1
|
Dupouy B, Cotos L, Binder A, Slavikova L, Rottmann M, Mäser P, Jacquemin D, Ganter M, Davioud‐Charvet E, Elhabiri M. Click Coupling of Flavylium Dyes with Plasmodione Analogues: Towards New Redox-Sensitive Pro-Fluorophores. Chemistry 2025; 31:e202403691. [PMID: 39654502 PMCID: PMC11771622 DOI: 10.1002/chem.202403691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Indexed: 12/17/2024]
Abstract
The development of redox-sensitive molecular fluorescent probes for the detection of redox changes in Plasmodium falciparum-parasitized red blood cells remains of interest due to the limitations of current genetically encoded biosensors. This study describes the design, screening and synthesis of new pro-fluorophores based on flavylium azido dyes coupled by CuAAC click chemistry to alkynyl analogues of plasmodione oxide, the key metabolite of the potent redox-active antimalarial plasmodione. The photophysical and electrochemical properties of these probes were evaluated, focusing on their fluorogenic responses. The influence of both the redox status of the quinone and the length of the PEG chain separating the fluorophore from the electrophore on the photophysical properties was investigated. The fluorescence quenching by photoinduced electron transfer is reversible and of high amplitude for probes in oxidized quinone forms and fluorescence is reinstated for reduced hydroquinone forms. Our results demonstrate that shortening the PEG chain has the effect of enhancing the fluorogenic response, likely due to non-covalent interactions between the two chromophores. All these systems were evaluated for their antiparasitic activities and fluorescence imaging suggests the efficacy of the fluorescent flavylium dyes in P. falciparum-parasitized red blood cells, paving the way for future parasite imaging studies to monitor cellular redox processes.
Collapse
Affiliation(s)
- Baptiste Dupouy
- Laboratoire d'Innovation Moléculaire et Applications, UMR 7042CNRS-Unistra-UHA, ECPM25 Rue Becquerel67200StrasbourgFrance
| | - Leandro Cotos
- Laboratoire d'Innovation Moléculaire et Applications, UMR 7042CNRS-Unistra-UHA, ECPM25 Rue Becquerel67200StrasbourgFrance
| | - Annika Binder
- Heidelberg UniversityMedical Faculty, Centre for Infectious DiseasesIm Neuenheimer Feld 324/34469120HeidelbergGermany
| | - Lucie Slavikova
- Laboratoire d'Innovation Moléculaire et Applications, UMR 7042CNRS-Unistra-UHA, ECPM25 Rue Becquerel67200StrasbourgFrance
| | - Matthias Rottmann
- Swiss Tropical and Public Health InstituteKreuzstrasse 2CH-4123AllschwilSwitzerland
| | - Pascal Mäser
- Swiss Tropical and Public Health InstituteKreuzstrasse 2CH-4123AllschwilSwitzerland
- University of BaselPetersgraben 1CH-4001BaselSwitzerland
| | - Denis Jacquemin
- Nantes Université, CNRS, CEISAMF-44000NantesFrance
- Institut Universitaire de France (IUF)F-75005ParisFrance
| | - Markus Ganter
- Heidelberg UniversityMedical Faculty, Centre for Infectious DiseasesIm Neuenheimer Feld 324/34469120HeidelbergGermany
| | - Elisabeth Davioud‐Charvet
- Laboratoire d'Innovation Moléculaire et Applications, UMR 7042CNRS-Unistra-UHA, ECPM25 Rue Becquerel67200StrasbourgFrance
| | - Mourad Elhabiri
- Laboratoire d'Innovation Moléculaire et Applications, UMR 7042CNRS-Unistra-UHA, ECPM25 Rue Becquerel67200StrasbourgFrance
| |
Collapse
|
2
|
Jiang T, Bai X, Li M. Advances in the Development of Bacterial Bioluminescence Imaging. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2024; 17:265-288. [PMID: 38640069 DOI: 10.1146/annurev-anchem-061622-034229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Abstract
Bioluminescence imaging (BLI) is a powerful method for visualizing biological processes and tracking cells. Engineered bioluminescent bacteria that utilize luciferase-catalyzed biochemical reactions to generate luminescence have become useful analytical tools for in vitro and in vivo bacterial imaging. Accordingly, this review initially introduces the development of engineered bioluminescent bacteria that use different luciferase-luciferin pairs as analytical tools and their applications for in vivo BLI, including real-time bacterial tracking of infection, probiotic investigation, tumor-targeted therapy, and drug screening. Applications of engineered bioluminescent bacteria as whole-cell biosensors for sensing biological changes in vitro and in vivo are then discussed. Finally, we review the optimizations and future directions of bioluminescent bacteria for imaging. This review aims to provide fundamental insights into bacterial BLI and highlight the potential development of this technique in the future.
Collapse
Affiliation(s)
- Tianyu Jiang
- 1Helmholtz International Lab for Anti-Infectives, State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao, Shandong, China
| | - Xiaoyu Bai
- 1Helmholtz International Lab for Anti-Infectives, State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao, Shandong, China
- 2School of Life Sciences, Shandong University, Qingdao, Shandong, China
| | - Minyong Li
- 3Key Laboratory of Chemical Biology (MOE), Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China;
| |
Collapse
|
3
|
Pan T, Su L, Zhang Y, Xu L, Chen Y. Advances in Bio-Optical Imaging Systems for Spatiotemporal Monitoring of Intestinal Bacteria. Mol Nutr Food Res 2024; 68:e2300760. [PMID: 38491399 DOI: 10.1002/mnfr.202300760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/26/2024] [Indexed: 03/18/2024]
Abstract
Vast and complex intestinal communities are regulated and balanced through interactions with their host organisms, and disruption of gut microbial balance can cause a variety of diseases. Studying the mechanisms of pathogenic intestinal flora in the host and early detection of bacterial translocation and colonization can guide clinical diagnosis, provide targeted treatments, and improve patient prognosis. The use of in vivo imaging techniques to track microorganisms in the intestine, and study structural and functional changes of both cells and proteins, may clarify the governing equilibrium between the flora and host. Despite the recent rapid development of in vivo imaging of intestinal microecology, determining the ideal methodology for clinical use remains a challenge. Advances in optics, computer technology, and molecular biology promise to expand the horizons of research and development, thereby providing exciting opportunities to study the spatio-temporal dynamics of gut microbiota and the origins of disease. Here, this study reviews the characteristics and problems associated with optical imaging techniques, including bioluminescence, conventional fluorescence, novel metabolic labeling methods, nanomaterials, intelligently activated imaging agents, and photoacoustic (PA) imaging. It hopes to provide a valuable theoretical basis for future bio-intelligent imaging of intestinal bacteria.
Collapse
Affiliation(s)
- Tongtong Pan
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University & Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Ouhai District, Wenzhou, Zhejiang, 325035, China
| | - Lihuang Su
- The First Affiliated Hospital of Wenzhou Medical University, Ouhai District, Wenzhou, Zhejiang, 325035, China
| | - Yiying Zhang
- Alberta Institute, Wenzhou Medical University, Ouhai District, Wenzhou, Zhejiang, 325035, China
| | - Liang Xu
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yongping Chen
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University & Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Ouhai District, Wenzhou, Zhejiang, 325035, China
| |
Collapse
|
4
|
Broudic K, Laurent S, Perkov V, Simon C, Garinot M, Truchot N, Latour J, Désert P. Nonclinical safety assessment of an mRNA Covid-19 vaccine candidate following repeated administrations and biodistribution. J Appl Toxicol 2024; 44:371-390. [PMID: 37723625 DOI: 10.1002/jat.4548] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/28/2023] [Accepted: 09/04/2023] [Indexed: 09/20/2023]
Abstract
Messenger RNA (mRNA) vaccines have demonstrated efficacy against Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) in humans. mRNA technology holds tremendous potential for rapid control and prevention of emergencies due to its flexibility with respect to production, application, and design for an efficacious and safe use in humans. We assessed the toxicity and biodistribution of MRT5500, an mRNA vaccine encoding for the full-length of the SARS-CoV-2 spike protein and delivered by lipid nanoparticles (LNPs) containing a novel ionizable lipid, Lipid-1 in preclinical animal models. In the repeated dose toxicity study, rabbits received three intramuscular (IM) injections of MRT5500 at 3-week interval followed by a 4-week observation period. In an exploratory biodistribution study in mice receiving a single IM injection of an mRNA encoding luciferase encapsulated in an LNP containing Lipid-1, the expression of the luciferase protein was monitored in vivo and ex vivo at several time points. In the regulatory biodistribution study in rabbits receiving a single IM injection of MRT5500, the quantification of the mRNA and the ionizable Lipid-1 were monitored in the same organs and time points as in the exploratory biodistribution study. MRT5500 was safe and well-tolerated with a transient acute phase response/inflammation and an expected vaccine-related immunological response, typical of those observed following a vaccine administration. The biodistribution data demonstrated that the mRNA and Lipid-1 components of the vaccine formulations were mainly detected at the injection site and in the draining lymph nodes. These results support the use of MRT5500 and its deployment into clinical trials.
Collapse
Affiliation(s)
- Karine Broudic
- Research and Development, Sanofi, Marcy l'Etoile, France
| | | | | | - Charlene Simon
- Research and Development, Sanofi, Marcy l'Etoile, France
| | - Marie Garinot
- Research and Development, Sanofi, Marcy l'Etoile, France
| | - Nathalie Truchot
- France Safety Assessment SAS, Charles River Laboratories, Saint-Germain-Nuelles, France
| | - Julie Latour
- Research and Development, Sanofi, Marcy l'Etoile, France
| | - Paul Désert
- Research and Development, Sanofi, Marcy l'Etoile, France
| |
Collapse
|
5
|
Dunuweera AN, Dunuweera SP, Ranganathan K. A Comprehensive Exploration of Bioluminescence Systems, Mechanisms, and Advanced Assays for Versatile Applications. Biochem Res Int 2024; 2024:8273237. [PMID: 38347947 PMCID: PMC10861286 DOI: 10.1155/2024/8273237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/10/2023] [Accepted: 01/21/2024] [Indexed: 02/15/2024] Open
Abstract
Bioluminescence has been a fascinating natural phenomenon of light emission from living creatures. It happens when the enzyme luciferase facilitates the oxidation of luciferin, resulting in the creation of an excited-state species that emits light. Although there are many bioluminescent systems, few have been identified. D-luciferin-dependent systems, coelenterazine-dependent systems, Cypridina luciferin-based systems, tetrapyrrole-based luciferins, bacterial bioluminescent systems, and fungal bioluminescent systems are natural bioluminescent systems. Since different bioluminescence systems, such as various combinations of luciferin-luciferase pair reactions, have different light emission wavelengths, they benefit industrial applications such as drug discovery, protein-protein interactions, in vivo imaging in small animals, and controlling neurons. Due to the expression of luciferase and easy permeation of luciferin into most cells and tissues, bioluminescence assays are applied nowadays with modern technologies in most cell and tissue types. It is a versatile technique in a variety of biomedical research. Furthermore, there are some investigated blue-sky research projects, such as bioluminescent plants and lamps. This review article is mainly based on the theory of diverse bioluminescence systems and their past, present, and future applications.
Collapse
Affiliation(s)
| | | | - K. Ranganathan
- Department of Botany, University of Jaffna, Jaffna 40000, Sri Lanka
| |
Collapse
|
6
|
Reyne N, McCarron A, Cmielewski P, Parsons D, Donnelley M. To bead or not to bead: A review of Pseudomonas aeruginosa lung infection models for cystic fibrosis. Front Physiol 2023; 14:1104856. [PMID: 36824474 PMCID: PMC9942929 DOI: 10.3389/fphys.2023.1104856] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/25/2023] [Indexed: 02/10/2023] Open
Abstract
Cystic fibrosis (CF) lung disease is characterised by recurring bacterial infections resulting in inflammation, lung damage and ultimately respiratory failure. Pseudomonas aeruginosa is considered one of the most important lung pathogens in those with cystic fibrosis. While multiple cystic fibrosis animal models have been developed, many fail to mirror the cystic fibrosis lung disease of humans, including the colonisation by opportunistic environmental pathogens. Delivering bacteria to the lungs of animals in different forms is a way to model cystic fibrosis bacterial lung infections and disease. This review presents an overview of previous models, and factors to consider when generating a new P. aeruginosa lung infection model. The future development and application of lung infection models that more accurately reflect human cystic fibrosis lung disease has the potential to assist in understanding the pathophysiology of cystic fibrosis lung disease and for developing treatments.
Collapse
Affiliation(s)
- Nicole Reyne
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia,Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia,Respiratory and Sleep Medicine, Women’s and Children’s Hospital, North Adelaide, SA, Australia,*Correspondence: Nicole Reyne,
| | - Alexandra McCarron
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia,Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia,Respiratory and Sleep Medicine, Women’s and Children’s Hospital, North Adelaide, SA, Australia
| | - Patricia Cmielewski
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia,Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia,Respiratory and Sleep Medicine, Women’s and Children’s Hospital, North Adelaide, SA, Australia
| | - David Parsons
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia,Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia,Respiratory and Sleep Medicine, Women’s and Children’s Hospital, North Adelaide, SA, Australia
| | - Martin Donnelley
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia,Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia,Respiratory and Sleep Medicine, Women’s and Children’s Hospital, North Adelaide, SA, Australia
| |
Collapse
|
7
|
Liebert A, Capon W, Pang V, Vila D, Bicknell B, McLachlan C, Kiat H. Photophysical Mechanisms of Photobiomodulation Therapy as Precision Medicine. Biomedicines 2023; 11:biomedicines11020237. [PMID: 36830774 PMCID: PMC9953702 DOI: 10.3390/biomedicines11020237] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
Despite a significant focus on the photochemical and photoelectrical mechanisms underlying photobiomodulation (PBM), its complex functions are yet to be fully elucidated. To date, there has been limited attention to the photophysical aspects of PBM. One effect of photobiomodulation relates to the non-visual phototransduction pathway, which involves mechanotransduction and modulation to cytoskeletal structures, biophotonic signaling, and micro-oscillatory cellular interactions. Herein, we propose a number of mechanisms of PBM that do not depend on cytochrome c oxidase. These include the photophysical aspects of PBM and the interactions with biophotons and mechanotransductive processes. These hypotheses are contingent on the effect of light on ion channels and the cytoskeleton, the production of biophotons, and the properties of light and biological molecules. Specifically, the processes we review are supported by the resonant recognition model (RRM). This previous research demonstrated that protein micro-oscillations act as a signature of their function that can be activated by resonant wavelengths of light. We extend this work by exploring the local oscillatory interactions of proteins and light because they may affect global body circuits and could explain the observed effect of PBM on neuro-cortical electroencephalogram (EEG) oscillations. In particular, since dysrhythmic gamma oscillations are associated with neurodegenerative diseases and pain syndromes, including migraine with aura and fibromyalgia, we suggest that transcranial PBM should target diseases where patients are affected by impaired neural oscillations and aberrant brain wave patterns. This review also highlights examples of disorders potentially treatable with precise wavelengths of light by mimicking protein activity in other tissues, such as the liver, with, for example, Crigler-Najjar syndrome and conditions involving the dysregulation of the cytoskeleton. PBM as a novel therapeutic modality may thus behave as "precision medicine" for the treatment of various neurological diseases and other morbidities. The perspectives presented herein offer a new understanding of the photophysical effects of PBM, which is important when considering the relevance of PBM therapy (PBMt) in clinical applications, including the treatment of diseases and the optimization of health outcomes and performance.
Collapse
Affiliation(s)
- Ann Liebert
- Faculty of Medicine and Health, University of Sydney, Sydney 2006, Australia
- Adventist Hospital Group, Wahroonga 2076, Australia
- NICM Health Research Institute, Western Sydney University, Westmead 2145, Australia
- Correspondence:
| | - William Capon
- Faculty of Medicine and Health, University of Sydney, Sydney 2006, Australia
| | - Vincent Pang
- NICM Health Research Institute, Western Sydney University, Westmead 2145, Australia
| | - Damien Vila
- Faculty of Medicine of Montpellier-Nîmes, University of Montpellier, 34090 Montpellier, France
| | - Brian Bicknell
- NICM Health Research Institute, Western Sydney University, Westmead 2145, Australia
| | - Craig McLachlan
- Faculty of Health, Torrens University, Adelaide 5000, Australia
| | - Hosen Kiat
- NICM Health Research Institute, Western Sydney University, Westmead 2145, Australia
- Faculty of Health, Torrens University, Adelaide 5000, Australia
- Cardiac Health Institute, Sydney 2121, Australia
- ANU College of Health and Medicine, Australian National University, Canberra 2600, Australia
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Macquarie Park 2109, Australia
| |
Collapse
|
8
|
Leanse LG, Hong W, de Souza Rastelli AN. Editorial: Photonics-based diagnosis and treatment of infectious and inflammatory diseases. Front Microbiol 2023; 14:1205032. [PMID: 37187541 PMCID: PMC10175852 DOI: 10.3389/fmicb.2023.1205032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Affiliation(s)
- Leon G. Leanse
- Health and Sport Sciences Hub, University of Gibraltar, Gibraltar, Gibraltar
- Wellman Center for Photomedicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
- *Correspondence: Leon G. Leanse
| | - Weili Hong
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Alessandra Nara de Souza Rastelli
- Department of Restorative Dentistry, Araraquara School of Dentistry, São Paulo State University-UNESP, Araraquara, São Paulo, Brazil
| |
Collapse
|
9
|
Johnson K, Juelich T, Smith J, Lee B, Freiberg AN. In Vivo Imaging of Nipah Virus Infection in Small Animal Rodent Models. Methods Mol Biol 2023; 2682:149-157. [PMID: 37610580 DOI: 10.1007/978-1-0716-3283-3_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
In vivo imaging system (IVIS) is a powerful tool for the study of infectious diseases, providing the ability to non-invasively follow viral infection in an individual animal over time. Recombinant henipaviruses expressing bioluminescent or fluorescent reporter proteins can be used both to monitor the spatial and temporal progression of Nipah virus (NiV) infection in vivo as well as in ex vivo tissues. Virally produced luciferases react with systemically administered substrate to produce bioluminescence that can then be detected via IVIS imaging, while fluorescent reporters inherently generate detectable fluorescence without a substrate. Here we describe protocols applying bioluminescent or fluorescent reporter expressing recombinant viruses to in vivo or ex vivo imaging of NiV infection.
Collapse
Affiliation(s)
- Kendra Johnson
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Terry Juelich
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jennifer Smith
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander N Freiberg
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
10
|
Marasini S, Dean SJ, Swift S, Perera J, Rupenthal ID, Wang T, Read H, Craig JP. Preclinical confirmation of UVC efficacy in treating infectious keratitis. Ocul Surf 2022; 25:76-86. [PMID: 35568373 DOI: 10.1016/j.jtos.2022.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 05/06/2022] [Accepted: 05/07/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE Preclinical evaluation of the therapeutic potential of antimicrobial 265 nm UVC for infectious keratitis. METHODS Four experiments explored UVC: 1) impact on bacterial and fungal lawns on agar, in individual or mixed culture, 2) bacterial inactivation dose in an in vitro deep corneal infection model, 3) dose validation in an ex vivo porcine keratitis model and 4) efficacy in a masked, randomised, controlled murine keratitis trial using bioluminescent Pseudomonas aeruginosa. RESULTS Minimum effective UVC exposures ranged between 2 s and 5 s for lawn bacteria and fungi in individual or mixed culture. Significant P. aeruginosa growth inhibition in the in vitro infection model was achieved with 15 s UVC, that resulted in a >3.5 log10 reduction of bacteria in a subsequent ex vivo keratitis model (p < 0.05). Bioluminescence fell below baseline levels in all treated animals, within 8 h of treatment (p < 0.05), in the in vivo study. Re-epithelialisation with corneal clarity occurred within 24 h in 75% of UVC-treated cases, with no relapse at 48 h. On plating, bacteria were recovered only from untreated controls. CONCLUSIONS UVC inhibited all tested bacteria and fungi, including mixed culture and strains linked to antibiotic resistance, in vitro, with exposures of ≤ 5 s. In vitro and ex vivo testing confirmed therapeutic potential of 15 s UVC. In vivo, 15 s UVC administered in two doses, 4 h apart, proved effective in treating murine bacterial keratitis.
Collapse
Affiliation(s)
- Sanjay Marasini
- Department of Ophthalmology, New Zealand National Eye Centre, The University of Auckland, Auckland, New Zealand
| | - Simon J Dean
- Department of Ophthalmology, New Zealand National Eye Centre, The University of Auckland, Auckland, New Zealand
| | - Simon Swift
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
| | - Janesha Perera
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
| | - Ilva D Rupenthal
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, The University of Auckland, Auckland, New Zealand
| | - Tao Wang
- Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Hannah Read
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
| | - Jennifer P Craig
- Department of Ophthalmology, New Zealand National Eye Centre, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
11
|
Marcelo GA, Galhano J, Duarte MP, Capelo-Martínez JL, Lodeiro C, Oliveira E. Validation of a Standard Luminescence Method for the Fast Determination of the Antimicrobial Activity of Nanoparticles in Escherichia coli. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:2164. [PMID: 35807997 PMCID: PMC9268724 DOI: 10.3390/nano12132164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 01/25/2023]
Abstract
The use of nanoparticles in multiple industries has raised concerned voices about the assessment of their toxicity/antimicrobial activity and the development of standardized handling protocols. Issues emerge during the antimicrobial assaying of multiple cargo, colorimetric, colloidal nanoformulations, as standard protocols often rely on visual evaluations, or optical density (OD) measurements, leading to high variance inhibitory concentrations (MIC). Thus, a fast, luminescence-based assay for the effective assessment of the antimicrobial activity of nanoparticles is herein reported, using the bioluminescence of an in-house E. coli ATCC® 8739TM construct with the pMV306G13 + Lux plasmid (E. coli Lux). The new strain's sensitivity to ofloxacin as a standard antibiotic was confirmed, and the methodology robustness verified against multiple nanoparticles and colorimetric drugs. The reduction of incubation from 24 to only 8 h, and the sole use of luminescence (LUX490) to accurately determine and distinguish MIC50 and MIC90, are two main advantages of the method. By discarding OD measurements, one can avoid turbidity and color interferences when calculating bacterial growth. This approach is an important tool that contributes to the standardization of methods, reducing samples' background interference and focusing on luminescence as a direct probe for bacterial metabolic activity, growth and, most importantly, the correct assessment of nanomaterials' antimicrobial activity.
Collapse
Affiliation(s)
- Gonçalo A. Marcelo
- BIOSCOPE Group, LAQV-REQUIMTE, Chemistry Department, NOVA School of Science and Technology, FCT NOVA, NOVA University Lisbon, 2829-516 Caparica, Portugal; (G.A.M.); (J.G.); (J.L.C.-M.); (C.L.)
| | - Joana Galhano
- BIOSCOPE Group, LAQV-REQUIMTE, Chemistry Department, NOVA School of Science and Technology, FCT NOVA, NOVA University Lisbon, 2829-516 Caparica, Portugal; (G.A.M.); (J.G.); (J.L.C.-M.); (C.L.)
| | - Maria Paula Duarte
- MEtRICs, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - José Luis Capelo-Martínez
- BIOSCOPE Group, LAQV-REQUIMTE, Chemistry Department, NOVA School of Science and Technology, FCT NOVA, NOVA University Lisbon, 2829-516 Caparica, Portugal; (G.A.M.); (J.G.); (J.L.C.-M.); (C.L.)
- PROTEOMASS Scientific Society, Rua dos Inventores, Madam Parque, Caparica Campus, 2825-182 Caparica, Portugal
| | - Carlos Lodeiro
- BIOSCOPE Group, LAQV-REQUIMTE, Chemistry Department, NOVA School of Science and Technology, FCT NOVA, NOVA University Lisbon, 2829-516 Caparica, Portugal; (G.A.M.); (J.G.); (J.L.C.-M.); (C.L.)
- PROTEOMASS Scientific Society, Rua dos Inventores, Madam Parque, Caparica Campus, 2825-182 Caparica, Portugal
| | - Elisabete Oliveira
- BIOSCOPE Group, LAQV-REQUIMTE, Chemistry Department, NOVA School of Science and Technology, FCT NOVA, NOVA University Lisbon, 2829-516 Caparica, Portugal; (G.A.M.); (J.G.); (J.L.C.-M.); (C.L.)
- PROTEOMASS Scientific Society, Rua dos Inventores, Madam Parque, Caparica Campus, 2825-182 Caparica, Portugal
| |
Collapse
|
12
|
Lian X, Scott-Thomas A, Lewis JG, Bhatia M, MacPherson SA, Zeng Y, Chambers ST. Monoclonal Antibodies and Invasive Aspergillosis: Diagnostic and Therapeutic Perspectives. Int J Mol Sci 2022; 23:5563. [PMID: 35628374 PMCID: PMC9146623 DOI: 10.3390/ijms23105563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 12/13/2022] Open
Abstract
Invasive aspergillosis (IA) is a life-threatening fungal disease that causes high morbidity and mortality in immunosuppressed patients. Early and accurate diagnosis and treatment of IA remain challenging. Given the broad range of non-specific clinical symptoms and the shortcomings of current diagnostic techniques, most patients are either diagnosed as "possible" or "probable" cases but not "proven". Moreover, because of the lack of sensitive and specific tests, many high-risk patients receive an empirical therapy or a prolonged treatment of high-priced antifungal agents, leading to unnecessary adverse effects and a high risk of drug resistance. More precise diagnostic techniques alongside a targeted antifungal treatment are fundamental requirements for reducing the morbidity and mortality of IA. Monoclonal antibodies (mAbs) with high specificity in targeting the corresponding antigen(s) may have the potential to improve diagnostic tests and form the basis for novel IA treatments. This review summarizes the up-to-date application of mAb-based approaches in assisting IA diagnosis and therapy.
Collapse
Affiliation(s)
- Xihua Lian
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand; (X.L.); (A.S.-T.); (J.G.L.); (M.B.); (S.A.M.)
- Department of Medical Imaging, The Second Clinical Medical School of Fujian Medical University, Quanzhou 362000, China
| | - Amy Scott-Thomas
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand; (X.L.); (A.S.-T.); (J.G.L.); (M.B.); (S.A.M.)
| | - John G. Lewis
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand; (X.L.); (A.S.-T.); (J.G.L.); (M.B.); (S.A.M.)
- Steroid and Immunobiochemistry Laboratory, Canterbury Health Laboratories, Christchurch 8140, New Zealand
| | - Madhav Bhatia
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand; (X.L.); (A.S.-T.); (J.G.L.); (M.B.); (S.A.M.)
| | - Sean A. MacPherson
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand; (X.L.); (A.S.-T.); (J.G.L.); (M.B.); (S.A.M.)
- Haematology Department, Christchurch Hospital, Christchurch 8011, New Zealand
| | - Yiming Zeng
- Department of Internal Medicine (Pulmonary and Critical Care Medicine), The Second Clinical Medical School of Fujian Medical University, Quanzhou 362000, China;
| | - Stephen T. Chambers
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand; (X.L.); (A.S.-T.); (J.G.L.); (M.B.); (S.A.M.)
| |
Collapse
|
13
|
A Novel In Vitro Method to Assess the Microbial Barrier Function of Tissue Adhesives Using Bioluminescence Imaging Technique. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3483238. [PMID: 35047631 PMCID: PMC8763484 DOI: 10.1155/2022/3483238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/14/2021] [Indexed: 11/17/2022]
Abstract
Background. Tissue glues can minimize treatment invasiveness, mitigate the risk of infection, and reduce surgery time; ergo, they have been developed and used in surgical procedures as wound closure devices beside sutures, staples, and metallic grafts. Regardless of their structure or function, tissue glues should show an acceptable microbial barrier function before being used in humans. This study proposes a novel in vitro method using Escherichia coli Lux and bioluminescence imaging technique to assess the microbial barrier function of tissue glues. Different volumes and concentrations of E. coli Lux were applied to precured or cured polyurethane-based tissue glue placed on agar plates. Plates were cultured for 1 h, 24 h, 48 h, and 72 h with bioluminescence signal measurement subsequently. Herein, protocol established a volume of 5 μL of a 1 : 100 dilution of E. coli Lux containing around 2 × 107 CFU/mL as optimal for testing polyurethane-based tissue glue. Measurement of OD600nm, determination of CFU/mL, and correlation with the bioluminescence measurement in p/s unit resulted in a good correlation between CFU/mL and p/s and demonstrated good reproducibility of our method. In addition, this in vitro method could show that the tested polyurethane-based tissue glue can provide a reasonable barrier against the microbial penetration and act as a bacterial barrier for up to 48 h with no penetration and up to 72 h with a low level of penetration through the material. Overall, we have established a novel, sensitive, and reproducible in vitro method using the bioluminescence imaging technique for testing the microbial barrier function of new tissue glues.
Collapse
|
14
|
Pan T, Lu D, Xin H, Li B. Biophotonic probes for bio-detection and imaging. LIGHT, SCIENCE & APPLICATIONS 2021; 10:124. [PMID: 34108445 PMCID: PMC8190087 DOI: 10.1038/s41377-021-00561-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/10/2021] [Accepted: 05/21/2021] [Indexed: 05/08/2023]
Abstract
The rapid development of biophotonics and biomedical sciences makes a high demand on photonic structures to be interfaced with biological systems that are capable of manipulating light at small scales for sensitive detection of biological signals and precise imaging of cellular structures. However, conventional photonic structures based on artificial materials (either inorganic or toxic organic) inevitably show incompatibility and invasiveness when interfacing with biological systems. The design of biophotonic probes from the abundant natural materials, particularly biological entities such as virus, cells and tissues, with the capability of multifunctional light manipulation at target sites greatly increases the biocompatibility and minimizes the invasiveness to biological microenvironment. In this review, advances in biophotonic probes for bio-detection and imaging are reviewed. We emphatically and systematically describe biological entities-based photonic probes that offer appropriate optical properties, biocompatibility, and biodegradability with different optical functions from light generation, to light transportation and light modulation. Three representative biophotonic probes, i.e., biological lasers, cell-based biophotonic waveguides and bio-microlenses, are reviewed with applications for bio-detection and imaging. Finally, perspectives on future opportunities and potential improvements of biophotonic probes are also provided.
Collapse
Affiliation(s)
- Ting Pan
- Institute of Nanophotonics, Jinan University, Guangzhou, 511443, China
| | - Dengyun Lu
- Institute of Nanophotonics, Jinan University, Guangzhou, 511443, China
| | - Hongbao Xin
- Institute of Nanophotonics, Jinan University, Guangzhou, 511443, China.
| | - Baojun Li
- Institute of Nanophotonics, Jinan University, Guangzhou, 511443, China.
| |
Collapse
|
15
|
Robertson J, McGoverin C, White JR, Vanholsbeeck F, Swift S. Rapid Detection of Escherichia coli Antibiotic Susceptibility Using Live/Dead Spectrometry for Lytic Agents. Microorganisms 2021; 9:924. [PMID: 33925816 PMCID: PMC8147107 DOI: 10.3390/microorganisms9050924] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023] Open
Abstract
Antibiotic resistance is a serious threat to public health. The empiric use of the wrong antibiotic occurs due to urgency in treatment combined with slow, culture-based diagnostic techniques. Inappropriate antibiotic choice can promote the development of antibiotic resistance. We investigated live/dead spectrometry using a fluorimeter (Optrode) as a rapid alternative to culture-based techniques through application of the LIVE/DEAD® BacLightTM Bacterial Viability Kit. Killing was detected by the Optrode in near real-time when Escherichia coli was treated with lytic antibiotics-ampicillin and polymyxin B-and stained with SYTO 9 and/or propidium iodide. Antibiotic concentration, bacterial growth phase, and treatment time used affected the efficacy of this detection method. Quantification methods of the lethal action and inhibitory action of the non-lytic antibiotics, ciprofloxacin and chloramphenicol, respectively, remain to be elucidated.
Collapse
Affiliation(s)
- Julia Robertson
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland 1023, New Zealand; (J.R.W.); (S.S.)
- The Dodd-Walls Centre for Photonic and Quantum Technologies, Auckland 1010, New Zealand; (C.M.); (F.V.)
| | - Cushla McGoverin
- The Dodd-Walls Centre for Photonic and Quantum Technologies, Auckland 1010, New Zealand; (C.M.); (F.V.)
- Department of Physics, The University of Auckland, Auckland 1010, New Zealand
| | - Joni R. White
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland 1023, New Zealand; (J.R.W.); (S.S.)
- The Dodd-Walls Centre for Photonic and Quantum Technologies, Auckland 1010, New Zealand; (C.M.); (F.V.)
| | - Frédérique Vanholsbeeck
- The Dodd-Walls Centre for Photonic and Quantum Technologies, Auckland 1010, New Zealand; (C.M.); (F.V.)
- Department of Physics, The University of Auckland, Auckland 1010, New Zealand
| | - Simon Swift
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland 1023, New Zealand; (J.R.W.); (S.S.)
| |
Collapse
|
16
|
Chaconas G, Moriarty TJ, Skare J, Hyde JA. Live Imaging. Curr Issues Mol Biol 2020; 42:385-408. [PMID: 33310914 PMCID: PMC7946808 DOI: 10.21775/cimb.042.385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Being able to vizualize a pathogen at a site of interaction with a host is an aesthetically appealing idea and the resulting images can be both informative as well as enjoyable to view. Moreover, the approaches used to derive these images can be powerful in terms of offering data unobtainable by other methods. In this article, we review three primary modalities for live imaging Borrelia spirochetes: whole animal imaging, intravital microscopy and live cell imaging. Each method has strengths and weaknesses, which we review, as well as specific purposes for which they are optimally utilized. Live imaging borriliae is a relatively recent development and there was a need of a review to cover the area. Here, in addition to the methods themselves, we also review areas of spirochete biology that have been significantly impacted by live imaging and present a collection of images associated with the forward motion in the field driven by imaging studies.
Collapse
Affiliation(s)
- George Chaconas
- Department of Biochemistry and Molecular Biology and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Tara J. Moriarty
- Faculties of Dentistry and Medicine (Laboratory Medicine and Pathobiology), University of Toronto, Toronto, Ontario, M5G 1G6, Canada
| | - Jon Skare
- Department of Microbial Pathogenesis and Immunology, Texas A & M University Health Science Center, Bryan, Texas, 77807, USA
| | - Jenny A. Hyde
- Department of Microbial Pathogenesis and Immunology, Texas A & M University Health Science Center, Bryan, Texas, 77807, USA
| |
Collapse
|
17
|
van Zyl WF, Deane SM, Dicks LM. Molecular insights into probiotic mechanisms of action employed against intestinal pathogenic bacteria. Gut Microbes 2020; 12:1831339. [PMID: 33112695 PMCID: PMC7595611 DOI: 10.1080/19490976.2020.1831339] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal (GI) diseases, and in particular those caused by bacterial infections, are a major cause of morbidity and mortality worldwide. Treatment is becoming increasingly difficult due to the increase in number of species that have developed resistance to antibiotics. Probiotic lactic acid bacteria (LAB) have considerable potential as alternatives to antibiotics, both in prophylactic and therapeutic applications. Several studies have documented a reduction, or prevention, of GI diseases by probiotic bacteria. Since the activities of probiotic bacteria are closely linked with conditions in the host's GI-tract (GIT) and changes in the population of enteric microorganisms, a deeper understanding of gut-microbial interactions is required in the selection of the most suitable probiotic. This necessitates a deeper understanding of the molecular capabilities of probiotic bacteria. In this review, we explore how probiotic microorganisms interact with enteric pathogens in the GIT. The significance of probiotic colonization and persistence in the GIT is also addressed.
Collapse
Affiliation(s)
- Winschau F. van Zyl
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa
| | - Shelly M. Deane
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa
| | - Leon M.T. Dicks
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa,CONTACT Leon M.T. Dicks; Department of Microbiology; Stellenbosch University, Stellenbosch7602, South Africa
| |
Collapse
|
18
|
Saccharomyces cerevisiae and Candida albicans Yeast Cells Labeled with Fe(III) Complexes as MRI Probes. MAGNETOCHEMISTRY 2020. [DOI: 10.3390/magnetochemistry6030041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The development of MRI probes is of interest for labeling antibiotic-resistant fungal infections based on yeast. Our work showed that yeast cells can be labeled with high-spin Fe(III) complexes to produce enhanced T2 water proton relaxation. These Fe(III)-based macrocyclic complexes contained a 1,4,7-triazacyclononane framework, two pendant alcohol groups, and either a non-coordinating ancillary group and a bound water molecule or a third coordinating pendant. The Fe(III) complexes that had an open coordination site associated strongly with Saccharomyces cerevisiae upon incubation, as shown by screening using Z-spectra analysis. The incubation of one Fe(III) complex with either Saccharomyces cerevisiae or Candida albicans yeast led to an interaction with the β-glucan-based cell wall, as shown by the ready retrieval of the complex by the bidentate chelator called maltol. Other conditions, such as a heat shock treatment of the complexes, produced Fe(III) complex uptake that could not be reversed by the addition of maltol. Appending a fluorescence dye to Fe(TOB) led to uptake through secretory pathways, as shown by confocal fluorescence microscopy and by the incomplete retrieval of the Fe(III) complex by the maltol treatment. Yeast cells that were labeled with these Fe(III) complexes displayed enhanced water proton T2 relaxation, both for S. cerevisiae and for yeast and hyphal forms of C. albicans.
Collapse
|
19
|
Eberhardt E, Bulté D, Van Bockstal L, Van den Kerkhof M, Cos P, Delputte P, Hendrickx S, Maes L, Caljon G. Miltefosine enhances the fitness of a non-virulent drug-resistant Leishmania infantum strain. J Antimicrob Chemother 2020; 74:395-406. [PMID: 30412253 DOI: 10.1093/jac/dky450] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 10/05/2018] [Indexed: 01/17/2023] Open
Abstract
Objectives Miltefosine is currently the only oral drug for visceral leishmaniasis, and although deficiency in an aminophospholipid/miltefosine transporter (MT) is sufficient to elicit drug resistance, very few naturally miltefosine-resistant (MIL-R) strains have yet been isolated. This study aimed to make a detailed analysis of the impact of acquired miltefosine resistance and miltefosine treatment on in vivo infection. Methods Bioluminescent versions of a MIL-R strain and its syngeneic parental line were generated by integration of the red-shifted firefly luciferase PpyRE9. The fitness of both lines was compared in vitro (growth rate, metacyclogenesis and macrophage infectivity) and in BALB/c mice through non-invasive bioluminescence imaging under conditions with and without drug pressure. Results This study demonstrated a severe fitness loss of MT-deficient parasites, resulting in a complete inability to multiply and cause a typical visceral leishmaniasis infection pattern in BALB/c mice. The observed fitness loss could not be rescued by host immune suppression with cyclophosphamide, whereas episomal reconstitution with a wild-type MT restored parasite virulence, hence linking parasite fitness to MT mutation. Remarkably, in vivo miltefosine treatment or in vitro miltefosine pre-exposure significantly rescued MIL-R parasite virulence. The in vitro pre-exposed MIL-R promastigotes showed a longer and more slender morphology, suggesting an altered membrane composition. Conclusions The profound fitness loss of MT-deficient parasites most likely explains the low frequency of MIL-R clinical isolates. The observation that miltefosine can reverse this phenotype indicates a drug dependency of the MT-deficient parasites and emphasizes the importance of resistance profiling prior to miltefosine administration.
Collapse
Affiliation(s)
- Eline Eberhardt
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Wilrijk, Belgium
| | - Dimitri Bulté
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Wilrijk, Belgium
| | - Lieselotte Van Bockstal
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Wilrijk, Belgium
| | - Magali Van den Kerkhof
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Wilrijk, Belgium
| | - Paul Cos
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Wilrijk, Belgium
| | - Peter Delputte
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Wilrijk, Belgium
| | - Sarah Hendrickx
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Wilrijk, Belgium
| | - Louis Maes
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Wilrijk, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
20
|
Álvarez-Bardón M, Pérez-Pertejo Y, Ordóñez C, Sepúlveda-Crespo D, Carballeira NM, Tekwani BL, Murugesan S, Martinez-Valladares M, García-Estrada C, Reguera RM, Balaña-Fouce R. Screening Marine Natural Products for New Drug Leads against Trypanosomatids and Malaria. Mar Drugs 2020; 18:E187. [PMID: 32244488 PMCID: PMC7230869 DOI: 10.3390/md18040187] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023] Open
Abstract
Neglected Tropical Diseases (NTD) represent a serious threat to humans, especially for those living in poor or developing countries. Almost one-sixth of the world population is at risk of suffering from these diseases and many thousands die because of NTDs, to which we should add the sanitary, labor and social issues that hinder the economic development of these countries. Protozoan-borne diseases are responsible for more than one million deaths every year. Visceral leishmaniasis, Chagas disease or sleeping sickness are among the most lethal NTDs. Despite not being considered an NTD by the World Health Organization (WHO), malaria must be added to this sinister group. Malaria, caused by the apicomplexan parasite Plasmodium falciparum, is responsible for thousands of deaths each year. The treatment of this disease has been losing effectiveness year after year. Many of the medicines currently in use are obsolete due to their gradual loss of efficacy, their intrinsic toxicity and the emergence of drug resistance or a lack of adherence to treatment. Therefore, there is an urgent and global need for new drugs. Despite this, the scant interest shown by most of the stakeholders involved in the pharmaceutical industry makes our present therapeutic arsenal scarce, and until recently, the search for new drugs has not been seriously addressed. The sources of new drugs for these and other pathologies include natural products, synthetic molecules or repurposing drugs. The most frequent sources of natural products are microorganisms, e.g., bacteria, fungi, yeasts, algae and plants, which are able to synthesize many drugs that are currently in use (e.g. antimicrobials, antitumor, immunosuppressants, etc.). The marine environment is another well-established source of bioactive natural products, with recent applications against parasites, bacteria and other pathogens which affect humans and animals. Drug discovery techniques have rapidly advanced since the beginning of the millennium. The combination of novel techniques that include the genetic modification of pathogens, bioimaging and robotics has given rise to the standardization of High-Performance Screening platforms in the discovery of drugs. These advancements have accelerated the discovery of new chemical entities with antiparasitic effects. This review presents critical updates regarding the use of High-Throughput Screening (HTS) in the discovery of drugs for NTDs transmitted by protozoa, including malaria, and its application in the discovery of new drugs of marine origin.
Collapse
Affiliation(s)
- María Álvarez-Bardón
- Department of Biomedical Sciences; University of León, 24071 León, Spain; (M.Á.-B.); (Y.P.-P.); (C.O.); (D.S.-C.); (R.M.R.)
| | - Yolanda Pérez-Pertejo
- Department of Biomedical Sciences; University of León, 24071 León, Spain; (M.Á.-B.); (Y.P.-P.); (C.O.); (D.S.-C.); (R.M.R.)
| | - César Ordóñez
- Department of Biomedical Sciences; University of León, 24071 León, Spain; (M.Á.-B.); (Y.P.-P.); (C.O.); (D.S.-C.); (R.M.R.)
| | - Daniel Sepúlveda-Crespo
- Department of Biomedical Sciences; University of León, 24071 León, Spain; (M.Á.-B.); (Y.P.-P.); (C.O.); (D.S.-C.); (R.M.R.)
| | - Nestor M. Carballeira
- Department of Chemistry, University of Puerto Rico, Río Piedras 00925-2537, San Juan, Puerto Rico;
| | - Babu L. Tekwani
- Department of Infectious Diseases, Division of Drug Discovery, Southern Research, Birmingham, AL 35205, USA;
| | - Sankaranarayanan Murugesan
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Vidya Vihar, Pilani 333031, India;
| | - Maria Martinez-Valladares
- Department of Animal Health, Instituto de Ganadería de Montaña (CSIC-Universidad de León), Grulleros, 24346 León, Spain;
| | - Carlos García-Estrada
- INBIOTEC (Instituto de Biotecnología de León), Avda. Real 1-Parque Científico de León, 24006 León, Spain;
| | - Rosa M. Reguera
- Department of Biomedical Sciences; University of León, 24071 León, Spain; (M.Á.-B.); (Y.P.-P.); (C.O.); (D.S.-C.); (R.M.R.)
| | - Rafael Balaña-Fouce
- Department of Biomedical Sciences; University of León, 24071 León, Spain; (M.Á.-B.); (Y.P.-P.); (C.O.); (D.S.-C.); (R.M.R.)
| |
Collapse
|
21
|
Jex AR, Svärd S, Hagen KD, Starcevich H, Emery-Corbin SJ, Balan B, Nosala C, Dawson SC. Recent advances in functional research in Giardia intestinalis. ADVANCES IN PARASITOLOGY 2020; 107:97-137. [PMID: 32122532 PMCID: PMC7878119 DOI: 10.1016/bs.apar.2019.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This review considers current advances in tools to investigate the functional biology of Giardia, it's coding and non-coding genes, features and cellular and molecular biology. We consider major gaps in current knowledge of the parasite and discuss the present state-of-the-art in its in vivo and in vitro cultivation. Advances in in silico tools, including for the modelling non-coding RNAs and genomic elements, as well as detailed exploration of coding genes through inferred homology to model organisms, have provided significant, primary level insight. Improved methods to model the three-dimensional structure of proteins offer new insights into their function, and binding interactions with ligands, other proteins or precursor drugs, and offer substantial opportunities to prioritise proteins for further study and experimentation. These approaches can be supplemented by the growing and highly accessible arsenal of systems-based methods now being applied to Giardia, led by genomic, transcriptomic and proteomic methods, but rapidly incorporating advanced tools for detection of real-time transcription, evaluation of chromatin states and direct measurement of macromolecular complexes. Methods to directly interrogate and perturb gene function have made major leaps in recent years, with CRISPr-interference now available. These approaches, coupled with protein over-expression, fluorescent labelling and in vitro and in vivo imaging, are set to revolutionize the field and herald an exciting time during which the field may finally realise Giardia's long proposed potential as a model parasite and eukaryote.
Collapse
Affiliation(s)
- Aaron R Jex
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, VIC, Australia; Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia.
| | - Staffan Svärd
- Centre for Biomedicine, Uppsala University, Uppsala, Sweden
| | - Kari D Hagen
- College of Biological Sciences, University of California-Davis, Davis, CA, United States
| | - Hannah Starcevich
- College of Biological Sciences, University of California-Davis, Davis, CA, United States
| | - Samantha J Emery-Corbin
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, VIC, Australia
| | - Balu Balan
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, VIC, Australia
| | - Chris Nosala
- College of Biological Sciences, University of California-Davis, Davis, CA, United States
| | - Scott C Dawson
- College of Biological Sciences, University of California-Davis, Davis, CA, United States
| |
Collapse
|
22
|
Archer NK, Wang Y, Ortines RV, Liu H, Nolan SJ, Liu Q, Alphonse MP, Dikeman DA, Mazhar M, Miller RJ, Anderson LS, Francis KP, Simon SI, Miller LS. Preclinical Models and Methodologies for Monitoring Staphylococcus aureus Infections Using Noninvasive Optical Imaging. Methods Mol Biol 2020; 2069:197-228. [PMID: 31523776 PMCID: PMC7745539 DOI: 10.1007/978-1-4939-9849-4_15] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In vivo whole-animal optical (bioluminescence and fluorescence) imaging of Staphylococcus aureus infections has provided the opportunity to noninvasively and longitudinally monitor the dynamics of the bacterial burden and ensuing host immune responses in live anesthetized animals. Herein, we describe several different mouse models of S. aureus skin infection, skin inflammation, incisional/excisional wound infections, as well as mouse and rabbit models of orthopedic implant infection, which utilized this imaging technology. These animal models and imaging methodologies provide insights into the pathogenesis of these infections and innate and adaptive immune responses, as well as the preclinical evaluation of diagnostic and treatment modalities. Noninvasive approaches to investigate host-pathogen interactions are extremely important as virulent community-acquired methicillin-resistant S. aureus strains (CA-MRSA) are spreading through the normal human population, becoming more antibiotic resistant and creating a serious threat to public health.
Collapse
Affiliation(s)
- Nathan K Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yu Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Roger V Ortines
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Haiyun Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sabrina J Nolan
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Qi Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Martin P Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dustin A Dikeman
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Momina Mazhar
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert J Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Leif S Anderson
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | | | - Scott I Simon
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
23
|
Zhao N, Liu JM, Liu S, Ji XM, Lv H, Hu YZ, Wang ZH, Lv SW, Li CY, Wang S. A novel universal nano-luciferase-involved reporter system for long-term probing food-borne probiotics and pathogenic bacteria in mice by in situ bioluminescence imaging. RSC Adv 2020; 10:13029-13036. [PMID: 35492135 PMCID: PMC9051406 DOI: 10.1039/d0ra01283a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 03/23/2020] [Indexed: 01/18/2023] Open
Abstract
Food-borne bacteria have received increasing attention due to their great impact on human health. Bioimaging makes it possible to monitor bacteria inside the living body in real time and in situ. Nano-luciferase (NLuc) as a new member of the luciferase family exhibits superior properties than the commonly used luciferases, including small size, high stability and improved luminescence. Herein, NLuc, CBRLuc and FLuc were well expressed in varied food-borne bacteria. Results showed that the signal intensity of E. coli-NLuc was about 41 times higher than E. coli-CBRLuc, L. plantarum-NLuc was nearly 227 times that of L. plantarum-FLuc in vitro. Moreover, NLuc was applied to trace L. plantarum and E. coli in vivo through the whole body and separated digestive tract imaging, as well as the feces bacterium counting and probing. The persistence of bioluminescent strains was predominantly localized in colon and cecum of mice after oral administration. The NLuc system showed its incomparable superiority, especially in the application of intestinal imaging and the universality for food-borne bacteria. We demonstrated that the NLuc system was a brilliant alternative for specific application of food-borne bacteria in vivo, aiming to collect more accurate and real-time information of food-borne bacteria from the living body for further investigation of their damage mechanism and nutrition effect. Schematic illustration of the preparation of bioluminescent bacteria and the experimental design of tracing of the foodborne bacteria in vivo.![]()
Collapse
|
24
|
Rodrigues CJC, Sanches JM, de Carvalho CCCR. Determining transaminase activity in bacterial libraries by time-lapse imaging. Chem Commun (Camb) 2019; 55:13538-13541. [PMID: 31647085 DOI: 10.1039/c9cc07507k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Transaminase activity was determined by time-lapse imaging using a colourimetric reaction and image analysis. A correlation between the benzaldehyde conversion and relative luminance was determined, allowing the identification of the most promising biocatalysts, the determination of kinetic parameters, and the assessment of the effect of the substrate concentration on activity.
Collapse
Affiliation(s)
- Carlos J C Rodrigues
- iBB-Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.
| | - João M Sanches
- Institute for Systems and Robotics (ISR/IST), LARSyS, Bioengineering Department, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Carla C C R de Carvalho
- iBB-Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
25
|
Thompson JM, Miller LS. Preclinical Optical Imaging to Study Pathogenesis, Novel Therapeutics and Diagnostics Against Orthopaedic Infection. J Orthop Res 2019; 37:2269-2277. [PMID: 31342546 DOI: 10.1002/jor.24428] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/15/2019] [Indexed: 02/04/2023]
Abstract
Preclinical in vivo optical imaging includes bioluminescence imaging (BLI) and fluorescence imaging (FLI), which provide noninvasive and longitudinal monitoring of biological processes in an in vivo context. In vivo BLI involves the detection of photons of light from bioluminescent bacteria engineered to naturally emit light in preclinical animal models of infection. Meanwhile, in vivo FLI involves the detection of photons of a longer emission wavelength of light after exposure of a fluorophore to a shorter excitation wavelength of light. In vivo FLI has been used in preclinical animal models to detect fluorescent-labeled host proteins or cells (often in engineered fluorescent reporter mice) to understand host-related processes, or to detect injectable near-infrared fluorescent probes as a novel approach for diagnosing infection. This review describes the use of in vivo optical imaging in preclinical models of orthopaedic implant-associated infection (OIAI), including (i) pathogenesis of the infectious course, (ii) monitoring efficacy of antimicrobial prophylaxis and therapy and (iii) evaluating novel near-infrared fluorescent probes for diagnosing infection. Finally, we describe optoacoustic imaging and fluorescence image-guided surgery, which are recent technologies that have the potential to translate to diagnosing and treating OIAI in humans. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:2269-2277, 2019.
Collapse
Affiliation(s)
- John M Thompson
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287
| | - Lloyd S Miller
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21231
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, 21218
| |
Collapse
|
26
|
Monitoring Tuberculosis Drug Activity in Live Animals by Near-Infrared Fluorescence Imaging. Antimicrob Agents Chemother 2019:AAC.01280-19. [PMID: 31527027 DOI: 10.1128/aac.01280-19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Worldwide, tuberculosis (TB) is the leading cause of death due to infection with a single pathogenic agent, Mycobacterium tuberculosis In the absence of an effective vaccine, new, more powerful antibiotics are required to halt the growing spread of multidrug-resistant strains and to shorten the duration of TB treatment. However, assessing drug efficacy at the preclinical stage remains a long and fastidious procedure that delays progression of drugs down the pipeline and towards the clinic. In this investigation, we report the construction, optimization and characterization of genetically engineered near-infrared (NIR) fluorescent reporter strains of the pathogens Mycobacterium marinum and Mycobacterium tuberculosis that enable direct visualization of bacteria in infected zebrafish and mice, respectively. Fluorescence could be measured precisely in infected immunodeficient mice, while its intensity appeared to be below the limit of detection in immunocompetent mice, probably because of the lower bacterial load obtained in these animals. Furthermore, we show that the fluorescence level accurately reflects the bacterial load, as determined by colony forming unit (CFU) enumeration, thus enabling the efficacy of antibiotic treatment to be assessed in live animals in real time. The NIR fluorescent imaging system disclosed here is a valuable resource for TB research and can serve to accelerate drug development.
Collapse
|
27
|
Castañeda CD, McDaniel CD, Abdelhamed H, Karsi A, Kiess AS. Evaluating bacterial colonization of a developing broiler embryo after in ovo injection with a bioluminescent bacteria. Poult Sci 2019; 98:2997-3006. [PMID: 30789222 DOI: 10.3382/ps/pez053] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 02/06/2019] [Indexed: 02/04/2023] Open
Abstract
In ovo injection of probiotics has been of interest for achieving early health benefits. However, there is limited research demonstrating where bacteria could migrate within the embryo after injection. The objective of this study was to evaluate bacterial colonization or migration after in ovo injection of broiler embryo with bioluminescent Escherichia coli. Injection using 106 CFU/mL nonpathogenic E. coli was applied to amniotic and air cell regions on day 18 of incubation. On days 18, 19, 20, and 21 the amnion, skin, lung, gastrointestinal tract (GIT), bursa, and spleen were collected. On day 21, the GIT was separated into crop, duodenum, jejunum, ileum, and ceca sections. All tissues were visualized using anin vivo imaging system to confirm the presence of bioluminescent E. coli. Samples were homogenized, 10-fold serially diluted, and spread onto appropriate agar to determine bacterial loads in all tissues. Results indicated that eggs injected into the amnion had significantly high numbers of E. coli cells in all tissues compared to air cell injected and control treatments 2 h post-injection (P < 0.0001). E. coli was also found on the lungs, spleen, and bursa of eggs injected either in the amnion or air cell (P < 0.05). Results indicated that in ovo injection into the amnion was more efficient than air cell injection, yielding a higher bacterial concentration in the evaluated tissues, specifically the ileum and ceca. Future research using bioluminescent probiotic bacteria may establish sites of preference for different probiotics leading to site-specific application that can maximize their overall impact when in ovo injected.
Collapse
Affiliation(s)
- Claudia D Castañeda
- Department of Poultry Science, Mississippi State University, Mississippi State MS 39762
| | - Christopher D McDaniel
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State MS 39762
| | - Hossam Abdelhamed
- Department of Poultry Science, Mississippi State University, Mississippi State MS 39762
| | - Attila Karsi
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State MS 39762
| | - Aaron S Kiess
- Department of Poultry Science, Mississippi State University, Mississippi State MS 39762
| |
Collapse
|
28
|
Vanherp L, Ristani A, Poelmans J, Hillen A, Lagrou K, Janbon G, Brock M, Himmelreich U, Vande Velde G. Sensitive bioluminescence imaging of fungal dissemination to the brain in mouse models of cryptococcosis. Dis Model Mech 2019; 12:dmm.039123. [PMID: 31101657 PMCID: PMC6602310 DOI: 10.1242/dmm.039123] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/08/2019] [Indexed: 12/18/2022] Open
Abstract
Cryptococcus neoformans is a leading cause of fungal brain infection, but the mechanism of dissemination and dynamics of cerebral infection following pulmonary disease are poorly understood. To address these questions, non-invasive techniques that can study the dynamic processes of disease development and progression in living animal models or patients are required. As such, bioluminescence imaging (BLI) has emerged as a powerful tool to evaluate the spatial and temporal distribution of infection in living animals. We aimed to study the time profile of the dissemination of cryptococcosis from the lung to the brain in murine models by engineering the first bioluminescent C. neoformans KN99α strain, expressing a sequence-optimized red-shifted luciferase. The high pathogen specificity and sensitivity of BLI was complemented by the three-dimensional anatomical information from micro-computed tomography (μCT) of the lung and magnetic resonance imaging (MRI) of the brain. These non-invasive imaging techniques provided longitudinal readouts on the spatial and temporal distribution of infection following intravenous, intranasal or endotracheal routes of inoculation. Furthermore, the imaging results correlated strongly with the fungal load in the respective organs. By obtaining dynamic and quantitative information about the extent and timing of brain infections for individual animals, we found that dissemination to the brain after primary infection of the lung is likely a late-stage event with a timeframe that is variable between animals. This novel tool in Cryptococcus research can aid the identification of host and pathogen factors involved in this process, and supports development of novel preventive or therapeutic approaches. Summary: A novel combination of bioluminescence and anatomical imaging non-invasively identified the timeframe and extent of Cryptococcus neoformans dissemination to the brain in animal models of systemic and pulmonary fungal infection.
Collapse
Affiliation(s)
- Liesbeth Vanherp
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium.,Molecular Small Animal Imaging Center (MoSAIC), KU Leuven, 3000 Leuven, Belgium
| | - Alexandra Ristani
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium.,Molecular Small Animal Imaging Center (MoSAIC), KU Leuven, 3000 Leuven, Belgium
| | - Jennifer Poelmans
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium.,Molecular Small Animal Imaging Center (MoSAIC), KU Leuven, 3000 Leuven, Belgium
| | - Amy Hillen
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium.,Molecular Small Animal Imaging Center (MoSAIC), KU Leuven, 3000 Leuven, Belgium
| | - Katrien Lagrou
- Laboratory of Clinical Bacteriology and Mycology, Department of Microbiology and Immunology, KU Leuven, 3000 Leuven, Belgium
| | - Guilhem Janbon
- RNA Biology of Fungal Pathogens, Department of Mycology, Pasteur Institute, Paris 75015, France
| | - Matthias Brock
- Fungal Biology Group, School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Uwe Himmelreich
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium.,Molecular Small Animal Imaging Center (MoSAIC), KU Leuven, 3000 Leuven, Belgium
| | - Greetje Vande Velde
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium .,Molecular Small Animal Imaging Center (MoSAIC), KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
29
|
Yeh HW, Ai HW. Development and Applications of Bioluminescent and Chemiluminescent Reporters and Biosensors. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2019; 12:129-150. [PMID: 30786216 PMCID: PMC6565457 DOI: 10.1146/annurev-anchem-061318-115027] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Although fluorescent reporters and biosensors have become indispensable tools in biological and biomedical fields, fluorescence measurements require external excitation light, thereby limiting their use in thick tissues and live animals. Bioluminescent reporters and biosensors may potentially overcome this hurdle because they use enzyme-catalyzed exothermic biochemical reactions to generate excited-state emitters. This review first introduces the development of bioluminescent reporters, and next, their applications in sensing biological changes in vitro and in vivo as biosensors. Lastly, we discuss chemiluminescent sensors that produce photons in the absence of luciferases. This review aims to explore fundamentals and experimental insights and to emphasize the yet-to-be-reached potential of next-generation luminescent reporters and biosensors.
Collapse
Affiliation(s)
- Hsien-Wei Yeh
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, and Department of Chemistry, University of Virginia, Charlottesville, Virginia 22908, USA;
| | - Hui-Wang Ai
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, and Department of Chemistry, University of Virginia, Charlottesville, Virginia 22908, USA;
| |
Collapse
|
30
|
Balaña-Fouce R, Pérez Pertejo MY, Domínguez-Asenjo B, Gutiérrez-Corbo C, Reguera RM. Walking a tightrope: drug discovery in visceral leishmaniasis. Drug Discov Today 2019; 24:1209-1216. [PMID: 30876846 DOI: 10.1016/j.drudis.2019.03.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/29/2019] [Accepted: 03/06/2019] [Indexed: 12/14/2022]
Abstract
The current commitment of the pharma industry, nongovernmental organizations and academia to find better treatments against neglected tropical diseases should end decades of challenge caused by these global scourges. The initial result of these efforts has been the introduction of enhanced combinations of drugs, currently in clinical use, or formulations thereof. Phenotypic screening based on intracellular parasite infections has been revealed as the first key tool of antileishmanial drug discovery, because most first-in-class drugs entering Phase I trials were discovered this way. The professional commitment among stakeholders has enabled the availability of a plethora of new chemical entities that fit the target product profile for these diseases. However, the rate of hit discovery in leishmaniasis is far behind that for other neglected diseases. This review defends the need to develop new screening methods that consider the part played not only by intracellular parasites but also by the host's immune system to generate disease-relevant assays and improve clinical outcomes.
Collapse
Affiliation(s)
- Rafael Balaña-Fouce
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana, E-24071 León, Spain
| | - M Yolanda Pérez Pertejo
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana, E-24071 León, Spain
| | - Bárbara Domínguez-Asenjo
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana, E-24071 León, Spain
| | - Camino Gutiérrez-Corbo
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana, E-24071 León, Spain
| | - Rosa M Reguera
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana, E-24071 León, Spain.
| |
Collapse
|
31
|
Miller RJ, Thompson JM, Zheng J, Marchitto MC, Archer NK, Pinsker BL, Ortines RV, Jiang X, Martin RA, Brown ID, Wang Y, Sterling RS, Mao HQ, Miller LS. In Vivo Bioluminescence Imaging in a Rabbit Model of Orthopaedic Implant-Associated Infection to Monitor Efficacy of an Antibiotic-Releasing Coating. J Bone Joint Surg Am 2019; 101:e12. [PMID: 30801375 PMCID: PMC6738548 DOI: 10.2106/jbjs.18.00425] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND In vivo bioluminescence imaging (BLI) provides noninvasive monitoring of bacterial burden in animal models of orthopaedic implant-associated infection (OIAI). However, technical limitations have limited its use to mouse and rat models of OIAI. The goal of this study was to develop a larger, rabbit model of OIAI using in vivo BLI to evaluate the efficacy of an antibiotic-releasing implant coating. METHODS A nanofiber coating loaded with or without linezolid-rifampin was electrospun onto a surgical-grade locking peg. To model OIAI in rabbits, a medial parapatellar arthrotomy was performed to ream the femoral canal, and a bright bioluminescent methicillin-resistant Staphylococcus aureus (MRSA) strain was inoculated into the canal, followed by retrograde insertion of the coated implant flush with the articular surface. In vivo BLI signals were confirmed by ex vivo colony-forming units (CFUs) from tissue, bone, and implant specimens. RESULTS In this rabbit model of OIAI (n = 6 rabbits per group), implants coated without antibiotics were associated with significantly increased knee width and in vivo BLI signals compared with implants coated with linezolid-rifampin (p < 0.001 and p < 0.05, respectively). On day 7, the implants without antibiotics were associated with significantly increased CFUs from tissue (mean [and standard error of the mean], 1.4 × 10 ± 2.1 × 10 CFUs; p < 0.001), bone (6.9 × 10 ± 3.1 × 10 CFUs; p < 0.05), and implant (5.1 × 10 ± 2.2 × 10 CFUs; p < 0.05) specimens compared with implants with linezolid-rifampin, which demonstrated no detectable CFUs from any source. CONCLUSIONS By combining a bright bioluminescent MRSA strain with modified techniques, in vivo BLI in a rabbit model of OIAI demonstrated the efficacy of an antibiotic-releasing coating. CLINICAL RELEVANCE The new capability of in vivo BLI for noninvasive monitoring of bacterial burden in larger-animal models of OIAI may have important preclinical relevance.
Collapse
Affiliation(s)
- Robert J. Miller
- Departments of Dermatology (R.J.M., M.C.M., N.K.A., B.L.P., R.V.O., I.D.B., Y.W., and L.S.M.) and Orthopaedic Surgery (J.M.T., R.S.S., and L.S.M.) and Division of Infectious Diseases, Department of Medicine (L.S.M.), Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - John M. Thompson
- Departments of Dermatology (R.J.M., M.C.M., N.K.A., B.L.P., R.V.O., I.D.B., Y.W., and L.S.M.) and Orthopaedic Surgery (J.M.T., R.S.S., and L.S.M.) and Division of Infectious Diseases, Department of Medicine (L.S.M.), Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jesse Zheng
- Departments of Biomedical Engineering (J.Z.) and Materials Science and Engineering (X.J., R.A.M., H.-Q.M., and L.S.M.), Translational Tissue Engineering Center (X.J., R.A.M., H.-Q.M., and L.S.M.), Institute for NanoBioTechnology (X.J., R.A.M., and H.-Q.M.), and Whitaker Biomedical Engineering Institute (H.-Q.M.), Johns Hopkins University, Baltimore, Maryland
| | - Mark C. Marchitto
- Departments of Dermatology (R.J.M., M.C.M., N.K.A., B.L.P., R.V.O., I.D.B., Y.W., and L.S.M.) and Orthopaedic Surgery (J.M.T., R.S.S., and L.S.M.) and Division of Infectious Diseases, Department of Medicine (L.S.M.), Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nathan K. Archer
- Departments of Dermatology (R.J.M., M.C.M., N.K.A., B.L.P., R.V.O., I.D.B., Y.W., and L.S.M.) and Orthopaedic Surgery (J.M.T., R.S.S., and L.S.M.) and Division of Infectious Diseases, Department of Medicine (L.S.M.), Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Bret L. Pinsker
- Departments of Dermatology (R.J.M., M.C.M., N.K.A., B.L.P., R.V.O., I.D.B., Y.W., and L.S.M.) and Orthopaedic Surgery (J.M.T., R.S.S., and L.S.M.) and Division of Infectious Diseases, Department of Medicine (L.S.M.), Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Roger V. Ortines
- Departments of Dermatology (R.J.M., M.C.M., N.K.A., B.L.P., R.V.O., I.D.B., Y.W., and L.S.M.) and Orthopaedic Surgery (J.M.T., R.S.S., and L.S.M.) and Division of Infectious Diseases, Department of Medicine (L.S.M.), Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xuesong Jiang
- Departments of Biomedical Engineering (J.Z.) and Materials Science and Engineering (X.J., R.A.M., H.-Q.M., and L.S.M.), Translational Tissue Engineering Center (X.J., R.A.M., H.-Q.M., and L.S.M.), Institute for NanoBioTechnology (X.J., R.A.M., and H.-Q.M.), and Whitaker Biomedical Engineering Institute (H.-Q.M.), Johns Hopkins University, Baltimore, Maryland
| | - Russell A. Martin
- Departments of Biomedical Engineering (J.Z.) and Materials Science and Engineering (X.J., R.A.M., H.-Q.M., and L.S.M.), Translational Tissue Engineering Center (X.J., R.A.M., H.-Q.M., and L.S.M.), Institute for NanoBioTechnology (X.J., R.A.M., and H.-Q.M.), and Whitaker Biomedical Engineering Institute (H.-Q.M.), Johns Hopkins University, Baltimore, Maryland
| | - Isabelle D. Brown
- Departments of Dermatology (R.J.M., M.C.M., N.K.A., B.L.P., R.V.O., I.D.B., Y.W., and L.S.M.) and Orthopaedic Surgery (J.M.T., R.S.S., and L.S.M.) and Division of Infectious Diseases, Department of Medicine (L.S.M.), Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yu Wang
- Departments of Dermatology (R.J.M., M.C.M., N.K.A., B.L.P., R.V.O., I.D.B., Y.W., and L.S.M.) and Orthopaedic Surgery (J.M.T., R.S.S., and L.S.M.) and Division of Infectious Diseases, Department of Medicine (L.S.M.), Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert S. Sterling
- Departments of Dermatology (R.J.M., M.C.M., N.K.A., B.L.P., R.V.O., I.D.B., Y.W., and L.S.M.) and Orthopaedic Surgery (J.M.T., R.S.S., and L.S.M.) and Division of Infectious Diseases, Department of Medicine (L.S.M.), Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hai-Quan Mao
- Departments of Biomedical Engineering (J.Z.) and Materials Science and Engineering (X.J., R.A.M., H.-Q.M., and L.S.M.), Translational Tissue Engineering Center (X.J., R.A.M., H.-Q.M., and L.S.M.), Institute for NanoBioTechnology (X.J., R.A.M., and H.-Q.M.), and Whitaker Biomedical Engineering Institute (H.-Q.M.), Johns Hopkins University, Baltimore, Maryland
| | - Lloyd S. Miller
- Departments of Dermatology (R.J.M., M.C.M., N.K.A., B.L.P., R.V.O., I.D.B., Y.W., and L.S.M.) and Orthopaedic Surgery (J.M.T., R.S.S., and L.S.M.) and Division of Infectious Diseases, Department of Medicine (L.S.M.), Johns Hopkins University School of Medicine, Baltimore, Maryland,Departments of Biomedical Engineering (J.Z.) and Materials Science and Engineering (X.J., R.A.M., H.-Q.M., and L.S.M.), Translational Tissue Engineering Center (X.J., R.A.M., H.-Q.M., and L.S.M.), Institute for NanoBioTechnology (X.J., R.A.M., and H.-Q.M.), and Whitaker Biomedical Engineering Institute (H.-Q.M.), Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
32
|
Kendall LV, Owiny JR, Dohm ED, Knapek KJ, Lee ES, Kopanke JH, Fink M, Hansen SA, Ayers JD. Replacement, Refinement, and Reduction in Animal Studies With Biohazardous Agents. ILAR J 2019; 59:177-194. [DOI: 10.1093/ilar/ily021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 06/11/2018] [Indexed: 12/17/2022] Open
Abstract
Abstract
Animal models are critical to the advancement of our knowledge of infectious disease pathogenesis, diagnostics, therapeutics, and prevention strategies. The use of animal models requires thoughtful consideration for their well-being, as infections can significantly impact the general health of an animal and impair their welfare. Application of the 3Rs—replacement, refinement, and reduction—to animal models using biohazardous agents can improve the scientific merit and animal welfare. Replacement of animal models can use in vitro techniques such as cell culture systems, mathematical models, and engineered tissues or invertebrate animal hosts such as amoeba, worms, fruit flies, and cockroaches. Refinements can use a variety of techniques to more closely monitor the course of disease. These include the use of biomarkers, body temperature, behavioral observations, and clinical scoring systems. Reduction is possible using advanced technologies such as in vivo telemetry and imaging, allowing longitudinal assessment of animals during the course of disease. While there is no single method to universally replace, refine, or reduce animal models, the alternatives and techniques discussed are broadly applicable and they should be considered when infectious disease animal models are developed.
Collapse
Affiliation(s)
- Lon V Kendall
- Department of Microbiology, Immunology and Pathology, and Laboratory Animal Resources, Colorado State University, Fort Collins, Colorado
| | - James R Owiny
- Laboratory Animal Resources, Colorado State University, Fort Collins, Colorado
| | - Erik D Dohm
- Animal Resources Program, University of Alabama, Birmingham, Alabama
| | - Katie J Knapek
- Comparative Medicine Training Program, Colorado State University, Fort Collins, Colorado
| | - Erin S Lee
- Animal Resource Center, University of Texas Medical Branch, Galveston, Texas
| | - Jennifer H Kopanke
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado
| | - Michael Fink
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri
| | - Sarah A Hansen
- Office of Animal Resources, University of Iowa, Iowa City, Iowa
| | - Jessica D Ayers
- Laboratory Animal Resources, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
33
|
Perdiguero B, Gómez CE, Esteban M. Bioluminescence Imaging as a Tool for Poxvirus Biology. Methods Mol Biol 2019; 2023:269-285. [PMID: 31240684 DOI: 10.1007/978-1-4939-9593-6_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Bioluminescence imaging, with luciferase as a reporter-encoding gene, has been successfully and widely used for studies to follow viral infection in an organism and to measure therapeutic efficacy of antiviral agents in small animal models. Bioluminescence is produced by the reaction of a luciferase enzyme stably inserted into the viral genome with a defined substrate systemically delivered into the animal. The light emitted is captured allowing the detection of viral infection sites and the quantification of viral replication in the context of tissues of a living animal. The goal of this chapter is to provide a technical background for the evaluation of poxvirus infection in cells and animals through bioluminescence imaging technology using luciferase-expressing recombinant poxviruses.
Collapse
Affiliation(s)
- Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Madrid, Spain
| | - Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Madrid, Spain.
| |
Collapse
|
34
|
Hernandez-Quintanar L, Rodriguez-Salvador M. Discovering new 3D bioprinting applications: Analyzing the case of optical tissue phantoms. Int J Bioprint 2018; 5:178. [PMID: 32596533 PMCID: PMC7294689 DOI: 10.18063/ijb.v5i1.178] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 12/18/2018] [Indexed: 11/25/2022] Open
Abstract
Optical tissue phantoms enable to mimic the optical properties of biological tissues for biomedical device calibration, new equipment validation, and clinical training for the detection, and treatment of diseases. Unfortunately, current methods for their development present some problems, such as a lack of repeatability in their optical properties. Where the use of three-dimensional (3D) printing or 3D bioprinting could address these issues. This paper aims to evaluate the use of this technology in the development of optical tissue phantoms. A competitive technology intelligence methodology was applied by analyzing Scopus, Web of Science, and patents from January 1, 2000, to July 31, 2018. The main trends regarding methods, materials, and uses, as well as predominant countries, institutions, and journals, were determined. The results revealed that, while 3D printing is already employed (in total, 108 scientific papers and 18 patent families were identified), 3D bioprinting is not yet applied for optical tissue phantoms. Nevertheless, it is expected to have significant growth. This research gives biomedical scientists a new window of opportunity for exploring the use of 3D bioprinting in a new area that may support testing of new equipment and development of techniques for the diagnosis and treatment of diseases.
Collapse
|
35
|
Van Zyl WF, Deane SM, Dicks LMT. In vivo bioluminescence imaging of the spatial and temporal colonization of lactobacillus plantarum 423 and enterococcus mundtii ST4SA in the intestinal tract of mice. BMC Microbiol 2018; 18:171. [PMID: 30376820 PMCID: PMC6208077 DOI: 10.1186/s12866-018-1315-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 10/14/2018] [Indexed: 12/23/2022] Open
Abstract
Background Lactic acid bacteria (LAB) are major inhabitants and part of the normal microflora of the gastrointestinal tract (GIT) of humans and animals. Despite substantial evidence supporting the beneficial properties of LAB, only a few studies have addressed the migration and colonization of probiotic bacteria in the GIT. The reason for this is mostly due to the limitations, or lack of, efficient reporter systems. Here we describe the development and application of a non-invasive in vivo bioluminescence reporter system to study, in real-time, the spatial and temporal persistence of Lactobacillus plantarum 423 and Enterococcus mundtii ST4SA in the intestinal tract of mice. Results This study reports on the application of the firefly luciferase gene (ffluc) from Photinus pyralis to develop luciferase-expressing L. plantarum 423 and E. mundtii ST4SA, using a Lactococcus lactis NICE system on a high copy number plasmid (pNZ8048) and strong constitutive lactate dehydrogenase gene promoters (Pldh and STldh). The reporter system was used for in vivo and ex vivo monitoring of both probiotic LAB strains in the GIT of mice after single and multiple oral administrations. Enterococcus mundtii ST4SA reached the large intestine 45 min after gavage, while L. plantarum 423 reached the cecum/colon after 90 min. Both strains predominantly colonized the cecum and colon after five consecutive daily administrations. Enterococcus mundtii ST4SA persisted in faeces at higher numbers and for more days compared to L. plantarum 423. Conclusions Our findings demonstrate the efficiency of a high-copy number vector, constitutive promoters and bioluminescence imaging to study the colonization and persistence of L. plantarum 423 and E. mundtii ST4SA in the murine GIT. The system allowed us to differentiate between intestinal transit times of the two strains in the digestive tract. This is the first report of bioluminescence imaging of a luciferase-expressing E. mundtii strain to study colonization dynamics in the murine model. The bioluminescence system developed in this study may be used to study the in vivo colonization dynamics of other probiotic LAB. Electronic supplementary material The online version of this article (10.1186/s12866-018-1315-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Winschau F Van Zyl
- Department of Microbiology, Stellenbosch University, Private Bag X1, Matieland, Stellenbosch, 7600, South Africa
| | - Shelly M Deane
- Department of Microbiology, Stellenbosch University, Private Bag X1, 7 Matieland, Stellenbosch, 7602, South Africa
| | - Leon M T Dicks
- Department of Microbiology, Stellenbosch University, Private Bag X1, 7 Matieland, Stellenbosch, 7602, South Africa.
| |
Collapse
|
36
|
Dehghani H, Guggenheim JA, Taylor SL, Xu X, Wang KKH. Quantitative bioluminescence tomography using spectral derivative data. BIOMEDICAL OPTICS EXPRESS 2018; 9:4163-4174. [PMID: 30615705 PMCID: PMC6157772 DOI: 10.1364/boe.9.004163] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/23/2018] [Accepted: 07/31/2018] [Indexed: 05/15/2023]
Abstract
Bioluminescence imaging (BLI) is a non-contact, optical imaging technique based on measurement of emitted light due to an internal source, which is then often directly related to cellular activity. It is widely used in pre-clinical small animal imaging studies to assess the progression of diseases such as cancer, aiding in the development of new treatments and therapies. For many applications, the quantitative assessment of accurate cellular activity and spatial distribution is desirable as it would enable direct monitoring for prognostic evaluation. This requires quantitative spatially-resolved measurements of bioluminescence source strength inside the animal to be obtained from BLI images. This is the goal of bioluminescence tomography (BLT) in which a model of light propagation through tissue is combined with an optimization algorithm to reconstruct a map of the underlying source distribution. As most models consider only the propagation of light from internal sources to the animal skin surface, an additional challenge is accounting for the light propagation from the skin to the optical detector (e.g. camera). Existing approaches typically use a model of the imaging system optics (e.g. ray-tracing, analytical optical models) or approximate corrections derived from calibration measurements. However, these approaches are typically computationally intensive or of limited accuracy. In this work, a new approach is presented in which, rather than directly using BLI images acquired at several wavelengths, the spectral derivative of that data (difference of BLI images at adjacent wavelengths) is used in BLT. As light at similar wavelengths encounters a near-identical system response (path through the optics etc.) this eliminates the need for additional corrections or system models. This approach is applied to BLT with simulated and experimental phantom data and shown that the error in reconstructed source intensity is reduced from 49% to 4%. Qualitatively, the accuracy of source localization is improved in both simulated and experimental data, as compared to reconstruction using the standard approach. The outlined algorithm can widely be adapted to all commercial systems without any further technological modifications.
Collapse
Affiliation(s)
- Hamid Dehghani
- School of Computer Science, University of Birmingham, Birmingham, B15 2TT, UK
| | - James A. Guggenheim
- Department of Medical Physics & Biomedical Engineering, University College London, London, UK
| | - Shelley L. Taylor
- School of Computer Science, University of Birmingham, Birmingham, B15 2TT, UK
| | - Xiangkun Xu
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Ken Kang-Hsin Wang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
37
|
Stable Expression of Modified Green Fluorescent Protein in Group B Streptococci To Enable Visualization in Experimental Systems. Appl Environ Microbiol 2018; 84:AEM.01262-18. [PMID: 30006391 DOI: 10.1128/aem.01262-18] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 07/04/2018] [Indexed: 12/17/2022] Open
Abstract
Group B streptococcus (GBS) is a Gram-positive bacterium associated with various diseases in humans and animals. Many studies have examined GBS physiology, virulence, and microbe-host interactions using diverse imaging approaches, including fluorescence microscopy. Strategies to label and visualize GBS using fluorescence biomarkers have been limited to antibody-based methods or nonspecific stains that bind DNA or protein; an effective plasmid-based system to label GBS with a fluorescence biomarker would represent a useful visualization tool. In this study, we developed and validated a green fluorescent protein (GFP)-variant-expressing plasmid, pGU2664, which can be applied as a marker to visualize GBS in experimental studies. The synthetic constitutively active CP25 promoter drives strong and stable expression of the GFPmut3 biomarker in GBS strains carrying pGU2664. GBS maintains GFPmut3 activity at different phases of growth. The application of fluorescence polarization enables easy discrimination of GBS GFPmut3 activity from the autofluorescence of culture media commonly used to grow GBS. Differential interference contrast microscopy, in combination with epifluorescence microscopy to detect GFPmut3 in GBS, enabled visualization of bacterial attachment to live human epithelial cells in real time. Plasmid pGU2664 was also used to visualize phenotypic differences in the adherence of wild-type GBS and an isogenic gene-deficient mutant strain lacking CovR (the control of virulence regulator) in adhesion assays. The system for GFPmut3 expression in GBS described in this study provides a new tool for the visualization of this organism in diverse research applications. We discuss the advantages and consider the limitations of this fluorescent biomarker system developed for GBS.IMPORTANCE Group B streptococcus (GBS) is a bacterium associated with various diseases in humans and animals. This study describes the development of a strategy to label and visualize GBS using a fluorescence biomarker, termed GFPmut3. We show that this biomarker can be successfully applied to track the growth of bacteria in liquid medium, and it enables the detailed visualization of GBS in the context of live human cells in real-time microscopic analysis. The system for GFPmut3 expression in GBS described in this study provides a new tool for the visualization of this organism in diverse research applications.
Collapse
|
38
|
Lamb LE, Zhi X, Alam F, Pyzio M, Scudamore CL, Wiles S, Sriskandan S. Modelling invasive group A streptococcal disease using bioluminescence. BMC Microbiol 2018; 18:60. [PMID: 29921240 PMCID: PMC6006931 DOI: 10.1186/s12866-018-1200-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 06/01/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The development of vaccines and evaluation of novel treatment strategies for invasive group A streptococcal (iGAS) disease requires suitable models of human infection that can be monitored longitudinally and are preferably non-invasive. Bio-photonic imaging provides an opportunity to reduce use of animals in infection modelling and refine the information that can be obtained, however the range of bioluminescent GAS strains available is limited. In this study we set out to develop bioluminescent iGAS strains for use in in vivo pneumonia and soft tissue disease models. RESULTS Using clinical emm1, emm3, and emm89 GAS strains that were transformed with constructs carrying the luxABCDE operon, growth and bioluminescence of transformed strains were characterised in vitro and in vivo. Emm3 and emm89 strains expressed detectable bioluminescence when transformed with a replicating plasmid and light production correlated with viable bacterial counts in vitro, however plasmid instability precluded use in the absence of antimicrobial pressure. Emm89 GAS transformed with an integrating construct demonstrated stable bioluminescence that was maintained in the absence of antibiotics. Bioluminescence of the emm89 strain correlated with viable bacterial counts both in vitro and immediately following infection in vivo. Although bioluminescence conferred a detectable fitness burden to the emm89 strain during soft tissue infection in vivo, it did not prevent dissemination to distant tissues. CONCLUSION Development of stably bioluminescent GAS for use in vitro and in vivo models of infection should facilitate development of novel therapeutics and vaccines while also increasing our understanding of infection progression and transmission routes.
Collapse
Affiliation(s)
- L E Lamb
- Section of Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London, UK.,Royal Centre Defence Medicine, Academia and Research, University of Birmingham, Birmingham, B15 2SQ, UK
| | - X Zhi
- Section of Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London, UK
| | - F Alam
- Section of Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London, UK
| | - M Pyzio
- Section of Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London, UK
| | - C L Scudamore
- MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, UK
| | - S Wiles
- Section of Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London, UK.,Bioluminescent Superbugs Lab, Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - S Sriskandan
- Section of Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London, UK.
| |
Collapse
|
39
|
Investigation of Polyaniline and a Functionalised Derivative as Antimicrobial Additives to Create Contamination Resistant Surfaces. MATERIALS 2018; 11:ma11030436. [PMID: 29547572 PMCID: PMC5873015 DOI: 10.3390/ma11030436] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 03/13/2018] [Accepted: 03/13/2018] [Indexed: 02/06/2023]
Abstract
Antimicrobial surfaces can be applied to break transmission pathways in hospitals. Polyaniline (PANI) and poly(3-aminobenzoic acid) (P3ABA) are novel antimicrobial agents with potential as non-leaching additives to provide contamination resistant surfaces. The activity of PANI and P3ABA were investigated in suspension and as part of absorbent and non-absorbent surfaces. The effect of inoculum size and the presence of organic matter on surface activity was determined. PANI and P3ABA both demonstrated bactericidal activity against Escherichia coli and Staphylococcus aureus in suspension and as part of an absorbent surface. Only P3ABA showed antimicrobial activity in non-absorbent films. The results that are presented in this work support the use of P3ABA to create contamination resistant surfaces.
Collapse
|
40
|
Roach DR, Leung CY, Henry M, Morello E, Singh D, Di Santo JP, Weitz JS, Debarbieux L. Synergy between the Host Immune System and Bacteriophage Is Essential for Successful Phage Therapy against an Acute Respiratory Pathogen. Cell Host Microbe 2018; 22:38-47.e4. [PMID: 28704651 DOI: 10.1016/j.chom.2017.06.018] [Citation(s) in RCA: 281] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/05/2017] [Accepted: 06/23/2017] [Indexed: 01/11/2023]
Abstract
The rise of multi-drug-resistant (MDR) bacteria has spurred renewed interest in the use of bacteriophages in therapy. However, mechanisms contributing to phage-mediated bacterial clearance in an animal host remain unclear. We investigated the effects of host immunity on the efficacy of phage therapy for acute pneumonia caused by MDR Pseudomonas aeruginosa in a mouse model. Comparing efficacies of phage-curative and prophylactic treatments in healthy immunocompetent, MyD88-deficient, lymphocyte-deficient, and neutrophil-depleted murine hosts revealed that neutrophil-phage synergy is essential for the resolution of pneumonia. Population modeling of in vivo results further showed that neutrophils are required to control both phage-sensitive and emergent phage-resistant variants to clear infection. This "immunophage synergy" contrasts with the paradigm that phage therapy success is largely due to bacterial permissiveness to phage killing. Lastly, therapeutic phages were not cleared by pulmonary immune effector cells and were immunologically well tolerated by lung tissues.
Collapse
Affiliation(s)
- Dwayne R Roach
- Department of Microbiology, Institut Pasteur, Paris 75015, France
| | - Chung Yin Leung
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA; School of Physics, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Marine Henry
- Department of Microbiology, Institut Pasteur, Paris 75015, France
| | - Eric Morello
- Department of Microbiology, Institut Pasteur, Paris 75015, France
| | - Devika Singh
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - James P Di Santo
- Innate Immunity Unit, Department of Immunology, Institut Pasteur, Paris 75015, France; Inserm U1223, Paris 75015, France
| | - Joshua S Weitz
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA; School of Physics, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| | | |
Collapse
|
41
|
Conway TP, Daniels KJ, Park YN, Soll DR. Generating a Battery of Monoclonal Antibodies Against Firefly Luciferase for Dot Blot Analysis, Western Blot Analysis, and Immunostaining of Cells in Culture and Paraffin Sections. Monoclon Antib Immunodiagn Immunother 2018; 37:45-51. [PMID: 29474160 DOI: 10.1089/mab.2017.0057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Firefly luciferase (FLuc) is commonly used as a reporter gene PpyLuc1 in bioanalytical assays. We have produced five mouse-derived monoclonal antibodies (mAbs) that recognize FLuc. The mAbs, DSHB-LUC-2, DSHB-LUC-3, DSHB-LUC-9, DSHB-LUC-16, and DSHB-LUC-24, were generated by immunizing mice with purified 6xHIS-tagged FLuc (6xHis-FLuc) in suspension with an adjuvant. All five were validated by dot blots. Four of the mAbs provided strong signals in western blot analysis, and one a weak signal. All five were validated for immunostaining in fixed cell culture. Only one stained cells embedded in paraffin. The five mAbs are available at cost through the Developmental Studies Hybridoma Bank (DSHB), a nonprofit National Resource created by the National Institutes of Health.
Collapse
Affiliation(s)
- Thomas P Conway
- Developmental Studies Hybridoma Bank and W.M. Keck Dynamic Image Analysis, Department of Biology, The University of Iowa , Iowa City, Iowa
| | - Karla J Daniels
- Developmental Studies Hybridoma Bank and W.M. Keck Dynamic Image Analysis, Department of Biology, The University of Iowa , Iowa City, Iowa
| | - Yang-Nim Park
- Developmental Studies Hybridoma Bank and W.M. Keck Dynamic Image Analysis, Department of Biology, The University of Iowa , Iowa City, Iowa
| | - David R Soll
- Developmental Studies Hybridoma Bank and W.M. Keck Dynamic Image Analysis, Department of Biology, The University of Iowa , Iowa City, Iowa
| |
Collapse
|
42
|
L. plantarum WCFS1 enhances Treg frequencies by activating DCs even in absence of sampling of bacteria in the Peyer Patches. Sci Rep 2018; 8:1785. [PMID: 29379071 PMCID: PMC5788989 DOI: 10.1038/s41598-018-20243-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/15/2018] [Indexed: 12/14/2022] Open
Abstract
Probiotics such as L. plantarum WCFS1 can modulate immune responses in healthy subjects but how this occurs is still largely unknown. Immune-sampling in the Peyer Patches has been suggested to be one of the mechanisms. Here we studied the systemic and intestinal immune effects in combination with a trafficking study through the intestine of a well-established immunomodulating probiotic, i.e. L. plantarum WCFS1. We demonstrate that not more than 2–3 bacteria were sampled and in many animals not any bacterium could be found in the PP. Despite this, L. plantarum was associated with a strong increase in infiltration of regulatory CD103+ DCs and generation of regulatory T cells in the spleen. Also, a reduced splenic T helper cell cytokine response was observed after ex vivo restimulation. L. plantarum enhanced Treg cells and attenuated the T helper 2 response in healthy mice. We demonstrate that, in healthy mice, immune sampling is a rare phenomenon and not required for immunomodulation. Also in absence of any sampling immune activation was found illustrating that host-microbe interaction on the Peyer Patches was enough to induce immunomodulation of DCs and T-cells.
Collapse
|
43
|
Hyde JA, Skare JT. Detection of Bioluminescent Borrelia burgdorferi from In Vitro Cultivation and During Murine Infection. Methods Mol Biol 2018; 1690:241-257. [PMID: 29032549 PMCID: PMC8786108 DOI: 10.1007/978-1-4939-7383-5_18] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Borrelia burgdorferi, etiologic agent of Lyme disease, is the leading tick-borne disease in the United States with approximately 300,000 cases diagnosed annually. Disease occurs in stages beginning localized infection at the site of a tick bite and progresses to disseminated infection when antibiotic treatment is not administered in a timely manner. A multi-systemic infection develops following dissemination to numerous immunoprotective tissues, such as the heart, bladder, and joints, resulting in late Lyme disease. B. burgdorferi undergoes dynamic genetic regulation throughout mammalian infection and defining the exact role of virulence genes at distinct stages of disease is challenging. The murine model allows for the characterization of the pathogenic function of genes in B. burgdorferi, but traditional end point studies limit the ability to gather data throughout an infection study and greatly increase the required number of mice. Molecular genetic techniques to evaluate and quantitate B. burgdorferi infection are laborious and costly. To partly circumvent these issues, a codon optimized firefly luciferase, under the control of a constitutive borrelial promoter, was introduced into B. burgdorferi enabling the characterization of mutant or modified strains under in vitro growth conditions and throughout murine infection. The detection of bioluminescent B. burgdorferi is highly sensitive and allows for the repeated real-time quantitative evaluation of borrelial load during murine infection. Furthermore, bioluminescence has also been utilized to evaluate alteration in tissue localization and tissue-specific gene expression of B. burgdorferi. In this chapter, we describe the generation of bioluminescent borrelial strains along with methods for in vitro, in vivo, and ex vivo B. burgdorferi studies.
Collapse
Affiliation(s)
- Jenny A Hyde
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science Center, Riverside Pkwy, Bryan, Texas, 77807, USA.
| | - Jon T Skare
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science Center, Riverside Pkwy, Bryan, Texas, 77807, USA
| |
Collapse
|
44
|
McCallum H, Fenton A, Hudson PJ, Lee B, Levick B, Norman R, Perkins SE, Viney M, Wilson AJ, Lello J. Breaking beta: deconstructing the parasite transmission function. Philos Trans R Soc Lond B Biol Sci 2017; 372:rstb.2016.0084. [PMID: 28289252 PMCID: PMC5352811 DOI: 10.1098/rstb.2016.0084] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2016] [Indexed: 01/29/2023] Open
Abstract
Transmission is a fundamental step in the life cycle of every parasite but it is also one of the most challenging processes to model and quantify. In most host–parasite models, the transmission process is encapsulated by a single parameter β. Many different biological processes and interactions, acting on both hosts and infectious organisms, are subsumed in this single term. There are, however, at least two undesirable consequences of this high level of abstraction. First, nonlinearities and heterogeneities that can be critical to the dynamic behaviour of infections are poorly represented; second, estimating the transmission coefficient β from field data is often very difficult. In this paper, we present a conceptual model, which breaks the transmission process into its component parts. This deconstruction enables us to identify circumstances that generate nonlinearities in transmission, with potential implications for emergent transmission behaviour at individual and population scales. Such behaviour cannot be explained by the traditional linear transmission frameworks. The deconstruction also provides a clearer link to the empirical estimation of key components of transmission and enables the construction of flexible models that produce a unified understanding of the spread of both micro- and macro-parasite infectious disease agents. This article is part of the themed issue ‘Opening the black box: re-examining the ecology and evolution of parasite transmission’.
Collapse
Affiliation(s)
- Hamish McCallum
- Environmental Futures Research Institute, Griffith University, Nathan 4111, Queensland, Australia
| | - Andy Fenton
- Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Peter J Hudson
- Center for Infectious Disease Dynamics, Penn State University, University Park, PA 16802, USA
| | - Brian Lee
- School of Natural Sciences, University of Stirling, Stirling FK9 4LA, UK
| | - Beth Levick
- Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Rachel Norman
- School of Natural Sciences, University of Stirling, Stirling FK9 4LA, UK
| | - Sarah E Perkins
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK.,Department of Biodiversity and Molecular Ecology, Research and Innovation Centre, Fondazione Edmund Mach, Via E. Mach 1, 38010 S. Michele all'Adige, Trentino, Italy
| | - Mark Viney
- School of Biological Sciences, University of Bristol, Tyndall Avenue, Bristol BS8 1TQ, UK
| | - Anthony J Wilson
- Vector-borne Viral Diseases Programme, The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK
| | - Joanne Lello
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK .,Department of Biodiversity and Molecular Ecology, Research and Innovation Centre, Fondazione Edmund Mach, Via E. Mach 1, 38010 S. Michele all'Adige, Trentino, Italy
| |
Collapse
|
45
|
Sharifian S, Homaei A, Hemmati R, Khajeh K. Light emission miracle in the sea and preeminent applications of bioluminescence in recent new biotechnology. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2017; 172:115-128. [DOI: 10.1016/j.jphotobiol.2017.05.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 05/16/2017] [Indexed: 02/08/2023]
|
46
|
Giardia Colonizes and Encysts in High-Density Foci in the Murine Small Intestine. mSphere 2017; 2:mSphere00343-16. [PMID: 28656177 PMCID: PMC5480036 DOI: 10.1128/msphere.00343-16] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 04/21/2017] [Indexed: 01/28/2023] Open
Abstract
Giardia is a single-celled parasite causing significant diarrheal disease in several hundred million people worldwide. Due to limited access to the site of infection in the gastrointestinal tract, our understanding of the dynamics of Giardia infections in the host has remained limited and largely inferred from laboratory culture. To better understand Giardia physiology and colonization in the host, we developed imaging methods to quantify Giardia expressing bioluminescent physiological reporters in two relevant animal models. We discovered that parasites primarily colonize and encyst in the proximal small intestine in discrete, high-density foci. We also show that high parasite density contributes to encystation initiation. Giardia lamblia is a highly prevalent yet understudied protistan parasite causing significant diarrheal disease worldwide. Hosts ingest Giardia cysts from contaminated sources. In the gastrointestinal tract, cysts excyst to become motile trophozoites, colonizing and attaching to the gut epithelium. Trophozoites later differentiate into infectious cysts that are excreted and contaminate the environment. Due to the limited accessibility of the gut, the temporospatial dynamics of giardiasis in the host are largely inferred from laboratory culture and thus may not mirror Giardia physiology in the host. Here, we have developed bioluminescent imaging (BLI) to directly interrogate and quantify the in vivo temporospatial dynamics of Giardia infection, thereby providing an improved murine model to evaluate anti-Giardia drugs. Using BLI, we determined that parasites primarily colonize the proximal small intestine nonuniformly in high-density foci. By imaging encystation-specific bioreporters, we show that encystation initiates shortly after inoculation and continues throughout the duration of infection. Encystation also initiates in high-density foci in the proximal small intestine, and high density contributes to the initiation of encystation in laboratory culture. We suggest that these high-density in vivo foci of colonizing and encysting Giardia likely result in localized disruption to the epithelium. This more accurate visualization of giardiasis redefines the dynamics of the in vivo Giardia life cycle, paving the way for future mechanistic studies of density-dependent parasitic processes in the host. IMPORTANCEGiardia is a single-celled parasite causing significant diarrheal disease in several hundred million people worldwide. Due to limited access to the site of infection in the gastrointestinal tract, our understanding of the dynamics of Giardia infections in the host has remained limited and largely inferred from laboratory culture. To better understand Giardia physiology and colonization in the host, we developed imaging methods to quantify Giardia expressing bioluminescent physiological reporters in two relevant animal models. We discovered that parasites primarily colonize and encyst in the proximal small intestine in discrete, high-density foci. We also show that high parasite density contributes to encystation initiation.
Collapse
|
47
|
Adams PP, Flores Avile C, Jewett MW. A Dual Luciferase Reporter System for B. burgdorferi Measures Transcriptional Activity during Tick-Pathogen Interactions. Front Cell Infect Microbiol 2017; 7:225. [PMID: 28620587 PMCID: PMC5449462 DOI: 10.3389/fcimb.2017.00225] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 05/16/2017] [Indexed: 12/23/2022] Open
Abstract
Knowledge of the transcriptional responses of vector-borne pathogens at the vector-pathogen interface is critical for understanding disease transmission. Borrelia (Borreliella) burgdorferi, the causative agent of Lyme disease in the United States, is transmitted by the bite of infected Ixodes sp. ticks. It is known that B. burgdorferi has altered patterns of gene expression during tick acquisition, persistence and transmission. Recently, we and others have discovered in vitro expression of RNAs found internal, overlapping, and antisense to annotated open reading frames in the B. burgdorferi genome. However, there is a lack of molecular genetic tools for B. burgdorferi for quantitative, strand-specific, comparative analysis of these transcripts in distinct environments such as the arthropod vector. To address this need, we have developed a dual luciferase reporter system to quantify B. burgdorferi promoter activities in a strand-specific manner. We demonstrate that constitutive expression of a B. burgdorferi codon-optimized Renilla reniformis luciferase gene (rlucBb ) allows normalization of the activity of a promoter of interest when fused to the B. burgdorferi codon-optimized Photinus pyralis luciferase gene (flucBb) on the same plasmid. Using the well characterized, differentially regulated, promoters for flagellin (flaBp), outer surface protein A (ospAp) and outer surface protein C (ospCp), we document the efficacy of the dual luciferase system for quantitation of promoter activities during in vitro growth and in infected ticks. Cumulatively, the dual luciferase method outlined herein is the first dual reporter system for B. burgdorferi, providing a novel and highly versatile approach for strand-specific molecular genetic analyses.
Collapse
Affiliation(s)
- Philip P Adams
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, University of Central Florida College of MedicineOrlando, FL, United States
| | - Carlos Flores Avile
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, University of Central Florida College of MedicineOrlando, FL, United States
| | - Mollie W Jewett
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, University of Central Florida College of MedicineOrlando, FL, United States
| |
Collapse
|
48
|
Pham JK, Nosala C, Scott EY, Nguyen KF, Hagen KD, Starcevich HN, Dawson SC. Transcriptomic Profiling of High-Density Giardia Foci Encysting in the Murine Proximal Intestine. Front Cell Infect Microbiol 2017; 7:227. [PMID: 28620589 PMCID: PMC5450421 DOI: 10.3389/fcimb.2017.00227] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 05/16/2017] [Indexed: 12/15/2022] Open
Abstract
Giardia is a highly prevalent, understudied protistan parasite causing significant diarrheal disease worldwide. Its life cycle consists of two stages: infectious cysts ingested from contaminated food or water sources, and motile trophozoites that colonize and attach to the gut epithelium, later encysting to form new cysts that are excreted into the environment. Current understanding of parasite physiology in the host is largely inferred from transcriptomic studies using Giardia grown axenically or in co-culture with mammalian cell lines. The dearth of information about the diversity of host-parasite interactions occurring within distinct regions of the gastrointestinal tract has been exacerbated by a lack of methods to directly and non-invasively interrogate disease progression and parasite physiology in live animal hosts. By visualizing Giardia infections in the mouse gastrointestinal tract using bioluminescent imaging (BLI) of tagged parasites, we recently showed that parasites colonize the gut in high-density foci. Encystation is initiated in these foci throughout the entire course of infection, yet how the physiology of parasites within high-density foci in the host gut differs from that of cells in laboratory culture is unclear. Here we use BLI to precisely select parasite samples from high-density foci in the proximal intestine to interrogate in vivo Giardia gene expression in the host. Relative to axenic culture, we noted significantly higher expression (>10-fold) of oxidative stress, membrane transporter, and metabolic and structural genes associated with encystation in the high-density foci. These differences in gene expression within parasite foci in the host may reflect physiological changes associated with high-density growth in localized regions of the gut. We also identified and verified six novel cyst-specific proteins, including new components of the cyst wall that were highly expressed in these foci. Our in vivo transcriptome data support an emerging view that parasites encyst early in localized regions in the gut, possibly as a consequence of nutrient limitation, and also impact local metabolism and physiology.
Collapse
Affiliation(s)
- Jonathan K Pham
- Department of Microbiology and Molecular Genetics, University of California, DavisDavis, CA, United States
| | - Christopher Nosala
- Department of Microbiology and Molecular Genetics, University of California, DavisDavis, CA, United States
| | - Erica Y Scott
- Department of Animal Science, University of California, DavisDavis, CA, United States
| | - Kristofer F Nguyen
- Department of Microbiology and Molecular Genetics, University of California, DavisDavis, CA, United States
| | - Kari D Hagen
- Department of Microbiology and Molecular Genetics, University of California, DavisDavis, CA, United States
| | - Hannah N Starcevich
- Department of Microbiology and Molecular Genetics, University of California, DavisDavis, CA, United States
| | - Scott C Dawson
- Department of Microbiology and Molecular Genetics, University of California, DavisDavis, CA, United States
| |
Collapse
|
49
|
Ozuru R, Saito M, Kanemaru T, Miyahara S, Villanueva SYAM, Murray GL, Adler B, Fujii J, Yoshida SI. Adipose tissue is the first colonization site of Leptospira interrogans in subcutaneously infected hamsters. PLoS One 2017; 12:e0172973. [PMID: 28245231 PMCID: PMC5330501 DOI: 10.1371/journal.pone.0172973] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 02/12/2017] [Indexed: 01/29/2023] Open
Abstract
Leptospirosis is one of the most widespread zoonoses in the world, and its most severe form in humans, “Weil’s disease,” may lead to jaundice, hemorrhage, renal failure, pulmonary hemorrhage syndrome, and sometimes,fatal multiple organ failure. Although the mechanisms underlying jaundice in leptospirosis have been gradually unraveled, the pathophysiology and distribution of leptospires during the early stage of infection are not well understood. Therefore, we investigated the hamster leptospirosis model, which is the accepted animal model of human Weil’s disease, by using an in vivo imaging system to observe the whole bodies of animals infected with Leptospira interrogans and to identify the colonization and growth sites of the leptospires during the early phase of infection. Hamsters, infected subcutaneously with 104 bioluminescent leptospires, were analyzed by in vivo imaging, organ culture, and microscopy. The results showed that the luminescence from the leptospires spread through each hamster’s body sequentially. The luminescence was first detected at the injection site only, and finally spread to the central abdomen, in the liver area. Additionally, the luminescence observed in the adipose tissue was the earliest detectable compared with the other organs, indicating that the leptospires colonized the adipose tissue at the early stage of leptospirosis. Adipose tissue cultures of the leptospires became positive earlier than the blood cultures. Microscopic analysis revealed that the leptospires colonized the inner walls of the blood vessels in the adipose tissue. In conclusion, this is the first study to report that adipose tissue is an important colonization site for leptospires, as demonstrated by microscopy and culture analyses of adipose tissue in the hamster model of Weil’s disease.
Collapse
Affiliation(s)
- Ryo Ozuru
- Department of Bacteriology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Bacteriology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Tottori, Japan
- * E-mail:
| | - Mitsumasa Saito
- Department of Microbiology, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Takaaki Kanemaru
- Morphology Core Unit, Kyushu University Hospital, Fukuoka, Japan
| | - Satoshi Miyahara
- Department of Bacteriology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Gerald L. Murray
- Department of Microbiology, Monash University, Melbourne, Australia
| | - Ben Adler
- Department of Microbiology, Monash University, Melbourne, Australia
| | - Jun Fujii
- Division of Bacteriology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Shin-ichi Yoshida
- Department of Bacteriology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
50
|
Li XF, Li XD, Deng CL, Dong HL, Zhang QY, Ye Q, Ye HQ, Huang XY, Deng YQ, Zhang B, Qin CF. Visualization of a neurotropic flavivirus infection in mouse reveals unique viscerotropism controlled by host type I interferon signaling. Am J Cancer Res 2017; 7:912-925. [PMID: 28382163 PMCID: PMC5381253 DOI: 10.7150/thno.16615] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 12/17/2016] [Indexed: 01/07/2023] Open
Abstract
Flavivirus includes a large group of human pathogens with medical importance. Especially, neurotropic flaviviruses capable of invading central and peripheral nervous system, e.g. Japanese encephalitis virus (JEV) and Zika virus (ZIKV), are highly pathogenic to human and constitute major global health problems. However, the dynamic dissemination and pathogenesis of neurotropic flavivirus infections remain largely unknown. Here, using JEV as a model, we rationally designed and constructed a recombinant reporter virus that stably expressed Renilla luciferase (Rluc). The resulting JEV reporter virus (named Rluc-JEV) and parental JEV exhibited similar replication and infection characteristics, and the magnitude of Rluc activity correlated well with progeny viral production in vitro and in vivo. By using in vivo bioluminescence imaging (BLI) technology, we dissected the replication and dissemination dynamics of JEV infection in mice upon different inoculation routes. Interestingly, besides replicating in mouse brain, Rluc-JEV predominantly invaded the abdominal organs in mice with typical viscerotropism. Further tests in mice deficient in type I interferon (IFN) receptors demonstrated robust and prolonged viral replication in the intestine, spleen, liver, kidney and other abdominal organs. Combined with histopathological and immunohistochemical results, the host type I IFN signaling was evidenced as the major barrier to the viscerotropism and pathogenicity of this neurotropic flavivirus. Additionally, the Rluc-JEV platform was readily adapted for efficacy assay of known antiviral compounds and a live JE vaccine. Collectively, our study revealed abdominal organs as important targets of JEV infection in mice and profiled the unique viscerotropism trait controlled by the host type I IFN signaling. This in vivo visualization technology described here provides a powerful tool for testing antiviral agents and vaccine candidates for flaviviral infection.
Collapse
|