to screen novel antibiotics as they are developed. Due to the slow growth rate of mycobacteria, novel imaging technologies are a pressing need, since they can they can impact the rate of development of new therapeutics as well as improving our understanding of virulence mechanisms and the evaluation of novel vaccine candidates.
Collapse
|
71
|
The impact of "omic" and imaging technologies on assessing the host immune response to biodefence agents. J Immunol Res 2014; 2014:237043. [PMID: 25333059 PMCID: PMC4182007 DOI: 10.1155/2014/237043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 07/23/2014] [Accepted: 08/05/2014] [Indexed: 01/08/2023] Open
Abstract
Understanding the interactions between host and pathogen is important for the development and assessment of medical countermeasures to infectious agents, including potential biodefence pathogens such as Bacillus anthracis, Ebola virus, and Francisella tularensis. This review focuses on technological advances which allow this interaction to be studied in much greater detail. Namely, the use of “omic” technologies (next generation sequencing, DNA, and protein microarrays) for dissecting the underlying host response to infection at the molecular level; optical imaging techniques (flow cytometry and fluorescence microscopy) for assessing cellular responses to infection; and biophotonic imaging for visualising the infectious disease process. All of these technologies hold great promise for important breakthroughs in the rational development of vaccines and therapeutics for biodefence agents.
Collapse
|
72
|
Van Reet N, Van de Vyver H, Pyana PP, Van der Linden AM, Büscher P. A panel of Trypanosoma brucei strains tagged with blue and red-shifted luciferases for bioluminescent imaging in murine infection models. PLoS Negl Trop Dis 2014; 8:e3054. [PMID: 25144573 PMCID: PMC4140678 DOI: 10.1371/journal.pntd.0003054] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 06/17/2014] [Indexed: 11/19/2022] Open
Abstract
Background Genetic engineering with luciferase reporter genes allows monitoring Trypanosoma brucei (T.b.) infections in mice by in vivo bioluminescence imaging (BLI). Until recently, luminescent T.b. models were based on Renilla luciferase (RLuc) activity. Our study aimed at evaluating red-shifted luciferases for in vivo BLI in a set of diverse T.b. strains of all three subspecies, including some recently isolated from human patients. Methodology/Principal findings We transfected T.b. brucei, T.b. rhodesiense and T.b. gambiense strains with either RLuc, click beetle red (CBR) or Photinus pyralis RE9 (PpyRE9) luciferase and characterised their in vitro luciferase activity, growth profile and drug sensitivity, and their potential for in vivo BLI. Compared to RLuc, the red-shifted luciferases, CBR and PpyRE9, allow tracking of T.b. brucei AnTaR 1 trypanosomes with higher details on tissue distribution, and PpyRE9 allows detection of the parasites with a sensitivity of at least one order of magnitude higher than CBR luciferase. With CBR-tagged T.b. gambiense LiTaR1, T.b. rhodesiense RUMPHI and T.b. gambiense 348 BT in an acute, subacute and chronic infection model respectively, we observed differences in parasite tropism for murine tissues during in vivo BLI. Ex vivo BLI on the brain confirmed central nervous system infection by all luminescent strains of T.b. brucei AnTaR 1, T.b. rhodesiense RUMPHI and T.b. gambiense 348 BT. Conclusions/Significance We established a genetically and phenotypically diverse collection of bioluminescent T.b. brucei, T.b. gambiense and T.b. rhodesiense strains, including drug resistant strains. For in vivo BLI monitoring of murine infections, we recommend trypanosome strains transfected with red-shifted luciferase reporter genes, such as CBR and PpyRE9. Red-shifted luciferases can be detected with a higher sensitivity in vivo and at the same time they improve the spatial resolution of the parasites in the entire body due to the better kinetics of their substrate D-luciferin. Research on African trypanosomes heavily relies on rodent infection models. One way to reduce the number of laboratory rodents used in each experiment and effectively follow the progression of the infection in the same animals is to use genetically modified trypanosomes that allow monitoring of the infection over time with bioluminescence technology, without having to sacrifice the animals at multiple time points. In this study, we were able to establish a collection of bioluminescent strains of all three subspecies of Trypanosoma brucei (T.b.), including T.b. gambiense and T.b. rhodesiense that cause human African trypanosomiasis (HAT) or sleeping sickness. Making use of bioluminescence assays, we demonstrate the diversity of our collection in terms of in vitro and in vivo growth, drug sensitivity and in vivo parasite distribution, including central nervous system tropism. Growth characteristics and drug sensitivity are not affected by the genetic modification with luciferase reporter genes. Trypanosome strains transfected with red-shifted luciferase reporter genes have several advantages compared to the corresponding blue luciferase modified strains. Red light is less absorbed in the blood than blue light, which should lead to higher sensitivity of detection. Furthermore, the substrates that drive the light reaction are better distributed through the body for the red luciferase than for the blue luciferase, which greatly improves spatial resolution of the infection.
Collapse
Affiliation(s)
- Nick Van Reet
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
- * E-mail:
| | - Hélène Van de Vyver
- Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Patient Pati Pyana
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
- Département de Parasitologie, Institut National de Recherche Biomédicale, Kinshasa Gombe, Democratic Republic of the Congo
| | - Anne Marie Van der Linden
- Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Bio-Imaging Lab, Department of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Philippe Büscher
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| |
Collapse
|
73
|
Huang YK, Chu C, Wu CH, Chen CL, Chiu CH. Evaluation of Gram-negative bacterial infection by a stable and conjugative bioluminescence plasmid in a mouse model. J Biomed Sci 2014; 21:78. [PMID: 25135473 PMCID: PMC4237811 DOI: 10.1186/s12929-014-0078-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 08/11/2014] [Indexed: 12/27/2022] Open
Abstract
Background The green fluorescence protein (GFP)-associated fluorescence method and the luciferase-associated bioluminescence method are the two major methods for IVIS imaging system to investigate the bacterial infection in animal models. The aim of this study was to evaluate the infection route of Gram-negative bacteria carrying a stable and broad range of conjugative bioluminescence plasmid pSE-Lux1 in a mouse model. Results Both encapsulated and non-encapsulated Gram-negative bacteria were used as hosts to evaluate conjugation efficiency and plasmid stability of pSE-Lux1, a recombinant of pSE34 and luxABCDE operon. The plasmid conjugation efficiencies of pSE-Lux1 ranged from 10−3 to 10−7 in various Gram-negative bacteria. Plasmid pSE-Lux1 maintained in Escherichia coli, Klebsiella pneumoniae, and Salmonella enterica serovars Choleraesues (abbreviated S. Choleraesuis) and Typhimurium (S. Typhimurium), than in Acinetobacter baumannii and Serratia marcescens, was shown to be of better stability for at least four days. To investigate systemic bacterial infections, K. pneumoniae strain CG354 was intravenously injected, and then was clearly observed to be non-pathogenic to Balb/c mice for a long-term bioluminescence monitoring for 6 days. For examining dynamic distributions of gastrointestinal tract infection, the invasion protein SipB-deficient mutant OU5045△sipB and OU5046△sipB of S. serovar Typhimurium constructed in this study, compared to wild-type strain OU5045 and its virulence plasmid-less strain OU5046, were of less virulence to mice. Conclusions This is the first study to evaluate the conjugative and stable bioluminescence vehicle system of pSE-Lux1 in a wide range of Gram-negative bacteria, a system that can provide a useful reporter approach to trace systemic and gastrointestinal bacterial infections in a mouse model.
Collapse
Affiliation(s)
| | | | | | - Chyi-Liang Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | | |
Collapse
|
74
|
Pontier-Bres R, Munro P, Boyer L, Anty R, Imbert V, Terciolo C, André F, Rampal P, Lemichez E, Peyron JF, Czerucka D. Saccharomyces boulardii modifies Salmonella typhimurium traffic and host immune responses along the intestinal tract. PLoS One 2014; 9:e103069. [PMID: 25118595 PMCID: PMC4145484 DOI: 10.1371/journal.pone.0103069] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 06/27/2014] [Indexed: 12/12/2022] Open
Abstract
Salmonella enterica serovar Typhimurium (ST) is an enteropathogenic Gram-negative bacterium that causes infection following oral ingestion. ST spreads rapidly along the gastrointestinal tract (GIT) and invades the intestinal epithelium to ultimately reach internal body organs. The probiotic yeast Saccharomyces boulardii BIOCODEX (S.b-B) is prescribed for prophylaxis of diarrheal infectious diseases. We previously showed that S.b-B prevents weight loss in ST-infected mice and significantly decreases bacterial translocation to the spleen and liver. This study was designed to investigate the effect of S.b-B on ST migration along the GIT and the impact of the yeast on the host's early innate immune responses. Bioluminescent imaging (BLI) was used to evaluate the effect of S.b-B on the progression of luminescent Salmonella Typhimurium (ST-lux) in the GIT of mice pretreated with streptomycin. Photonic emission (PE) was measured in GIT extracts (stomach, small intestine, cecum and colon) at various time periods post-infection (PI). PE analysis revealed that, 45 min PI, ST-lux had migrated slightly faster in the mice treated with S.b-B than in the untreated infected animals. At 90 min PI, ST-lux had reached the cecum in both groups of mice. Adhesion of ST to S.b-B was visualized in the intestines of the mice and probably accounts for (1) the faster elimination of ST-lux in the feces, and (2) reduced translocation of ST to the spleen and liver. In the early phase of infection, S.b-B also modifies the host's immune responses by (1) increasing IFN-γ gene expression and decreasing IL-10 gene expression in the small intestine, and (2) elevating both IFN-γ, and IL-10 mRNA levels in the cecum. BLI revealed that S.b-B modifies ST migration and the host immune response along the GIT. Study findings shed new light on the protective mechanisms of S.b-B during the early phase of Salmonella pathogenesis.
Collapse
Affiliation(s)
- Rodolphe Pontier-Bres
- INSERM, U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 4 “Inflammation, Cancer, Cancer Stem Cells” Nice, France
- Université de Nice-Sophia Antipolis, UFR Médecine, IFR50, Faculté de Médecine, Nice, France
| | - Patrick Munro
- INSERM, U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 6 “Microbial toxins in host pathogen interactions” Nice, France
- Université de Nice-Sophia Antipolis, UFR Médecine, IFR50, Faculté de Médecine, Nice, France
| | - Laurent Boyer
- INSERM, U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 6 “Microbial toxins in host pathogen interactions” Nice, France
- Université de Nice-Sophia Antipolis, UFR Médecine, IFR50, Faculté de Médecine, Nice, France
| | - Rodolphe Anty
- INSERM, U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 8 “Hepatic complications in obesity” Nice, France
- Université de Nice-Sophia Antipolis, UFR Médecine, IFR50, Faculté de Médecine, Nice, France
| | - Véronique Imbert
- INSERM, U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 4 “Inflammation, Cancer, Cancer Stem Cells” Nice, France
- Université de Nice-Sophia Antipolis, UFR Médecine, IFR50, Faculté de Médecine, Nice, France
| | - Chloé Terciolo
- CRO2 INSERM U911, Campus Santé Timone, Université Aix-Marseille, Marseille, France
| | - Fréderic André
- CRO2 INSERM U911, Campus Santé Timone, Université Aix-Marseille, Marseille, France
| | | | - Emmanuel Lemichez
- INSERM, U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 6 “Microbial toxins in host pathogen interactions” Nice, France
- Université de Nice-Sophia Antipolis, UFR Médecine, IFR50, Faculté de Médecine, Nice, France
| | - Jean-François Peyron
- INSERM, U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 4 “Inflammation, Cancer, Cancer Stem Cells” Nice, France
- Université de Nice-Sophia Antipolis, UFR Médecine, IFR50, Faculté de Médecine, Nice, France
| | - Dorota Czerucka
- INSERM, U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 4 “Inflammation, Cancer, Cancer Stem Cells” Nice, France
- Université de Nice-Sophia Antipolis, UFR Médecine, IFR50, Faculté de Médecine, Nice, France
- * E-mail:
| |
Collapse
|
75
|
|
76
|
Munder A, Wölbeling F, Klockgether J, Wiehlmann L, Tümmler B. In vivoimaging of bioluminescentPseudomonas aeruginosain an acute murine airway infection model. Pathog Dis 2014; 72:74-7. [DOI: 10.1111/2049-632x.12184] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 04/29/2014] [Accepted: 05/02/2014] [Indexed: 11/29/2022] Open
Affiliation(s)
- Antje Munder
- Clinical Research Group; Hannover Medical School; Clinic for Pediatric Pneumology, Allergology and Neonatology; Hannover Germany
| | - Florian Wölbeling
- Clinical Research Group; Hannover Medical School; Clinic for Pediatric Pneumology, Allergology and Neonatology; Hannover Germany
| | - Jens Klockgether
- Clinical Research Group; Hannover Medical School; Clinic for Pediatric Pneumology, Allergology and Neonatology; Hannover Germany
| | - Lutz Wiehlmann
- Clinical Research Group; Hannover Medical School; Clinic for Pediatric Pneumology, Allergology and Neonatology; Hannover Germany
| | - Burkhard Tümmler
- Clinical Research Group; Hannover Medical School; Clinic for Pediatric Pneumology, Allergology and Neonatology; Hannover Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH); Member of the German Center for Lung Research; Hannover Germany
| |
Collapse
|
77
|
Jacobsen ID, Lüttich A, Kurzai O, Hube B, Brock M. In vivo imaging of disseminated murine Candida albicans infection reveals unexpected host sites of fungal persistence during antifungal therapy. J Antimicrob Chemother 2014; 69:2785-96. [PMID: 24951534 DOI: 10.1093/jac/dku198] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES Candida albicans is an important fungal pathogen that can cause life-threatening disseminated infections. To determine the efficacy of therapy in murine models, a determination of renal fungal burden as cfu is commonly used. However, this approach provides only a snapshot of the current situation in an individual animal and cryptic sites of infection may easily be missed. Thus, we aimed to develop real-time non-invasive imaging to monitor infection in vivo. METHODS Bioluminescent C. albicans reporter strains were developed based on a bioinformatical approach for codon optimization. The reporter strains were analysed in vitro and in vivo in the murine model of systemic candidiasis. RESULTS Reporter strains allowed the in vivo monitoring of infection and a determination of fungal burden, with a high correlation between bioluminescence and cfu count. We confirmed the kidney as the main target organ but additionally observed the translocation of C. albicans to the urinary bladder. The treatment of infected mice with caspofungin and fluconazole significantly improved the clinical outcome and clearance of C. albicans from the kidneys; however, unexpectedly, viable fungal cells persisted in the gall bladder. Fungi were secreted with bile and detected in the faeces, implicating the gall bladder as a reservoir for colonization by C. albicans after antifungal therapy. Bile extracts significantly decreased the susceptibility of C. albicans to various antifungals in vitro, thereby probably contributing to its persistence. CONCLUSIONS Using in vivo imaging, we identified cryptic sites of infection and persistence of C. albicans in the gall bladder during otherwise effective antifungal treatment. Bile appears to directly interfere with antifungal activity.
Collapse
Affiliation(s)
- Ilse D Jacobsen
- Microbial Immunology, Hans Knoell Institute, Leibniz Institute for Natural Product Research and Infection Biology, Beutenbergstrasse 11a, 07745 Jena, Germany
| | - Anja Lüttich
- Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Leibniz Institute for Natural Product Research and Infection Biology, Beutenbergstrasse 11a, 07745 Jena, Germany
| | - Oliver Kurzai
- Septomics Research Center, Friedrich-Schiller University Jena and Leibniz Institute for Natural Product Research and Infection Biology, Albert-Einstein Strasse 10, 07745 Jena, Germany
| | - Bernhard Hube
- Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Leibniz Institute for Natural Product Research and Infection Biology, Beutenbergstrasse 11a, 07745 Jena, Germany Friedrich Schiller University, Jena, Germany Center for Sepsis Control and Care, Universitätsklinikum Jena, Jena, Germany
| | - Matthias Brock
- Friedrich Schiller University, Jena, Germany Microbial Biochemistry and Physiology, Hans Knoell Institute, Leibniz Institute for Natural Product Research and Infection Biology, Beutenbergstrasse 11a, 07745 Jena, Germany
| |
Collapse
|
78
|
Carter KP, Young AM, Palmer AE. Fluorescent sensors for measuring metal ions in living systems. Chem Rev 2014; 114:4564-601. [PMID: 24588137 PMCID: PMC4096685 DOI: 10.1021/cr400546e] [Citation(s) in RCA: 1589] [Impact Index Per Article: 144.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Indexed: 02/06/2023]
Affiliation(s)
- Kyle P. Carter
- Department
of Chemistry and
Biochemistry, BioFrontiers Institute, University
of Colorado, UCB 596,
3415 Colorado AvenueBoulder, Colorado 80303, United
States
| | - Alexandra M. Young
- Department
of Chemistry and
Biochemistry, BioFrontiers Institute, University
of Colorado, UCB 596,
3415 Colorado AvenueBoulder, Colorado 80303, United
States
| | - Amy E. Palmer
- Department
of Chemistry and
Biochemistry, BioFrontiers Institute, University
of Colorado, UCB 596,
3415 Colorado AvenueBoulder, Colorado 80303, United
States
| |
Collapse
|
79
|
Paveley RA, Bickle QD. Automated imaging and other developments in whole-organism anthelmintic screening. Parasite Immunol 2014; 35:302-13. [PMID: 23581722 DOI: 10.1111/pim.12037] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 04/06/2013] [Indexed: 12/13/2022]
Abstract
Helminth infections still represent a huge public health problem throughout the developing world and in the absence of vaccines control is based on periodic mass drug administration. Poor efficacy of some anthelmintics and concerns about emergence of drug resistance has highlighted the need for new drug discovery. Most current anthelmintics were discovered through in vivo screening of selected compounds in animal models but recent approaches have shifted towards screening for activity against adult or larval stages in vitro. Larvae are normally available in greater numbers than adults, can often be produced in vitro and are small enough for microplate assays. However, the manual visualization of drug effects in vitro is subjective, laborious and slow. This can be overcome by application of automated readouts including high-content imaging. Incorporated into robotically controlled HTS platforms such methods allow the very large compound collections being made available by the pharmaceutical industry or academic organizations to be screened against helminths for the first time, invigorating the drug discovery pipeline. Here, we review the status of whole-organism screens based on in vitro activity against living worms and highlight the recent progress towards automated image-based readouts.
Collapse
Affiliation(s)
- R A Paveley
- Department of Infection and Immunity, London School of Hygiene and Tropical Medicine, London, UK
| | | |
Collapse
|
80
|
Secreted Gaussia princeps luciferase as a reporter of Escherichia coli replication in a mouse tissue cage model of infection. PLoS One 2014; 9:e90382. [PMID: 24595353 PMCID: PMC3942414 DOI: 10.1371/journal.pone.0090382] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 01/28/2014] [Indexed: 11/26/2022] Open
Abstract
Measurement of bacterial burden in animal infection models is a key component for both bacterial pathogenesis studies and therapeutic agent research. The traditional quantification means for in vivo bacterial burden requires frequent animal sacrifice and enumerating colony forming units (CFU) recovered from infection loci. To address these issues, researchers have developed a variety of luciferase-expressing bacterial reporter strains to enable bacterial detection in living animals. To date, all such luciferase-based bacterial reporters are in cell-associated form. Production of luciferase-secreting recombinant bacteria could provide the advantage of reporting CFU from both infection loci themselves and remote sampling (eg. body fluid and plasma). Toward this end, we have genetically manipulated a pathogenic Escherichia coli (E. coli) strain, ATCC25922, to secrete the marine copepod Gaussia princeps luciferase (Gluc), and assessed the use of Gluc as both an in situ and ex situ reporter for bacterial burden in mouse tissue cage infections. The E. coli expressing Gluc demonstrates in vivo imaging of bacteria in a tissue cage model of infection. Furthermore, secreted Gluc activity and bacterial CFUs recovered from tissue cage fluid (TCF) are correlated along 18 days of infection. Importantly, secreted Gluc can also be detected in plasma samples and serve as an ex situ indicator for the established tissue cage infection, once high bacterial burdens are achieved. We have demonstrated that Gluc from marine eukaryotes can be stably expressed and secreted by pathogenic E. coli in vivo to enable a facile tool for longitudinal evaluation of persistent bacterial infection.
Collapse
|
81
|
Expression of fluorescent proteins in bifidobacteria for analysis of host-microbe interactions. Appl Environ Microbiol 2014; 80:2842-50. [PMID: 24584243 DOI: 10.1128/aem.04261-13] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Bifidobacteria are an important component of the human gastrointestinal microbiota and are frequently used as probiotics. The genetic inaccessibility and lack of molecular tools commonly used in other bacteria have hampered a detailed analysis of the genetic determinants of bifidobacteria involved in their adaptation to, colonization of, and interaction with the host. In the present study, a range of molecular tools were developed that will allow the closing of some of the gaps in functional analysis of bifidobacteria. A number of promoters were tested for transcriptional activity in Bifidobacterium bifidum S17 using pMDY23, a previously published promoter probe vector. The promoter of the gap gene (Pgap) of B. bifidum S17 yielded the highest promoter activity among the promoters tested. Thus, this promoter and the pMDY23 backbone were used to construct a range of vectors for expression of different fluorescent proteins (FPs). Successful expression of cyan fluorescent protein (CFP), green fluorescent protein (GFP), yellow fluorescent protein (YFP), and mCherry could be shown for three strains representing three different Bifidobacterium spp. The red fluorescent B. bifidum S17/pVG-mCherry was further used to demonstrate application of fluorescent bifidobacteria for adhesion assays and detection in primary human macrophages cultured in vitro. Furthermore, pMGC-mCherry was cloned by combining a chloramphenicol resistance marker and expression of the FP mCherry under the control of Pgap. The chloramphenicol resistance marker of pMGC-mCherry was successfully used to determine gastrointestinal transit time of B. bifidum S17. Moreover, B. bifidum S17/pMGC-mCherry could be detected in fecal samples of mice after oral administration.
Collapse
|
82
|
Taylor MC, Kelly JM. Optimizing bioluminescence imaging to study protozoan parasite infections. Trends Parasitol 2014; 30:161-2. [PMID: 24485045 DOI: 10.1016/j.pt.2014.01.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 01/15/2014] [Accepted: 01/15/2014] [Indexed: 11/28/2022]
Abstract
Bioluminescence imaging is a non-invasive technique which can be used to monitor infections in real-time. However, its utility is restricted by difficulties in detecting pathogens in deep tissue. 'Red-shifted' luciferases, which emit light of longer wavelength than standard bioluminescence-generating proteins, greatly enhance sensitivity, and have wide applicability for studying parasite infections.
Collapse
Affiliation(s)
- Martin C Taylor
- Department of Pathogen Molecular Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - John M Kelly
- Department of Pathogen Molecular Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK.
| |
Collapse
|
83
|
Ong LC, Ang LY, Alonso S, Zhang Y. Bacterial imaging with photostable upconversion fluorescent nanoparticles. Biomaterials 2014; 35:2987-98. [PMID: 24412082 DOI: 10.1016/j.biomaterials.2013.12.060] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 12/19/2013] [Indexed: 10/25/2022]
Abstract
Autofluorescence, photodamage and photobleaching are often encountered when using downconverting fluorophores and fluorescent proteins for bacteria labeling. These caveats represent a serious limitation when trying to map bacteria dissemination for prolonged periods. Upconversion nanoparticles (UCNs), which are able to convert low energy near-infrared (NIR) excitation light into higher energy visible or NIR light, can address these limitations. These particles' unique optical properties translate into attractive advantages of minimal autofluorescence, reduced photodamage, deeper tissue penetration and prolonged photostability. Here, we report a UCN-based bacteria labeling strategy using Escherichia coli as prototypic bacteria. A comparative analysis highlighted the superior photostability of UCN-labeled bacteria over green fluorescent protein-expressing bacteria. Infection study of UCN-labeled bacteria in dendritic cells indicated co-localization of the UCN signal with bacterial position for up to 6 h post-infection. Furthermore, long-term monitoring of the same infected cells demonstrated the potential to utilize photostable UCN-based imaging for bacterial trafficking purposes.
Collapse
Affiliation(s)
- Li Ching Ong
- Graduate School for Integrative Sciences and Engineering, National University of Singapore, Centre for Life Sciences (CeLS), #05-01, 28 Medical Drive, Singapore 117456, Singapore
| | - Lei Yin Ang
- Department of Microbiology, Immunology Program, National University of Singapore, Centre for Life Sciences (CeLS), #03-05, 28 Medical Drive, Singapore 117456, Singapore
| | - Sylvie Alonso
- Graduate School for Integrative Sciences and Engineering, National University of Singapore, Centre for Life Sciences (CeLS), #05-01, 28 Medical Drive, Singapore 117456, Singapore; Department of Microbiology, Immunology Program, National University of Singapore, Centre for Life Sciences (CeLS), #03-05, 28 Medical Drive, Singapore 117456, Singapore.
| | - Yong Zhang
- Graduate School for Integrative Sciences and Engineering, National University of Singapore, Centre for Life Sciences (CeLS), #05-01, 28 Medical Drive, Singapore 117456, Singapore; Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Block EA #03-12, 9 Engineering Drive 1, Singapore 117575, Singapore.
| |
Collapse
|
84
|
Kassem II, Splitter GA, Miller S, Rajashekara G. Let There Be Light! Bioluminescent Imaging to Study Bacterial Pathogenesis in Live Animals and Plants. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2014; 154:119-45. [DOI: 10.1007/10_2014_280] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
85
|
Alam FM, Bateman C, Turner CE, Wiles S, Sriskandan S. Non-invasive monitoring of Streptococcus pyogenes vaccine efficacy using biophotonic imaging. PLoS One 2013; 8:e82123. [PMID: 24278474 PMCID: PMC3835743 DOI: 10.1371/journal.pone.0082123] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Accepted: 10/21/2013] [Indexed: 11/18/2022] Open
Abstract
Streptococcus pyogenes infection of the nasopharynx represents a key step in the pathogenic cycle of this organism and a major focus for vaccine development, requiring robust models to facilitate the screening of potentially protective antigens. One antigen that may be an important target for vaccination is the chemokine protease, SpyCEP, which is cell surface-associated and plays a role in pathogenesis. Biophotonic imaging (BPI) can non-invasively characterize the spatial location and abundance of bioluminescent bacteria in vivo. We have developed a bioluminescent derivative of a pharyngeal S. pyogenes strain by transformation of an emm75 clinical isolate with the luxABCDE operon. Evaluation of isogenic recombinant strains in vitro and in vivo confirmed that bioluminescence conferred a growth deficit that manifests as a fitness cost during infection. Notwithstanding this, bioluminescence expression permitted non-invasive longitudinal quantitation of S. pyogenes within the murine nasopharynx albeit with a detection limit corresponding to approximately 105 bacterial colony forming units (CFU) in this region. Vaccination of mice with heat killed streptococci, or with SpyCEP led to a specific IgG response in the serum. BPI demonstrated that both vaccine candidates reduced S. pyogenes bioluminescence emission over the course of nasopharyngeal infection. The work suggests the potential for BPI to be used in the non-invasive longitudinal evaluation of potential S. pyogenes vaccines.
Collapse
Affiliation(s)
- Faraz M. Alam
- Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London, United Kingdom
| | - Colin Bateman
- Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London, United Kingdom
| | - Claire E. Turner
- Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London, United Kingdom
| | - Siouxsie Wiles
- Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London, United Kingdom
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
- * E-mail: (SS); (SW)
| | - Shiranee Sriskandan
- Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London, United Kingdom
- * E-mail: (SS); (SW)
| |
Collapse
|
86
|
Loh JMS, Proft T. Toxin-antitoxin-stabilized reporter plasmids for biophotonic imaging of Group A streptococcus. Appl Microbiol Biotechnol 2013; 97:9737-45. [PMID: 24061415 DOI: 10.1007/s00253-013-5200-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 08/11/2013] [Accepted: 08/14/2013] [Indexed: 01/19/2023]
Abstract
Bioluminescence is a rapid and cost-efficient optical imaging technology that allows the detection of bacteria in real-time during disease development. Here, we report a novel strategy to generate a wide range of bioluminescent group A streptococcus (GAS) strains by using a toxin-antitoxin-stabilized plasmid. The bacterial luciferin-luciferase operon (lux) or the firefly luciferase gene (ffluc) was introduced into GAS via a stabilized plasmid. The FFluc reporter gave significantly stronger bioluminescent signals than the Lux reporter, and was generally more stable. Plasmid-based luciferase reporters could easily be introduced into a variety of GAS strains and the signals correlated linearly with viable cell counts. Co-expression of the streptococcal ω-ε-ζ toxin-antitoxin operon provided segregational stability in the absence of antibiotics for at least 17 passages in vitro and up to 7 days in a mouse infection model. In addition, genome-integrated reporter constructs were also generated by site-specific recombination, but were found to be technically more challenging. The quick and efficient generation of various M-type GAS strains expressing plasmid-based luciferase reporters with comparable and quantifiable bioluminescence signals allows for comparative analysis of different GAS strains in vitro and in vivo.
Collapse
Affiliation(s)
- Jacelyn M S Loh
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | | |
Collapse
|
87
|
Vrana NE, Lavalle P, Dokmeci MR, Dehghani F, Ghaemmaghami AM, Khademhosseini A. Engineering functional epithelium for regenerative medicine and in vitro organ models: a review. TISSUE ENGINEERING PART B-REVIEWS 2013; 19:529-43. [PMID: 23705900 DOI: 10.1089/ten.teb.2012.0603] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Recent advances in the fields of microfabrication, biomaterials, and tissue engineering have provided new opportunities for developing biomimetic and functional tissues with potential applications in disease modeling, drug discovery, and replacing damaged tissues. An intact epithelium plays an indispensable role in the functionality of several organs such as the trachea, esophagus, and cornea. Furthermore, the integrity of the epithelial barrier and its degree of differentiation would define the level of success in tissue engineering of other organs such as the bladder and the skin. In this review, we focus on the challenges and requirements associated with engineering of epithelial layers in different tissues. Functional epithelial layers can be achieved by methods such as cell sheets, cell homing, and in situ epithelialization. However, for organs composed of several tissues, other important factors such as (1) in vivo epithelial cell migration, (2) multicell-type differentiation within the epithelium, and (3) epithelial cell interactions with the underlying mesenchymal cells should also be considered. Recent successful clinical trials in tissue engineering of the trachea have highlighted the importance of a functional epithelium for long-term success and survival of tissue replacements. Hence, using the trachea as a model tissue in clinical use, we describe the optimal structure of an artificial epithelium as well as challenges of obtaining a fully functional epithelium in macroscale. One of the possible remedies to address such challenges is the use of bottom-up fabrication methods to obtain a functional epithelium. Modular approaches for the generation of functional epithelial layers are reviewed and other emerging applications of microscale epithelial tissue models for studying epithelial/mesenchymal interactions in healthy and diseased (e.g., cancer) tissues are described. These models can elucidate the epithelial/mesenchymal tissue interactions at the microscale and provide the necessary tools for the next generation of multicellular engineered tissues and organ-on-a-chip systems.
Collapse
Affiliation(s)
- Nihal E Vrana
- 1 Institut National de la Santé et de la Recherche Médicale , INSERM, UMR-S 1121, "Biomatériaux et Bioingénierie," Strasbourg Cedex, France
| | | | | | | | | | | |
Collapse
|
88
|
Foss CA, Harper JS, Wang H, Pomper MG, Jain SK. Noninvasive molecular imaging of tuberculosis-associated inflammation with radioiodinated DPA-713. J Infect Dis 2013; 208:2067-74. [PMID: 23901092 DOI: 10.1093/infdis/jit331] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Increased expression of translocator protein (TSPO) is a feature of microglial and macrophage activation. Since activated macrophages are key components of tuberculosis-associated inflammation, we evaluated radioiodinated DPA-713, a synthetic ligand of TSPO, for in vivo imaging of host response. METHODS Mice were infected with aerosolized Mycobacterium tuberculosis and evaluated using whole-body [(125)I]iodo-DPA-713 single-photon emission computed tomography (SPECT). Ex vivo biodistribution and correlative immunofluorescence studies were also performed. RESULTS [(125)I]Iodo-DPA-713 SPECT imaging clearly delineated tuberculosis-associated pulmonary inflammation in live animals. Biodistribution studies confirmed radiotracer specificity for inflamed pulmonary tissues. Immunofluorescence studies demonstrated that TSPO is highly expressed in CD68(+) macrophages and phagocytic cells within tuberculosis lesions and that [(125)I]DPA-713 specifically accumulates within these cells. Coadministration of excess unlabelled DPA-713 abrogated both the SPECT and ex vivo fluorescence signals. Lesion-specific signal-to-noise ratios were significantly higher with [(125)I]iodo-DPA-713 SPECT (4.06 ± 0.52) versus [(18)F]fluorodeoxyglucose (FDG) positron emission tomography (PET) (2.00 ± 0.28) performed in the same mice (P = .004). CONCLUSIONS [(125)I]Iodo-DPA-713 accumulates specifically in tuberculosis-associated inflammatory lesions by selective retention within macrophages and phagocytic cells. [(125)I]Iodo-DPA-713 SPECT provides higher lesion-specific signal-to-noise ratios than [(18)F]FDG PET and may prove to be a more specific biomarker to monitor tuberculosis in situ.
Collapse
|
89
|
ZELMER A, WARD T. Noninvasive fluorescence imaging of small animals. J Microsc 2013; 252:8-15. [DOI: 10.1111/jmi.12063] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 06/06/2013] [Indexed: 12/13/2022]
Affiliation(s)
- A. ZELMER
- Immunology and Infection Department; London School of Hygiene and Tropical Medicine; London UK
| | - T.H. WARD
- Immunology and Infection Department; London School of Hygiene and Tropical Medicine; London UK
| |
Collapse
|
90
|
Xu T, Close DM, Webb JD, Price SL, Ripp SA, Sayler GS. Continuous, real-time bioimaging of chemical bioavailability and toxicology using autonomously bioluminescent human cell lines. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2013; 8723:872310. [PMID: 26516295 DOI: 10.1117/12.2015030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Bioluminescent imaging is an emerging biomedical surveillance strategy that uses external cameras to detect in vivo light generated in small animal models of human physiology or in vitro light generated in tissue culture or tissue scaffold mimics of human anatomy. The most widely utilized of reporters is the firefly luciferase (luc) gene; however, it generates light only upon addition of a chemical substrate, thus only generating intermittent single time point data snapshots. To overcome this disadvantage, we have demonstrated substrate-independent bioluminescent imaging using an optimized bacterial bioluminescence (lux) system. The lux reporter produces bioluminescence autonomously using components found naturally within the cell, thereby allowing imaging to occur continuously and in real-time over the lifetime of the host. We have validated this technology in human cells with demonstrated chemical toxicological profiling against exotoxin exposures at signal strengths comparable to existing luc systems (~1.33 × 107 photons/second). As a proof-in-principle demonstration, we have engineered breast carcinoma cells to express bioluminescence for real-time screening of endocrine disrupting chemicals and validated detection of 17β-estradiol (EC50 = ~ 10 pM). These and other applications of this new reporter technology will be discussed as potential new pathways towards improved models of target chemical bioavailability, toxicology, efficacy, and human safety.
Collapse
Affiliation(s)
- Tingting Xu
- Joint Institute for Biological Sciences, The University of Tennessee, 676 Dabney Hall, Knoxville, TN, USA 37996
| | - Dan M Close
- 490 BioTech Inc., 2450 E. J. Chapman Drive, Knoxville, TN USA 37996
| | - James D Webb
- Center for Environmental Biotechnology, The University of Tennessee, 676 Dabney Hall, Knoxville, TN, USA 37996
| | - Sarah L Price
- Center for Environmental Biotechnology, The University of Tennessee, 676 Dabney Hall, Knoxville, TN, USA 37996
| | - Steven A Ripp
- 490 BioTech Inc., 2450 E. J. Chapman Drive, Knoxville, TN USA 37996 ; Center for Environmental Biotechnology, The University of Tennessee, 676 Dabney Hall, Knoxville, TN, USA 37996
| | - Gary S Sayler
- Joint Institute for Biological Sciences, The University of Tennessee, 676 Dabney Hall, Knoxville, TN, USA 37996 ; 490 BioTech Inc., 2450 E. J. Chapman Drive, Knoxville, TN USA 37996 ; Center for Environmental Biotechnology, The University of Tennessee, 676 Dabney Hall, Knoxville, TN, USA 37996
| |
Collapse
|
91
|
Andreu N, Zelmer A, Sampson SL, Ikeh M, Bancroft GJ, Schaible UE, Wiles S, Robertson BD. Rapid in vivo assessment of drug efficacy against Mycobacterium tuberculosis using an improved firefly luciferase. J Antimicrob Chemother 2013; 68:2118-27. [PMID: 23633686 PMCID: PMC3743513 DOI: 10.1093/jac/dkt155] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Objectives In vivo experimentation is costly and time-consuming, and presents a major bottleneck in anti-tuberculosis drug development. Conventional methods rely on the enumeration of bacterial colonies, and it can take up to 4 weeks for Mycobacterium tuberculosis to grow on agar plates. Light produced by recombinant bacteria expressing luciferase enzymes can be used as a marker of bacterial load, and disease progression can be easily followed non-invasively in live animals by using the appropriate imaging equipment. The objective of this work was to develop a bioluminescence-based mouse model of tuberculosis to assess antibiotic efficacy against M. tuberculosis in vivo. Methods We used an M. tuberculosis strain carrying a red-shifted derivative of the firefly luciferase gene (FFlucRT) to infect mice, and monitored disease progression in living animals by bioluminescence imaging before and after treatment with the frontline anti-tuberculosis drug isoniazid. The resulting images were analysed and the bioluminescence was correlated with bacterial counts. Results Using bioluminescence imaging we detected as few as 1.7 × 103 and 7.5 × 104 reporter bacteria ex vivo and in vivo, respectively, in the lungs of mice. A good correlation was found between bioluminescence and bacterial load in both cases. Furthermore, a marked reduction in luminescence was observed in living mice given isoniazid treatment. Conclusions We have shown that an improved bioluminescent strain of M. tuberculosis can be visualized by non-invasive imaging in live mice during an acute, progressive infection and that this technique can be used to rapidly visualize and quantify the effect of antibiotic treatment. We believe that the model presented here will be of great benefit in early drug discovery as an easy and rapid way to identify active compounds in vivo.
Collapse
Affiliation(s)
- Nuria Andreu
- MRC Centre for Molecular Bacteriology and Infection, Department of Medicine, Imperial College London, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
92
|
Assessment of efficacy of antifungals against Aspergillus fumigatus: value of real-time bioluminescence imaging. Antimicrob Agents Chemother 2013; 57:3046-59. [PMID: 23587947 DOI: 10.1128/aac.01660-12] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Aspergillus fumigatus causes life-threatening infections, especially in immunocompromised patients. Common drugs for therapy of aspergillosis are polyenes, azoles, and echinocandins. However, despite in vitro efficacy of these antifungals, treatment failure is frequently observed. In this study, we established bioluminescence imaging to monitor drug efficacy under in vitro and in vivo conditions. In vitro assays confirmed the effectiveness of liposomal amphotericin B, voriconazole, and anidulafungin. Liposomal amphotericin B and voriconazole were fungicidal, whereas anidulafungin allowed initial germination of conidia that stopped elongation but allowed the conidia to remain viable. In vivo studies were performed with a leukopenic murine model. Mice were challenged by intranasal instillation with a bioluminescent reporter strain (5 × 10(5) and 2.5 × 10(5) conidia), and therapy efficacies of liposomal amphotericin B, voriconazole, and anidulafungin were monitored. For monotherapy, the highest treatment efficacy was observed with liposomal amphotericin B, whereas the efficacies of voriconazole and anidulafungin were strongly dependent on the infectious dose. When therapy efficacy was studied with different drug combinations, all combinations improved the rate of treatment success compared to that with monotherapy. One hundred percent survival was obtained for treatment with a combination of liposomal amphotericin B and anidulafungin, which prevented not only pulmonary infections but also infections of the sinus. In conclusion, combination therapy increases treatment success, at least in the murine infection model. In addition, our novel approach based on real-time imaging enables in vivo monitoring of drug efficacy in different organs during therapy of invasive aspergillosis.
Collapse
|
93
|
Li XF, Deng YQ, Zhao H, Ye Q, Wang HJ, Li SH, Zhu SY, Shi PY, Qin ED, Zhang B, Qin CF. Noninvasive bioluminescence imaging of dengue virus infection in the brain of A129 mice. Appl Microbiol Biotechnol 2013; 97:4589-96. [PMID: 23467830 DOI: 10.1007/s00253-013-4799-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 02/17/2013] [Accepted: 02/19/2013] [Indexed: 11/29/2022]
Abstract
Dengue virus (DENV) infection is one of the most important public health threats globally; however, no vaccines or effective antivirals are currently available. The bioluminescence imaging technique has emerged as a powerful tool for studies on viral pathogenesis in vitro and in vivo. In this study, using a recombinant DENV that stably expressed Renilla luciferase (Rluc-DENV), we used bioluminescence for imaging of DENV infection in the brain of A129 mice that lacked type I interferon receptors. Upon intracranial inoculation with Rluc-DENV, A129 mice developed typical neurological symptoms and rapidly succumbed to viral infection. Real-time bioluminescence intensity analysis revealed the replication kinetics of Rluc-DENV in the brain of A129 mice. Linear regression analyses showed a good correlation between photon flux and viral titers (R(2) = 0.9923). Finally, the bioluminescence model was validated using a known mouse monoclonal antibody, 2A10G6, and the therapeutic effects of this neutralizing antibody were readily monitored by live imaging in the same animal. The noninvasive bioluminescence imaging of DENV infection as described here shows distinct advantages over traditional animal models and provides a powerful tool for potential antiviral or vaccine assays against DENV infection in vivo.
Collapse
Affiliation(s)
- Xiao-Feng Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
D'Archivio S, Cosson A, Medina M, Lang T, Minoprio P, Goyard S. Non-invasive in vivo study of the Trypanosoma vivax infectious process consolidates the brain commitment in late infections. PLoS Negl Trop Dis 2013; 7:e1976. [PMID: 23301112 PMCID: PMC3536815 DOI: 10.1371/journal.pntd.0001976] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 11/04/2012] [Indexed: 01/08/2023] Open
Abstract
Trypanosoma vivax, one of the leading parasites responsible for Animal African Trypanosomosis (Nagana), is generally cyclically transmitted by Glossina spp. but in areas devoid of the tsetse flies in Africa or in Latin American countries is mechanically transmitted across vertebrate hosts by other haematophagous insects, including tabanids. We followed on from our recent studies on the maintenance of this parasite in vivo and in vitro, and its genetic manipulation, by constructing a West African IL1392 T. vivax strain that stably expresses firefly luciferase and is fully virulent for immunocompetent mice. We report here on a study where murine infection with this strain was monitored in vivo using a non-invasive method. Study findings fully support the use of this strain in the assessment of parasite dynamics in vivo since a strong correlation was found between whole body light emission measured over the course of the infection and parasitemia determined microscopically. In addition, parasitemia and survival rates were very similar for mice infected by the intraperitoneal and sub-cutaneous routes, except for a longer prepatent period following sub-cutaneous inoculation with the parasite. Our results clearly show that when administered by the subcutaneous route, the parasite is retained few days in the skin close to the inoculation site where it multiplies before passing into the bloodstream. Ex vivo bioluminescence analyses of organs isolated from infected mice corroborated our previous histopathological observations with parasite infiltration into spleen, liver and lungs. Finally, our study reinforces previous observations on the presence of the parasite in the central nervous system and consequently the brain commitment in the very late phases of the experimental infection.
Collapse
Affiliation(s)
- Simon D'Archivio
- Institut Pasteur, Laboratoire des Processus Infectieux à Trypanosoma, Department of Infection and Epidemiology, Paris, France
| | - Alain Cosson
- Institut Pasteur, Laboratoire des Processus Infectieux à Trypanosoma, Department of Infection and Epidemiology, Paris, France
| | - Mathieu Medina
- Institut Pasteur, Laboratoire des Processus Infectieux à Trypanosoma, Department of Infection and Epidemiology, Paris, France
| | - Thierry Lang
- Institut Pasteur, Laboratoire d'Immunophysiologie et Parasitisme, Department of Parasitology, Paris, France
| | - Paola Minoprio
- Institut Pasteur, Laboratoire des Processus Infectieux à Trypanosoma, Department of Infection and Epidemiology, Paris, France
| | - Sophie Goyard
- Institut Pasteur, Laboratoire des Processus Infectieux à Trypanosoma, Department of Infection and Epidemiology, Paris, France
| |
Collapse
|
95
|
Liu D. Technical Advances in Veterinary Diagnostic Microbiology. ADVANCED TECHNIQUES IN DIAGNOSTIC MICROBIOLOGY 2013. [PMCID: PMC7121739 DOI: 10.1007/978-1-4614-3970-7_35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Forming a significant part of biomass on earth, microorganisms are renowned for their abundance and diversity. From submicroscopic infectious particles (viruses), small unicellular cells (bacteria and yeasts) to multinucleate and multicellular organisms (filamentous fungi, protozoa, and helminths), microorganisms have found their way into virtually every environmental niche, and show little restrain in making their presence felt. While a majority of microorganisms are free-living and involved in the degradation of plant debris and other organic materials, others lead a symbiotic, mutually beneficial life within their hosts. In addition, some microorganisms have the capacity to take advantage of temporary weaknesses in animal and human hosts, causing notable morbidity and mortality. Because clinical manifestations in animals and humans resulting from infections with various microorganisms are often nonspecific (e.g., general malaise and fever), it is necessary to apply laboratory diagnostic means to identify the culprit organisms for treatment and prevention purposes.
Collapse
|
96
|
Lass S, Hudson PJ, Thakar J, Saric J, Harvill E, Albert R, Perkins SE. Generating super-shedders: co-infection increases bacterial load and egg production of a gastrointestinal helminth. J R Soc Interface 2012; 10:20120588. [PMID: 23256186 PMCID: PMC3565725 DOI: 10.1098/rsif.2012.0588] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Co-infection by multiple parasites is common within individuals. Interactions between co-infecting parasites include resource competition, direct competition and immune-mediated interactions and each are likely to alter the dynamics of single parasites. We posit that co-infection is a driver of variation in parasite establishment and growth, ultimately altering the production of parasite transmission stages. To test this hypothesis, three different treatment groups of laboratory mice were infected with the gastrointestinal helminth Heligmosomoides polygyrus, the respiratory bacterial pathogen Bordetella bronchiseptica lux+ or co-infected with both parasites. To follow co-infection simultaneously, self-bioluminescent bacteria were used to quantify infection in vivo and in real-time, while helminth egg production was monitored in real-time using faecal samples. Co-infection resulted in high bacterial loads early in the infection (within the first 5 days) that could cause host mortality. Co-infection also produced helminth ‘super-shedders’; individuals that chronically shed the helminth eggs in larger than average numbers. Our study shows that co-infection may be one of the underlying mechanisms for the often-observed high variance in parasite load and shedding rates, and should thus be taken into consideration for disease management and control. Further, using self-bioluminescent bacterial reporters allowed quantification of the progression of infection within the whole animal of the same individuals at a fine temporal scale (daily) and significantly reduced the number of animals used (by 85%) compared with experiments that do not use in vivo techniques. Thus, we present bioluminescent imaging as a novel, non-invasive tool offering great potential to be taken forward into other applications of infectious disease ecology.
Collapse
Affiliation(s)
- Sandra Lass
- Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA 16802, USA
| | | | | | | | | | | | | |
Collapse
|
97
|
In vivo bioluminescence imaging to evaluate systemic and topical antibiotics against community-acquired methicillin-resistant Staphylococcus aureus-infected skin wounds in mice. Antimicrob Agents Chemother 2012. [PMID: 23208713 DOI: 10.1128/aac.01003-12] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Community-acquired methicillin-resistant Staphylococcus aureus (CA-MRSA) frequently causes skin and soft tissue infections, including impetigo, cellulitis, folliculitis, and infected wounds and ulcers. Uncomplicated CA-MRSA skin infections are typically managed in an outpatient setting with oral and topical antibiotics and/or incision and drainage, whereas complicated skin infections often require hospitalization, intravenous antibiotics, and sometimes surgery. The aim of this study was to develop a mouse model of CA-MRSA wound infection to compare the efficacy of commonly used systemic and topical antibiotics. A bioluminescent USA300 CA-MRSA strain was inoculated into full-thickness scalpel wounds on the backs of mice and digital photography/image analysis and in vivo bioluminescence imaging were used to measure wound healing and the bacterial burden. Subcutaneous vancomycin, daptomycin, and linezolid similarly reduced the lesion sizes and bacterial burden. Oral linezolid, clindamycin, and doxycycline all decreased the lesion sizes and bacterial burden. Oral trimethoprim-sulfamethoxazole decreased the bacterial burden but did not decrease the lesion size. Topical mupirocin and retapamulin ointments both reduced the bacterial burden. However, the petrolatum vehicle ointment for retapamulin, but not the polyethylene glycol vehicle ointment for mupirocin, promoted wound healing and initially increased the bacterial burden. Finally, in type 2 diabetic mice, subcutaneous linezolid and daptomycin had the most rapid therapeutic effect compared with vancomycin. Taken together, this mouse model of CA-MRSA wound infection, which utilizes in vivo bioluminescence imaging to monitor the bacterial burden, represents an alternative method to evaluate the preclinical in vivo efficacy of systemic and topical antimicrobial agents.
Collapse
|
98
|
Bioluminescence imaging study of spatial and temporal persistence of Lactobacillus plantarum and Lactococcus lactis in living mice. Appl Environ Microbiol 2012. [PMID: 23204409 DOI: 10.1128/aem.03221-12] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Lactic acid bacteria, especially lactobacilli, are common inhabitants of the gastrointestinal tract of mammals, for which they have received considerable attention due to their putative health-promoting properties. In this study, we describe the development and application of luciferase-expressing Lactobacillus plantarum and Lactococcus lactis strains for noninvasive in vivo monitoring in the digestive tract of mice. We report for the first time the functional in vitro expression in Lactobacillus plantarum NCIMB8826 and in Lactococcus lactis MG1363 of the click beetle luciferase (CBluc), as well as Gaussia and bacterial luciferases, using a combination of vectors, promoters, and codon-optimized genes. We demonstrate that a CBluc construction is the best-performing luciferase system for the noninvasive in vivo detection of lactic acid bacteria after oral administration. The persistence and viability of both strains was studied by bioluminescence imaging in anesthetized mice and in mouse feces. In vivo bioluminescence imaging confirmed that after a single or multiple oral administrations, L. lactis has shorter survival times in the mouse gastrointestinal tract than L. plantarum, and it also revealed the precise gut compartments where both strains persisted. The application of luciferase-labeled bacteria has significant potential to allow the in vivo and ex vivo study of the interactions of lactic acid bacteria with their mammalian host.
Collapse
|
99
|
Cho JS, Guo Y, Ramos RI, Hebroni F, Plaisier SB, Xuan C, Granick JL, Matsushima H, Takashima A, Iwakura Y, Cheung AL, Cheng G, Lee DJ, Simon SI, Miller LS. Neutrophil-derived IL-1β is sufficient for abscess formation in immunity against Staphylococcus aureus in mice. PLoS Pathog 2012; 8:e1003047. [PMID: 23209417 PMCID: PMC3510260 DOI: 10.1371/journal.ppat.1003047] [Citation(s) in RCA: 184] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 10/05/2012] [Indexed: 02/07/2023] Open
Abstract
Neutrophil abscess formation is critical in innate immunity against many pathogens. Here, the mechanism of neutrophil abscess formation was investigated using a mouse model of Staphylococcus aureus cutaneous infection. Gene expression analysis and in vivo multispectral noninvasive imaging during the S. aureus infection revealed a strong functional and temporal association between neutrophil recruitment and IL-1β/IL-1R activation. Unexpectedly, neutrophils but not monocytes/macrophages or other MHCII-expressing antigen presenting cells were the predominant source of IL-1β at the site of infection. Furthermore, neutrophil-derived IL-1β was essential for host defense since adoptive transfer of IL-1β-expressing neutrophils was sufficient to restore the impaired neutrophil abscess formation in S. aureus-infected IL-1β-deficient mice. S. aureus-induced IL-1β production by neutrophils required TLR2, NOD2, FPR1 and the ASC/NLRP3 inflammasome in an α-toxin-dependent mechanism. Taken together, IL-1β and neutrophil abscess formation during an infection are functionally, temporally and spatially linked as a consequence of direct IL-1β production by neutrophils. Invasive infections caused by the human pathogen Staphylococcus aureus result in more deaths annually than infections caused by any other single infectious agent in the United States. Although neutrophil recruitment and abscess formation is crucial for effective host defense against this pathogen, how neutrophils sense and mount an inflammatory response are not completely clear. Using gene expression analysis and in vivo bioluminescence and fluorescence imaging, we found that neutrophil recruitment during a S. aureus cutaneous infection is functionally and temporally linked to IL-1β/IL-1R activation. Surprisingly, neutrophils themselves were determined to be the most abundant cell type that produced IL-1β during infection. Further, neutrophil-derived IL-1β, in the absence of other cellular sources of IL-1β, was sufficient for neutrophil recruitment, abscess formation, and bacterial clearance. Finally, mouse neutrophils produced IL-1β in direct response to live S. aureus in vitro. These findings expand our understanding of the acute neutrophil response to infection in which early recruited neutrophils serve as a source of IL-1β that is essential for amplifying and sustaining the neutrophilic response to promote abscess formation and bacterial clearance. Therapies aimed at promoting IL-1β production by neutrophils may be an effective immunotherapeutic strategy to control S. aureus infections.
Collapse
Affiliation(s)
- John S. Cho
- Department of Medicine, Division of Dermatology, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California, United States of America
| | - Yi Guo
- Department of Medicine, Division of Dermatology, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California, United States of America
| | - Romela Irene Ramos
- Department of Medicine, Division of Dermatology, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California, United States of America
| | - Frank Hebroni
- Department of Medicine, Division of Dermatology, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California, United States of America
| | - Seema B. Plaisier
- Department of Translational Immunology, Dirks/Dougherty Laboratory for Cancer Research, John Wayne Cancer Institute, Santa Monica, California, United States of America
| | - Caiyun Xuan
- Department of Translational Immunology, Dirks/Dougherty Laboratory for Cancer Research, John Wayne Cancer Institute, Santa Monica, California, United States of America
| | - Jennifer L. Granick
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
| | - Hironori Matsushima
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine, Toledo, Ohio, United States of America
| | - Akira Takashima
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine, Toledo, Ohio, United States of America
| | - Yoichiro Iwakura
- Center for Experimental Medicine and Systems Biology, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan and Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Saitama, Japan
| | - Ambrose L. Cheung
- Department of Microbiology and Immunology, Dartmouth Medical School, Hanover, New Hampshire, United States of America
| | - Genhong Cheng
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California, United States of America
| | - Delphine J. Lee
- Department of Translational Immunology, Dirks/Dougherty Laboratory for Cancer Research, John Wayne Cancer Institute, Santa Monica, California, United States of America
| | - Scott I. Simon
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
| | - Lloyd S. Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
100
|
Niska JA, Meganck JA, Pribaz JR, Shahbazian JH, Lim E, Zhang N, Rice BW, Akin A, Ramos RI, Bernthal NM, Francis KP, Miller LS. Monitoring bacterial burden, inflammation and bone damage longitudinally using optical and μCT imaging in an orthopaedic implant infection in mice. PLoS One 2012; 7:e47397. [PMID: 23082163 PMCID: PMC3474799 DOI: 10.1371/journal.pone.0047397] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 09/12/2012] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Recent advances in non-invasive optical, radiographic and μCT imaging provide an opportunity to monitor biological processes longitudinally in an anatomical context. One particularly relevant application for combining these modalities is to study orthopaedic implant infections. These infections are characterized by the formation of persistent bacterial biofilms on the implanted materials, causing inflammation, periprosthetic osteolysis, osteomyelitis, and bone damage, resulting in implant loosening and failure. METHODOLOGY/PRINCIPAL FINDINGS An orthopaedic implant infection model was used in which a titanium Kirshner-wire was surgically placed in femurs of LysEGFP mice, which possess EGFP-fluorescent neutrophils, and a bioluminescent S. aureus strain (Xen29; 1×10(3) CFUs) was inoculated in the knee joint before closure. In vivo bioluminescent, fluorescent, X-ray and μCT imaging were performed on various postoperative days. The bacterial bioluminescent signals of the S. aureus-infected mice peaked on day 19, before decreasing to a basal level of light, which remained measurable for the entire 48 day experiment. Neutrophil EGFP-fluorescent signals of the S. aureus-infected mice were statistically greater than uninfected mice on days 2 and 5, but afterwards the signals for both groups approached background levels of detection. To visualize the three-dimensional location of the bacterial infection and neutrophil infiltration, a diffuse optical tomography reconstruction algorithm was used to co-register the bioluminescent and fluorescent signals with μCT images. To quantify the anatomical bone changes on the μCT images, the outer bone volume of the distal femurs were measured using a semi-automated contour based segmentation process. The outer bone volume increased through day 48, indicating that bone damage continued during the implant infection. CONCLUSIONS/SIGNIFICANCE Bioluminescent and fluorescent optical imaging was combined with X-ray and μCT imaging to provide noninvasive and longitudinal measurements of the dynamic changes in bacterial burden, neutrophil recruitment and bone damage in a mouse orthopaedic implant infection model.
Collapse
Affiliation(s)
- Jared A Niska
- Orthopaedic Hospital Research Center, Orthopaedic Hospital Department of Orthopaedic Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|