1
|
Shang W, Hu Z, Li M, Wang Y, Rao Y, Tan L, Chen J, Huang X, Liu L, Liu H, Guo Z, Peng H, Yang Y, Hu Q, Li S, Hu X, Zou J, Rao X. Optimizing a high-sensitivity NanoLuc-based bioluminescence system for in vivo evaluation of antimicrobial treatment. MLIFE 2023; 2:462-478. [PMID: 38818266 PMCID: PMC10989145 DOI: 10.1002/mlf2.12091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 08/05/2023] [Accepted: 08/28/2023] [Indexed: 06/01/2024]
Abstract
Focal and systemic infections are serious threats to human health. Preclinical models enable the development of new drugs and therapeutic regimens. In vivo, animal bioluminescence (BL) imaging has been used with bacterial reporter strains to evaluate antimicrobial treatment effects. However, high-sensitivity bioluminescent systems are required because of the limited tissue penetration and low brightness of the BL signals of existing approaches. Here, we report that NanoLuc (Nluc) showed better performance than LuxCDABE in bacteria. However, the retention rate of plasmid constructs in bacteria was low. To construct stable Staphylococcus aureus reporter strains, a partner protein enolase (Eno) was identified by screening of S. aureus strain USA300 for fusion expression of Nluc-based luciferases, including Nluc, Teluc, and Antares2. Different substrates, such as hydrofurimazine (HFZ), furimazine (FUR), and diphenylterazine (DTZ), were used to optimize a stable reporter strain/substrate pair for BL imaging. S. aureus USA300/Eno-Antares2/HFZ produced the highest number of photons of orange-red light in vitro and enabled sensitive BL tracking of S. aureus in vivo, with sensitivities of approximately 10 CFU from mouse skin and 750 CFU from mouse kidneys. USA300/Eno-Antares2/HFZ was a powerful combination based on the longitudinal evaluation of the therapeutic efficacy of antibiotics. The optimized S. aureus Eno-Antares2/HFZ pair provides a technological advancement for the in vivo evaluation of antimicrobial treatment.
Collapse
Affiliation(s)
- Weilong Shang
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing Army Medical University (Third Military Medical University) Chongqing China
| | - Zhen Hu
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing Army Medical University (Third Military Medical University) Chongqing China
| | - Mengyang Li
- Department of Microbiology, School of Medicine Chongqing University Chongqing China
| | - Yuting Wang
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing Army Medical University (Third Military Medical University) Chongqing China
| | - Yifan Rao
- Department of Emergency Medicine, Xinqiao Hospital Army Medical University (Third Military Medical University) Chongqing China
| | - Li Tan
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing Army Medical University (Third Military Medical University) Chongqing China
| | - Juan Chen
- Department of Pharmacy, Xinqiao Hospital Army Medical University (Third Military Medical University) Chongqing China
| | - Xiaonan Huang
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing Army Medical University (Third Military Medical University) Chongqing China
| | - Lu Liu
- Department of Microbiology, School of Medicine Chongqing University Chongqing China
| | - He Liu
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing Army Medical University (Third Military Medical University) Chongqing China
| | - Zuwen Guo
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing Army Medical University (Third Military Medical University) Chongqing China
| | - Huagang Peng
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing Army Medical University (Third Military Medical University) Chongqing China
| | - Yi Yang
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing Army Medical University (Third Military Medical University) Chongqing China
| | - Qiwen Hu
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing Army Medical University (Third Military Medical University) Chongqing China
| | - Shu Li
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing Army Medical University (Third Military Medical University) Chongqing China
| | - Xiaomei Hu
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing Army Medical University (Third Military Medical University) Chongqing China
| | - Jiao Zou
- Department of Military Cognitive Psychology, School of Psychology Army Medical University (Third Military Medical University) Chongqing China
| | - Xiancai Rao
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing Army Medical University (Third Military Medical University) Chongqing China
| |
Collapse
|
2
|
Bacterial bioluminescence assay for bioanalysis and bioimaging. Anal Bioanal Chem 2021; 414:75-83. [PMID: 34693470 DOI: 10.1007/s00216-021-03695-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/31/2021] [Accepted: 09/24/2021] [Indexed: 10/20/2022]
Abstract
Bioluminescence occurs through a chemical reaction in organisms that spontaneously produce light. Luminescent bacteria are unique among bioluminescent organisms. Their bioluminescence intensity is an indicator of their metabolic activity, which can directly reflect the influence of environmental factors on cell viability. Moreover, the whole bioluminescence process is totally gene encoded without the addition of extra substrates. As a result, bacterial bioluminescence has been a powerful tool for whole-cell biosensors and bio-reporters in bioanalysis and bioimaging. This review aims to cover the applications of wild-type and recombinant luminescent bacteria to detect the toxicity of environmental pollutants and biological molecules. The bacterial bioluminescence analytical assay has characteristics such as high sensitivity, short-term detection, and easy operation. Meanwhile, due to the development of gene engineering and optical technology, bacterial luciferase as a reporter protein has been successfully expressed in prokaryotic and eukaryotic cells, tissues, and organs of animals. The major applications for bacterial luciferase-based bioluminescence imaging, such as infectious diseases, cancer therapy, and stem cell tracing, are discussed in this review.
Collapse
|
3
|
Seele J, Ballüer M, Tauber SC, Bunkowski S, Schulz K, Stadelmann C, Beineke A, Pägelow D, Fulde M, Nau R. Neural Injury and Repair in a Novel Neonatal Mouse Model of Listeria Monocytogenes Meningoencephalitis. J Neuropathol Exp Neurol 2021; 80:861-867. [PMID: 34486672 DOI: 10.1093/jnen/nlab079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
To improve the therapy of neonatal central nervous system infections, well-characterized animal models are urgently needed. The present study analyzes neuropathological alterations with particular focus on neural injury and repair in brains of neonatal mice with Listeria monocytogenes (LM) meningitis/meningoencephalitis using a novel nasal infection model. The hippocampal formation and frontal cortex of 14 neonatal mice with LM meningitis/meningoencephalitis and 14 uninfected controls were analyzed by histology, immunohistochemistry, and in situ tailing for morphological alterations. In the dentate gyrus of the hippocampal formation of mice with LM meningitis/meningoencephalitis, an increased density of apoptotic neurons visualized by in situ tailing (p = 0.04) and in situ tailing plus immunohistochemistry for activated Caspase-3 (p < 0.0001) was found. A decreased density of dividing cells stained with an anti-PCNA-antibody (p < 0.0001) and less neurogenesis visualized by anti-calretinin (p < 0.0001) and anti-calbindin (p = 0.01) antibodies were detected compared to uninfected controls. The density of microglia was higher in LM meningitis (p < 0.0001), while the density of astrocytes remained unchanged. Infiltrating monocytes and neutrophilic granulocytes likely contributed to tissue damage. In conclusion, in the brains of LM-infected mice a strong immune response was observed which led to neuronal apoptosis and an impaired neural regeneration. This model appears very suitable to study therapies against long-term sequelae of neonatal LM meningitis.
Collapse
Affiliation(s)
- Jana Seele
- Department of Neuropathology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany.,Department of Geriatrics, Evangelisches Krankenhaus Göttingen-Weende, Göttingen, Germany
| | - Melissa Ballüer
- Department of Neuropathology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany.,Department of Geriatrics, Evangelisches Krankenhaus Göttingen-Weende, Göttingen, Germany
| | - Simone C Tauber
- Department of Neurology, RWTH University Hospital, Aachen, Germany
| | - Stephanie Bunkowski
- Department of Neuropathology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany
| | - Katja Schulz
- Department of Neuropathology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany
| | - Christine Stadelmann
- Department of Neuropathology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Dennis Pägelow
- Institute of Microbiology and Epizootics, Centre of Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Marcus Fulde
- Institute of Microbiology and Epizootics, Centre of Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Roland Nau
- Department of Neuropathology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany.,Department of Geriatrics, Evangelisches Krankenhaus Göttingen-Weende, Göttingen, Germany
| |
Collapse
|
4
|
Functional Imaging Using Bioluminescent Reporter Genes in Living Subjects. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00004-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
5
|
Miller RJ, Crosby HA, Schilcher K, Wang Y, Ortines RV, Mazhar M, Dikeman DA, Pinsker BL, Brown ID, Joyce DP, Zhang J, Archer NK, Liu H, Alphonse MP, Czupryna J, Anderson WR, Bernthal NM, Fortuno-Miranda L, Bulte JWM, Francis KP, Horswill AR, Miller LS. Development of a Staphylococcus aureus reporter strain with click beetle red luciferase for enhanced in vivo imaging of experimental bacteremia and mixed infections. Sci Rep 2019; 9:16663. [PMID: 31723175 PMCID: PMC6853927 DOI: 10.1038/s41598-019-52982-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/26/2019] [Indexed: 02/03/2023] Open
Abstract
In vivo bioluminescence imaging has been used to monitor Staphylococcus aureus infections in preclinical models by employing bacterial reporter strains possessing a modified lux operon from Photorhabdus luminescens. However, the relatively short emission wavelength of lux (peak 490 nm) has limited tissue penetration. To overcome this limitation, the gene for the click beetle (Pyrophorus plagiophtalamus) red luciferase (luc) (with a longer >600 emission wavelength), was introduced singly and in combination with the lux operon into a methicillin-resistant S. aureus strain. After administration of the substrate D-luciferin, the luc bioluminescent signal was substantially greater than the lux signal in vitro. The luc signal had enhanced tissue penetration and improved anatomical co-registration with infected internal organs compared with the lux signal in a mouse model of S. aureus bacteremia with a sensitivity of approximately 3 × 104 CFU from the kidneys. Finally, in an in vivo mixed bacterial wound infection mouse model, S. aureus luc signals could be spectrally unmixed from Pseudomonas aeruginosa lux signals to noninvasively monitor the bacterial burden of both strains. Therefore, the S. aureus luc reporter may provide a technological advance for monitoring invasive organ dissemination during S. aureus bacteremia and for studying bacterial dynamics during mixed infections.
Collapse
Affiliation(s)
- Robert J Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Heidi A Crosby
- Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045, USA
| | - Katrin Schilcher
- Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045, USA
| | - Yu Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Roger V Ortines
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Momina Mazhar
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dustin A Dikeman
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Bret L Pinsker
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Isabelle D Brown
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniel P Joyce
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeffrey Zhang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nathan K Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Haiyun Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Martin P Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | - Nicholas M Bernthal
- Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, Santa Monica, California, USA
| | - Lea Fortuno-Miranda
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Jeff W M Bulte
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA.,Department of Chemical & Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, Maryland, 21205, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Kevin P Francis
- PerkinElmer, Hopkinton, Massachusetts, USA.,Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, Santa Monica, California, USA
| | - Alexander R Horswill
- Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045, USA.,Denver VA Healthcare System, Denver, Colorado, USA
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA. .,Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA. .,Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA. .,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, 21218, USA.
| |
Collapse
|
6
|
Liu YH, Wu PH, Kang CC, Tsai YS, Chou CK, Liang CT, Wu JJ, Tsai PJ. Group A Streptococcus Subcutaneous Infection-Induced Central Nervous System Inflammation Is Attenuated by Blocking Peripheral TNF. Front Microbiol 2019; 10:265. [PMID: 30837977 PMCID: PMC6389723 DOI: 10.3389/fmicb.2019.00265] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 02/01/2019] [Indexed: 11/13/2022] Open
Abstract
Group A streptococcus (GAS) infection causes a strong inflammatory response associated with cytokine storms, leading to multiorgan failure, which is characterized as streptococcal toxic shock syndrome. However, little is known about GAS subcutaneous infection-mediated brain inflammation. Therefore, we used a bioluminescent GAS strain and reporter mice carrying firefly luciferase under transcriptional control of the nuclear factor-kappa B (NF-κB) promoter to concurrently monitor the host immune response and bacterial burden in a single mouse. Notably, in addition to the subcutaneous inoculation locus at the back of mice, we detected strong luminescence signals from NF-κB activation and increased inflammatory cytokine production in the brain, implying the existence of central nervous system inflammation after GAS subcutaneous infection. The inflamed brain exhibited an increased expression of glial fibrillary acidic protein and nicotinamide adenine dinucleotide phosphate oxidase components and greater microglial activation and blood–brain barrier (BBB) disruption. Furthermore, Fluoro-Jade C positive cells increased in the brain, indicating that neurons underwent degeneration. Peripheral tumor necrosis factor (TNF), which contributes to pathology in brain injury, was elevated in the circulation, and the expression of its receptor was also increased in the inflamed brain. Blockage of peripheral TNF effectively reduced brain inflammation and injury, thereby preventing BBB disruption and improving survival. Our study provides new insights into GAS-induced central nervous system inflammation, such as encephalopathy, which can be attenuated by circulating TNF blockage.
Collapse
Affiliation(s)
- Ya-Hui Liu
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Hua Wu
- Department of Medical Laboratory Science and Biotechnology, Medical College, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Cheng Kang
- Department of Medical Laboratory Science and Biotechnology, Medical College, National Cheng Kung University, Tainan, Taiwan
| | - Yau-Sheng Tsai
- Institute of Clinical Medicine, National Cheng Kung University, Tainan, Taiwan.,Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Chuan-Kai Chou
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Chung-Tiang Liang
- Novo Nordisk Research Centre China, Beijing, China.,Department of Animal Facility, Discovery Biology China, Beijing, China
| | - Jiunn-Jong Wu
- Department of Medical Laboratory Science and Biotechnology, Medical College, National Cheng Kung University, Tainan, Taiwan.,Department of Biotechnology and Laboratory Science in Medicine, School of Biomedical Science and Engineering, National Yang-Ming University, Taipei, Taiwan
| | - Pei-Jane Tsai
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Medical Laboratory Science and Biotechnology, Medical College, National Cheng Kung University, Tainan, Taiwan.,Research Center of Infectious Disease and Signaling, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
7
|
Avci P, Karimi M, Sadasivam M, Antunes-Melo WC, Carrasco E, Hamblin MR. In-vivo monitoring of infectious diseases in living animals using bioluminescence imaging. Virulence 2017; 9:28-63. [PMID: 28960132 PMCID: PMC6067836 DOI: 10.1080/21505594.2017.1371897] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Traditional methods of localizing and quantifying the presence of pathogenic microorganisms in living experimental animal models of infections have mostly relied on sacrificing the animals, dissociating the tissue and counting the number of colony forming units. However, the discovery of several varieties of the light producing enzyme, luciferase, and the genetic engineering of bacteria, fungi, parasites and mice to make them emit light, either after administration of the luciferase substrate, or in the case of the bacterial lux operon without any exogenous substrate, has provided a new alternative. Dedicated bioluminescence imaging (BLI) cameras can record the light emitted from living animals in real time allowing non-invasive, longitudinal monitoring of the anatomical location and growth of infectious microorganisms as measured by strength of the BLI signal. BLI technology has been used to follow bacterial infections in traumatic skin wounds and burns, osteomyelitis, infections in intestines, Mycobacterial infections, otitis media, lung infections, biofilm and endodontic infections and meningitis. Fungi that have been engineered to be bioluminescent have been used to study infections caused by yeasts (Candida) and by filamentous fungi. Parasitic infections caused by malaria, Leishmania, trypanosomes and toxoplasma have all been monitored by BLI. Viruses such as vaccinia, herpes simplex, hepatitis B and C and influenza, have been studied using BLI. This rapidly growing technology is expected to continue to provide much useful information, while drastically reducing the numbers of animals needed in experimental studies.
Collapse
Affiliation(s)
- Pinar Avci
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,b Department of Dermatology , Harvard Medical School , Boston , MA , USA
| | - Mahdi Karimi
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,c Department of Medical Nanotechnology , School of Advanced Technologies in Medicine, Iran University of Medical Sciences , Tehran , Iran.,d Cellular and Molecular Research Center, Iran University of Medical Sciences , Tehran , Iran
| | - Magesh Sadasivam
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,e Amity Institute of Nanotechnology, Amity University Uttar Pradesh , Noida , India
| | - Wanessa C Antunes-Melo
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,f University of Sao Paulo , Sao Carlos-SP , Brazil
| | - Elisa Carrasco
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,g Department of Biosciences , Durham University , Durham , United Kingdom
| | - Michael R Hamblin
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,b Department of Dermatology , Harvard Medical School , Boston , MA , USA.,h Harvard-MIT Division of Health Sciences and Technology , Cambridge , MA , USA
| |
Collapse
|
8
|
Suff N, Waddington SN. The power of bioluminescence imaging in understanding host-pathogen interactions. Methods 2017; 127:69-78. [PMID: 28694065 DOI: 10.1016/j.ymeth.2017.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/12/2017] [Accepted: 07/03/2017] [Indexed: 01/06/2023] Open
Abstract
Infectious diseases are one of the leading causes of death worldwide. Modelling and understanding human infection is imperative to developing treatments to reduce the global burden of infectious disease. Bioluminescence imaging is a highly sensitive, non-invasive technique based on the detection of light, produced by luciferase-catalysed reactions. In the study of infectious disease, bioluminescence imaging is a well-established technique; it can be used to detect, localize and quantify specific immune cells, pathogens or immunological processes. This enables longitudinal studies in which the spectrum of the disease process and its response to therapies can be monitored. Light producing transgenic rodents are emerging as key tools in the study of host response to infection. Here, we review the strategies for identifying biological processes in vivo, including the technology of bioluminescence imaging and illustrate how this technique is shedding light on the host-pathogen relationship.
Collapse
Affiliation(s)
- Natalie Suff
- Gene Transfer Technology Group, Institute for Women's Health, University College London, 86-96 Chenies Mews, London WC1E 6HX, United Kingdom.
| | - Simon N Waddington
- Gene Transfer Technology Group, Institute for Women's Health, University College London, 86-96 Chenies Mews, London WC1E 6HX, United Kingdom
| |
Collapse
|
9
|
Biodistribution and residence time of adenovector serotype 5 in normal and immunodeficient mice and rats detected with bioluminescent imaging. Sci Rep 2017; 7:3597. [PMID: 28620164 PMCID: PMC5472566 DOI: 10.1038/s41598-017-03852-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 05/08/2017] [Indexed: 12/03/2022] Open
Abstract
As concerns increase about adenovirus type 5 (Ad5) being a safe gene transfer vector, it is important to evaluate its distribution, residence time, and possible toxicity in immunodeficient populations. To characterize the potential risk associated with different Ad5 vector delivery modes, we used immunocompetent and immunodeficient Rag2−/− animals to establish mouse and rat models that could be monitored with bioluminescent imaging following intramuscular or intravascular infection with an engineered replication-incompetent Ad5 virus carrying the firefly luciferase gene (Ad5-Fluc). The Ad5 vector was less well-tolerated by Rag2−/− animals than by wildtype ones, with delayed residence time, wider virus dissemination, less weight gain, and relatively severe pathological changes. In intravascularly Ad5-Fluc-infected Rag2−/− mice, systemic virus dissemination extended from the abdomen to the limbs and head on day 9 post-infection. Additionally, significant increases in plasma TNF-α and IFN-γ, which may be important factors in the heightened immunopathology in the liver and brain, were detected in the Rag2−/− mice 30 days after intravascular delivery. The Ad5 vector was better tolerated after intramuscular delivery than after intravascular delivery. Ad5-Fluc/Rag2−/− mice and rats can be used as reliable models of an immunodeficient population in which to evaluate the safety of Ad5-vectored vaccines or gene therapy products.
Collapse
|
10
|
Neuroimaging. IMAGING INFECTIONS 2017. [PMCID: PMC7123586 DOI: 10.1007/978-3-319-54592-9_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Imaging of infection in the CNS has been handled using cross-sectional imaging for more than two decades now resulting in a large array of descriptive diagnostic criteria, capable, in most circumstances of narrowing the differential diagnosis, detecting life-threatening complications and establishing baseline for assessment of treatment response. Limitations however exist, and in many circumstances, both cross-sectional imaging and nonspecific molecular imaging, such as 18F-FDG, fail to establish a diagnosis. The availability of pathogen-specific imaging agents/ligands would have a great effect on the management of patients with CNS infection. Besides early diagnosis, avoidance of diagnostic brain biopsies can have significant effect on the mortality and morbidity of patients.
Collapse
|
11
|
Bakhsheshian J, Wei BR, Hall MD, Simpson RM, Gottesman MM. In Vivo Bioluminescent Imaging of ATP-Binding Cassette Transporter-Mediated Efflux at the Blood-Brain Barrier. Methods Mol Biol 2016; 1461:227-39. [PMID: 27424909 PMCID: PMC10758286 DOI: 10.1007/978-1-4939-3813-1_19] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
We provide a detailed protocol for imaging ATP-binding cassette subfamily G member 2 (ABCG2) function at the blood-brain barrier (BBB) of transgenic mice. D-Luciferin is specifically transported by ABCG2 found on the apical side of endothelial cells at the BBB. The luciferase-luciferin enzymatic reaction produces bioluminescence, which allows a direct measurement of ABCG2 function at the BBB. Therefore bioluminescence imaging (BLI) correlates with ABCG2 function at the BBB and this can be measured by administering luciferin in a mouse model that expresses luciferase in the brain parenchyma. BLI allows for a relatively low-cost alternative for studying transporter function in vivo compared to other strategies such as positron emission tomography. This method for imaging ABCG2 function at the BBB can be used to investigate pharmacokinetic inhibition of the transporter.
Collapse
Affiliation(s)
- Joshua Bakhsheshian
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Bih-Rong Wei
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Matthew D Hall
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - R Mark Simpson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Michael M Gottesman
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
12
|
New researches and application progress of commonly used optical molecular imaging technology. BIOMED RESEARCH INTERNATIONAL 2014; 2014:429198. [PMID: 24696850 PMCID: PMC3947735 DOI: 10.1155/2014/429198] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 12/20/2013] [Indexed: 12/26/2022]
Abstract
Optical molecular imaging, a new medical imaging technique, is developed based on genomics, proteomics and modern optical imaging technique, characterized by non-invasiveness, non-radiativity, high cost-effectiveness, high resolution, high sensitivity and simple operation in comparison with conventional imaging modalities. Currently, it has become one of the most widely used molecular imaging techniques and has been applied in gene expression regulation and activity detection, biological development and cytological detection, drug research and development, pathogenesis research, pharmaceutical effect evaluation and therapeutic effect evaluation, and so forth, This paper will review the latest researches and application progresses of commonly used optical molecular imaging techniques such as bioluminescence imaging and fluorescence molecular imaging.
Collapse
|
13
|
Bioluminescent imaging of drug efflux at the blood-brain barrier mediated by the transporter ABCG2. Proc Natl Acad Sci U S A 2013; 110:20801-6. [PMID: 24297888 DOI: 10.1073/pnas.1312159110] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
ATP-binding cassette (ABC) transporters are a group of transmembrane proteins that maintain chemical homeostasis through efflux of compounds out of organelles and cells. Among other functions, ABC transporters play a key role in protecting the brain parenchyma by efflux of xenobiotics from capillary endothelial cells at the blood-brain barrier (BBB). They also prevent the entry of therapeutic drugs at the BBB, thereby limiting their efficacy. One of the key transporters playing this role is ABCG2. Although other ABC transporters can be studied through various imaging modalities, no specific probe exists for imaging ABCG2 function in vivo. Here we show that D-luciferin, the endogenous substrate of firefly luciferase, is a specific substrate for ABCG2. We hypothesized that ABCG2 function at the BBB could be evaluated by using bioluminescence imaging in transgenic mice expressing firefly luciferase in the brain. Bioluminescence signal in the brain of mice increased with coadministration of the ABCG2 inhibitors Ko143, gefitinib, and nilotinib, but not an ABCB1 inhibitor. This method for imaging ABCG2 function at the BBB will facilitate understanding of the function and pharmacokinetic inhibition of this transporter.
Collapse
|
14
|
GFAP expression as an indicator of disease severity in mouse models of Alexander disease. ASN Neuro 2013; 5:e00109. [PMID: 23432455 PMCID: PMC3604736 DOI: 10.1042/an20130003] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 02/21/2013] [Accepted: 02/22/2013] [Indexed: 12/19/2022] Open
Abstract
AxD (Alexander disease) is a rare disorder caused by heterozygous mutations in GFAP (glial fibrillary acidic protein) resulting in accumulation of the GFAP protein and elevation of Gfap mRNA. To test whether GFAP itself can serve as a biomarker of disease status or progression, we investigated two independent measures of GFAP expression in AxD mouse models, one using a genetic reporter of promoter activity and the other quantifying GFAP protein directly in a manner that could also be employed in human studies. Using a transgenic reporter line that expresses firefly luciferase under the control of the murine Gfap promoter (Gfap-luc), we found that luciferase activity reflected the regional CNS (central nervous system) variability of Gfap mRNA in Gfap+/+ mice, and increased in mice containing a point mutation in Gfap that mimics a common human mutation in AxD (R239H in the human sequence, and R236H in the murine sequence). In a second set of studies, we quantified GFAP protein in CSF (cerebrospinal fluid) taken from three different AxD mouse models and littermate controls. GFAP levels in CSF were increased in all three AxD models, in a manner corresponding to the concentrations of GFAP in brain. These studies demonstrate that transactivation of the Gfap promoter is an early and sustained indicator of the disease process in the mouse. Furthermore, GFAP in CSF serves as a potential biomarker that is comparable between mouse models and human patients.
Collapse
|
15
|
Systemic immune activation leads to neuroinflammation and sickness behavior in mice. Mediators Inflamm 2013; 2013:271359. [PMID: 23935246 PMCID: PMC3723093 DOI: 10.1155/2013/271359] [Citation(s) in RCA: 244] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 05/24/2013] [Accepted: 06/12/2013] [Indexed: 01/01/2023] Open
Abstract
Substantial evidence indicates an association between clinical depression and altered immune function. Systemic administration of bacterial lipopolysaccharide (LPS) is commonly used to study inflammation-associated behavioral changes in rodents. In these experiments, we tested the hypothesis that peripheral immune activation leads to neuroinflammation and depressive-like behavior in mice. We report that systemic administration of LPS induced astrocyte activation in transgenic GFAP-luc mice and increased immunoreactivity against the microglial marker ionized calcium-binding adapter molecule 1 in the dentate gyrus of wild-type mice. Furthermore, LPS treatment caused a strong but transient increase in cytokine levels in the serum and brain. In addition to studying LPS-induced neuroinflammation, we tested whether sickness could be separated from depressive-like behavior by evaluating LPS-treated mice in a panel of behavioral paradigms. Our behavioral data indicate that systemic LPS administration caused sickness and mild depressive-like behavior. However, due to the overlapping time course and mild effects on depression-related behavior per se, it was not possible to separate sickness from depressive-like behavior in the present rodent model.
Collapse
|
16
|
Nanogel-based PspA intranasal vaccine prevents invasive disease and nasal colonization by Streptococcus pneumoniae. Infect Immun 2013; 81:1625-34. [PMID: 23460513 DOI: 10.1128/iai.00240-13] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
To establish a safer and more effective vaccine against pneumococcal respiratory infections, current knowledge regarding the antigens common among pneumococcal strains and improvements to the system for delivering these antigens across the mucosal barrier must be integrated. We developed a pneumococcal vaccine that combines the advantages of pneumococcal surface protein A (PspA) with a nontoxic intranasal vaccine delivery system based on a nanometer-sized hydrogel (nanogel) consisting of a cationic cholesteryl group-bearing pullulan (cCHP). The efficacy of the nanogel-based PspA nasal vaccine (cCHP-PspA) was tested in murine pneumococcal airway infection models. Intranasal vaccination with cCHP-PspA provided protective immunity against lethal challenge with Streptococcus pneumoniae Xen10, reduced colonization and invasion by bacteria in the upper and lower respiratory tracts, and induced systemic and nasal mucosal Th17 responses, high levels of PspA-specific serum immunoglobulin G (IgG), and nasal and bronchial IgA antibody responses. Moreover, there was no sign of PspA delivery by nanogel to either the olfactory bulbs or the central nervous system after intranasal administration. These results demonstrate the effectiveness and safety of the nanogel-based PspA nasal vaccine system as a universal mucosal vaccine against pneumococcal respiratory infection.
Collapse
|
17
|
Huang NF, Okogbaa J, Babakhanyan A, Cooke JP. Bioluminescence imaging of stem cell-based therapeutics for vascular regeneration. Theranostics 2012; 2:346-54. [PMID: 22509198 PMCID: PMC3326722 DOI: 10.7150/thno.3694] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2011] [Accepted: 01/18/2012] [Indexed: 12/18/2022] Open
Abstract
Stem cell-based therapeutics show promise for treatment of vascular diseases. However, the survival of the cells after in vivo injection into diseased tissues remains a concern. In the advent of non-invasive optical imaging techniques such as bioluminescence imaging (BLI), cell localization and survival can be easily monitored over time. This approach has recently been applied towards monitoring stem cell treatments for vascular regeneration of the coronary or peripheral arteries. In this review, we will describe the application of BLI for tracking transplanted stem cells and associating their viability with therapeutic efficacy, in preclinical disease models of vascular disease.
Collapse
|
18
|
Cordeau P, Kriz J. Real-time imaging after cerebral ischemia: model systems for visualization of inflammation and neuronal repair. Methods Enzymol 2012; 506:117-33. [PMID: 22341222 DOI: 10.1016/b978-0-12-391856-7.00031-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Brain response to ischemic injury is characterized by initiation of a complex pathophysiological cascade comprising the events that may evolve over hours or several days and weeks after initial attack. At present, spatial and temporal dynamics of these events is not completely understood. To enable better understanding of the brain response to ischemic injury we developed and validated several novel transgenic mouse models of bioluminescence and fluorescence, allowing the noninvasive and time-lapse imaging of neuroinflammation, neuronal damage/stress and repair. These mice represent a powerful analytical tool for understanding in vivo pathology as well as the evaluating pharmacokinetics and longitudinal responses to drug therapies. Here, we describe the basic procedures of generating biophotonic mouse models for live imaging of microglial activation and neuronal stress and recovery, followed by a detailed description of in vivo bioluminescence imaging protocols used after experimental stroke.
Collapse
Affiliation(s)
- Pierre Cordeau
- Department of Psychiatry and Neuroscience, Laval University, Centre de Recherche du Centre Hospitalier de l’Université Laval, Québec, Québec City, Canada
| | | |
Collapse
|
19
|
Kataria J, Rukmangadachar LA, Hariprasad G, O J, Tripathi M, Srinivasan A. Two dimensional difference gel electrophoresis analysis of cerebrospinal fluid in tuberculous meningitis patients. J Proteomics 2011; 74:2194-203. [DOI: 10.1016/j.jprot.2011.06.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 06/01/2011] [Accepted: 06/18/2011] [Indexed: 12/14/2022]
|
20
|
Kagadis GC, Loudos G, Katsanos K, Langer SG, Nikiforidis GC. In vivosmall animal imaging: Current status and future prospects. Med Phys 2010; 37:6421-42. [DOI: 10.1118/1.3515456] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
21
|
Abstract
According to World Health Organization estimates, infectious organisms are responsible for approximately one in four deaths worldwide. Animal models play an essential role in the development of vaccines and therapeutic agents but large numbers of animals are required to obtain quantitative microbiological data by tissue sampling. Biophotonic imaging (BPI) is a highly sensitive, nontoxic technique based on the detection of visible light, produced by luciferase-catalysed reactions (bioluminescence) or by excitation of fluorescent molecules, using sensitive photon detectors. The development of bioluminescent/fluorescent microorganisms therefore allows the real-time noninvasive detection of microorganisms within intact living animals. Multiple imaging of the same animal throughout an experiment allows disease progression to be followed with extreme accuracy, reducing the number of animals required to yield statistically meaningful data. In the study of infectious disease, the use of BPI is becoming widespread due to the novel insights it can provide into established models, as well as the impact of the technique on two of the guiding principles of using animals in research, namely reduction and refinement. Here, we review the technology of BPI, from the instrumentation through to the generation of a photonic signal, and illustrate how the technique is shedding light on infection dynamics in vivo.
Collapse
Affiliation(s)
- Nuria Andreu
- Department of Medicine, Imperial College London, London, UK
| | | | | |
Collapse
|
22
|
Messing A, LaPash Daniels CM, Hagemann TL. Strategies for treatment in Alexander disease. Neurotherapeutics 2010; 7:507-15. [PMID: 20880512 PMCID: PMC2948554 DOI: 10.1016/j.nurt.2010.05.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Revised: 05/07/2010] [Accepted: 05/10/2010] [Indexed: 02/07/2023] Open
Abstract
Alexander disease is a rare and generally fatal disorder of the CNS, originally classified among the leukodystrophies because of the prominent myelin deficits found in young patients. The most common form of this disease affects infants, who often have profound mental retardation and a variety of developmental delays, but later onset forms also occur, sometimes with little or no white matter pathology at all. The pathological hallmark of Alexander disease is the inclusion body, known as Rosenthal fiber, within the cell bodies and processes of astrocytes. Recent genetic studies identified heterozygous missense mutations in glial fibrillary acidic protein (GFAP), the major intermediate filament protein in astrocytes, as the cause of nearly all cases of Alexander disease. These studies have transformed our view of this disorder and opened new directions for investigation and clinical practice, particularly with respect to diagnosis. Mechanisms by which expression of mutant forms of glial fibrillary acidic protein (GFAP) lead to the pleiotropic manifestations of disease (afflicting cell types beyond the ones expressing the mutant gene) are slowly coming into focus. Ideas are beginning to emerge that suggest several compelling therapeutic targets for interventions that might slow or arrest the evolution of the disease. This review will outline the rationale for pursuing these strategies, and highlight some of the critical issues that must be addressed in the planning of future clinical trials.
Collapse
Affiliation(s)
- Albee Messing
- Waisman Center, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA.
| | | | | |
Collapse
|
23
|
Hutter E, Maysinger D. Gold nanoparticles and quantum dots for bioimaging. Microsc Res Tech 2010; 74:592-604. [DOI: 10.1002/jemt.20928] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Accepted: 07/26/2010] [Indexed: 12/21/2022]
|
24
|
Luker KE, Luker GD. Bioluminescence imaging of reporter mice for studies of infection and inflammation. Antiviral Res 2010; 86:93-100. [PMID: 20417377 DOI: 10.1016/j.antiviral.2010.02.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Revised: 02/01/2010] [Accepted: 02/01/2010] [Indexed: 01/02/2023]
Abstract
In vivo bioluminescence imaging offers the opportunity to study biological processes in living animals, and the study of viral infections and host immune responses can be enhanced substantially through this imaging modality. For most studies of viral pathogenesis and effects of anti-viral therapies, investigators have used recombinant viruses engineered to express a luciferase enzyme. This strategy requires stable insertion of an imaging reporter gene into the viral genome, which is not feasible for many RNA viruses, and provides data on the viral component of pathogenesis but not on the host. Genetically engineered mice with luciferase reporters for specific viral or host genes provide opportunities to overcome these limitations and expand applications of bioluminescence imaging in viral infection and therapy. We review several different types of reporter mice for bioluminescence imaging, including animals that permit in vivo detection of viral replication, trafficking of immune cells, activation of key genes in host immunity to viral infection, and response to tissue damage. By utilizing luciferase enzymes with different emission spectra and/or substrates, it is possible to monitor two different biologic processes in the same animal, such as pathogen replication and sites of tissue injury. Combining imaging reporter viruses with genetically engineered reporter mice is expected to substantially enhance the power of bioluminescence imaging for quantitative studies of viral and host factors that control disease outcome and effects of established and new therapeutic agents.
Collapse
Affiliation(s)
- Kathryn E Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School, Ann Arbor, MI 48019-2200, USA.
| | | |
Collapse
|
25
|
Signore A, Mather SJ, Piaggio G, Malviya G, Dierckx RA. Molecular imaging of inflammation/infection: nuclear medicine and optical imaging agents and methods. Chem Rev 2010; 110:3112-45. [PMID: 20415479 DOI: 10.1021/cr900351r] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- A Signore
- Nuclear Medicine Unit, II Faculty of Medicine and Surgery, Sapienza University of Rome, Rome, Italy.
| | | | | | | | | |
Collapse
|
26
|
Evaluation of biophotonic imaging to estimate bacterial burden in mice infected with highly virulent compared to less virulent Streptococcus pneumoniae serotypes. Antimicrob Agents Chemother 2010; 54:3155-60. [PMID: 20530224 DOI: 10.1128/aac.00310-10] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bioluminescence imaging is an innovative, noninvasive tool to analyze infectious disease progression under real-life conditions in small laboratory animals. However, the relevance of bioluminescence imaging to monitor invasive compared to noninvasive bacterial infections of the lung has not been examined so far. In the current study, we systematically evaluated the importance of bioluminescence imaging to monitor pneumococcal disease progression by correlating biophotonic signals with lung bacterial loads in two mouse strains (BALB/c, C57BL/6) infected with either self-glowing, bioluminescent serotype 19 Streptococcus pneumoniae causing focal pneumonia or serotype 2 S. pneumoniae causing invasive pneumococcal disease. The best correlations between bioluminescence signals and lung CFU counts were observed in BALB/c mice compared to C57BL/6 mice just on day 3 after infection with invasive serotype 2 S. pneumoniae, while excellent correlations between photon counts and bacterial loads were observed in isolated lungs of BALB/c and C57BL/6 mice, irrespective of the employed pneumococcal serotype. Moreover, good correlations between biophotonic signals and CFU counts were also observed in mice upon infection with serotype 19 S. pneumoniae causing focal pneumonia in mice, again with best correlation values obtained for BALB/c mice at day 3 postinfection. Collectively, we show that the relevance of biophotonic imaging to monitor S. pneumoniae-induced lung infections in mice is largely influenced by the disease model under investigation. The provided data may be important for studies of infectious diseases.
Collapse
|
27
|
Andreu N, Zelmer A, Fletcher T, Elkington PT, Ward TH, Ripoll J, Parish T, Bancroft GJ, Schaible U, Robertson BD, Wiles S. Optimisation of bioluminescent reporters for use with mycobacteria. PLoS One 2010; 5:e10777. [PMID: 20520722 PMCID: PMC2875389 DOI: 10.1371/journal.pone.0010777] [Citation(s) in RCA: 229] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 04/27/2010] [Indexed: 01/01/2023] Open
Abstract
Background Mycobacterium tuberculosis, the causative agent of tuberculosis, still represents a major public health threat in many countries. Bioluminescence, the production of light by luciferase-catalyzed reactions, is a versatile reporter technology with multiple applications both in vitro and in vivo. In vivo bioluminescence imaging (BLI) represents one of its most outstanding uses by allowing the non-invasive localization of luciferase-expressing cells within a live animal. Despite the extensive use of luminescent reporters in mycobacteria, the resultant luminescent strains have not been fully applied to BLI. Methodology/Principal Findings One of the main obstacles to the use of bioluminescence for in vivo imaging is the achievement of reporter protein expression levels high enough to obtain a signal that can be detected externally. Therefore, as a first step in the application of this technology to the study of mycobacterial infection in vivo, we have optimised the use of firefly, Gaussia and bacterial luciferases in mycobacteria using a combination of vectors, promoters, and codon-optimised genes. We report for the first time the functional expression of the whole bacterial lux operon in Mycobacterium tuberculosis and M. smegmatis thus allowing the development of auto-luminescent mycobacteria. We demonstrate that the Gaussia luciferase is secreted from bacterial cells and that this secretion does not require a signal sequence. Finally we prove that the signal produced by recombinant mycobacteria expressing either the firefly or bacterial luciferases can be non-invasively detected in the lungs of infected mice by bioluminescence imaging. Conclusions/Significance While much work remains to be done, the finding that both firefly and bacterial luciferases can be detected non-invasively in live mice is an important first step to using these reporters to study the pathogenesis of M. tuberculosis and other mycobacterial species in vivo. Furthermore, the development of auto-luminescent mycobacteria has enormous ramifications for high throughput mycobacterial drug screening assays which are currently carried out either in a destructive manner using LuxAB or the firefly luciferase.
Collapse
Affiliation(s)
- Nuria Andreu
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Andrea Zelmer
- Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Taryn Fletcher
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Paul T. Elkington
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Theresa H. Ward
- Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Jorge Ripoll
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
| | - Tanya Parish
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Infectious Diseases Research Institute, Seattle, Washington, United States of America
| | - Gregory J. Bancroft
- Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Ulrich Schaible
- Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Department of Molecular Infection Research, Research Center Borstel, Borstel, Germany
| | | | - Siouxsie Wiles
- Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
- * E-mail:
| |
Collapse
|
28
|
Sharma PK, Engels E, Van Oeveren W, Ploeg RJ, van Henny der Mei C, Busscher HJ, Van Dam GM, Rakhorst G. Spatiotemporal progression of localized bacterial peritonitis before and after open abdomen lavage monitored by in vivo bioluminescent imaging. Surgery 2009; 147:89-97. [PMID: 19733882 DOI: 10.1016/j.surg.2009.05.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Accepted: 05/21/2009] [Indexed: 01/13/2023]
Abstract
BACKGROUND Bacterial peritonitis is a life-threatening abdominal infection associated with high morbidity and mortality. The rat is a popular animal model for studying peritonitis and its treatment, but longitudinal monitoring of the progression of peritonitis in live animals has been impossible until now and thus required a large number of animals. Our objective was to develop a noninvasive in vivo imaging technique to monitor the spatiotemporal spread of bacterial peritonitis. METHODS Peritonitis was induced in 8 immunocompetent male Wistar rats by placing fibrin clots containing 5x10(8) cells of both Bacteroides fragilis (American Type Tissue Culture [ATCC)] 25,285 and bioluminescent Escherichia coli Xen14. After 1 or 2 days, infected clots were removed and open abdomen lavage was performed. In vivo bioluminescent imaging was used to monitor the spread of peritonitis. RESULTS Bioluminescent in vivo imaging showed an increase in the area of spread, and the number of E. coli tripled into the rat's abdominal cavity on day 1 after clot insertion; however, on day 2, encapsulation of the clot confined bacterial spread. Bioluminescent E. coli respread over the peritoneal cavity after lavage; within 10 days, however, in vivo imaging showed a decrease of 3-4 orders of magnitude in bacterial load. CONCLUSION Bioluminescent in vivo imaging can be effectively used to monitor the spatiotemporal behavior of the peritonitis during 3 different stages of the disease process: initiation, treatment, and follow-up. Imaging allows researchers to repeatedly image the same animal, thereby reducing variability and providing greater confidence in determining treatment efficacies for therapeutic interventions using a small number of animals.
Collapse
Affiliation(s)
- Prashant K Sharma
- Department of Biomedical Engineering, University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Prions are infectious proteins that cause fatal neurodegenerative diseases. Because astrocytic gliosis marked by the deposition of fibrils composed of GFAP is a prominent feature of prion disease, we asked whether GFAP might be used as a surrogate marker for prions. To interrogate this posit, we inoculated prions into transgenic (Tg) mice expressing luciferase (luc) under the GFAP gene (Gfap) promoter, denoted Tg(Gfap-luc) mice. Weekly noninvasive, bioluminescence imaging (BLI) detected an increase in light emitted from the brains of Tg(Gfap-luc) mice at approximately 55 d after inoculation and approximately 62 d before neurologic deficits appeared. To determine whether BLI could be used as a proxy bioassay for prion infectivity, we performed endpoint titrations of prions in Tg(Gfap-luc) mice. BLI bioassays were as or more sensitive than those determined by the onset of neurological dysfunction, and were completed in approximately half the time. Our studies argue that BLI is likely to be a suitable surrogate for measuring prion infectivity, and might be useful in the study of Tg mouse models for other neurodegenerative illnesses.
Collapse
|
30
|
Cho W, Hagemann TL, Johnson DA, Johnson JA, Messing A. Dual transgenic reporter mice as a tool for monitoring expression of glial fibrillary acidic protein. J Neurochem 2009; 110:343-51. [PMID: 19457099 DOI: 10.1111/j.1471-4159.2009.06146.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Glial fibrillary acidic protein (GFAP) is the major intermediate filament protein of astrocytes, and its expression changes dramatically during development and following injury. To facilitate study of the regulation of GFAP expression, we have generated dual transgenic mice expressing both firefly luciferase under the control of a 2.2 kb human GFAP promoter and Renilla luciferase under the control of a 0.5 kb human Glyceraldehyde 3 phosphate dehydrogenase (GAPDH) promoter for normalization of the GFAP signal. The GFAP-fLuc was highly expressed in brain compared to other tissues, and was limited to astrocytes, whereas the GAPDH-RLuc was more widely expressed. Normalization of the GFAP signal to the GAPDH signal reduced the inter-individual variability compared to using the GFAP signal alone. The GFAP/GAPDH ratio correctly reflected the up-regulation of GFAP that occurs following retinal degeneration in FVB/N mice because of the rd mutation. Following kainic acid-induced seizures, changes in the GFAP/GAPDH ratio precede those in total GFAP protein. In knock-in mice expressing the R236H Alexander disease mutant, GFAP promoter activity is only transiently elevated and may not entirely account for the accumulation of GFAP protein that takes place.
Collapse
Affiliation(s)
- Woosung Cho
- Waisman Center and Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA.
| | | | | | | | | |
Collapse
|
31
|
Lalancette-Hébert M, Phaneuf D, Soucy G, Weng YC, Kriz J. Live imaging of Toll-like receptor 2 response in cerebral ischaemia reveals a role of olfactory bulb microglia as modulators of inflammation. ACTA ACUST UNITED AC 2009; 132:940-54. [PMID: 19153151 DOI: 10.1093/brain/awn345] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Activation of microglial cells in response to ischaemic injury, inflammatory and/or immune stimuli is associated with the marked induction of Toll-like receptor 2 (TLR2). At present, little is known about the spatial and temporal sequence of events, micro-regional specificities and the potential long term role of the TLR2 response to brain injuries. To investigate microglial activation/TLR2 response in real time, we generated a transgenic mouse model bearing the dual reporter system luciferase/green fluorescent protein under transcriptional control of a murine TLR2 promoter. In this model, transcriptional activation of TLR2 was visualized in the brains of live animals using biophotonic/bioluminescence molecular imaging and a high resolution/sensitivity charged coupled device camera. It was found that TLR2 induction/microglial activation has a marked chronic component after ischaemic injury and may last several months after the initial attack. The pro-inflammatory response was not restricted to the site of ischaemic injury but was also evident in the olfactory bulb. A significant TLR2 response was first seen in the olfactory bulb 6 h after stroke and several hours before the increase in photon emission over the site of infarction. This sequence of events was further confirmed by immunohistochemistry. A similar early TLR2 response from olfactory bulb microglia was observed in the brain's immune response to pathogens. We therefore propose that, owing to their unique situation, receiving and translating numerous inputs from the brain as well as from the environment, olfactory bulb microglia may serve as sensors and/or modulators of brain inflammation.
Collapse
Affiliation(s)
- M Lalancette-Hébert
- Department of Anatomy and Physiology, Laval University, Centre de Recherche du Centre Hospitalier de l'Université Laval, Québec, Canada G1V 4G2
| | | | | | | | | |
Collapse
|
32
|
Luker KE, Luker GD. Applications of bioluminescence imaging to antiviral research and therapy: multiple luciferase enzymes and quantitation. Antiviral Res 2008; 78:179-87. [PMID: 18358543 DOI: 10.1016/j.antiviral.2008.01.158] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Revised: 01/22/2008] [Accepted: 01/25/2008] [Indexed: 11/18/2022]
Abstract
Bioluminescence imaging (BLI) has emerged as a powerful new modality for studies of viral infection and therapy in small animal models. BLI technology captures the light emitted from different luciferase enzymes to detect sites of viral infection and quantify viral replication in the context of a living animal. In this review, we discuss the biochemical features of various luciferase enzymes and modifications to these enzymes that can greatly enhance their ability to image viral infection, host responses and the effects of therapy. We also describe BLI instrumentation and technical aspects of BLI needed to optimize imaging data. Examples of BLI for quantitative analysis of viral infection and in vivo monitoring of antiviral and antibacterial therapy are presented to highlight the potential for BLI to accelerate discovery of new antiviral agents and determine efficacy of antiviral compounds. Ongoing research to use multiple luciferase enzymes to image viral infection, host immune signaling pathways, and cell trafficking in the same animal will continue to advance BLI for longitudinal, real-time quantification and analyses of viral infection and pre-clinical testing of promising therapeutic agents.
Collapse
Affiliation(s)
- Kathryn E Luker
- Department of Radiology, University of Michigan Medical School, 109 Zina Pitcher Place, A526 BSRB, Ann Arbor, MI 48109-2200, United States
| | | |
Collapse
|
33
|
Massoud TF, Singh A, Gambhir SS. Noninvasive molecular neuroimaging using reporter genes: part II, experimental, current, and future applications. AJNR Am J Neuroradiol 2008; 29:409-18. [PMID: 18272565 DOI: 10.3174/ajnr.a0863] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
SUMMARY In this second article, we review the various strategies and applications that make use of reporter genes for molecular imaging of the brain in living subjects. These approaches are emerging as valuable tools for monitoring gene expression in diverse applications in laboratory animals, including the study of gene-targeted and trafficking cells, gene therapies, transgenic animals, and more complex molecular interactions within the central nervous system. Further development of more sensitive and selective reporters, combined with improvements in detection technology, will consolidate the position of in vivo reporter gene imaging as a versatile technique for greater understanding of intracellular biologic processes and underlying molecular neuropathology and will potentially establish a future role in the clinical management of patients with neurologic diseases.
Collapse
Affiliation(s)
- T F Massoud
- Department of Radiology, Section of Neuroradiology, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, Cambridge, UK.
| | | | | |
Collapse
|
34
|
Brandt CT, Simonsen H, Liptrot M, Søgaard LV, Lundgren JD, Ostergaard C, Frimodt-Møller N, Rowland IJ. In vivo study of experimental pneumococcal meningitis using magnetic resonance imaging. BMC Med Imaging 2008; 8:1. [PMID: 18194516 PMCID: PMC2253532 DOI: 10.1186/1471-2342-8-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2007] [Accepted: 01/14/2008] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Magnetic Resonance Imaging (MRI) methods were evaluated as a tool for the study of experimental meningitis. The identification and characterisation of pathophysiological parameters that vary during the course of the disease could be used as markers for future studies of new treatment strategies. METHODS Rats infected intracisternally with S. pneumoniae (n = 29) or saline (n = 13) were randomized for imaging at 6, 12, 24, 30, 36, 42 or 48 hours after infection. T1W, T2W, quantitative diffusion, and post contrast T1W images were acquired at 4.7 T. Dynamic MRI (dMRI) was used to evaluate blood-brain-barrier (BBB) permeability and to obtain a measure of cerebral and muscle perfusion. Clinical- and motor scores, bacterial counts in CSF and blood, and WBC counts in CSF were measured. RESULTS MR images and dMRI revealed the development of a highly significant increase in BBB permeability (P < 0.002) and ventricle size (P < 0.0001) among infected rats. Clinical disease severity was closely related to ventricle expansion (P = 0.024). Changes in brain water distribution, assessed by ADC, and categorization of brain 'perfusion' by cortex DeltaSI(bolus) were subject to increased inter-rat variation as the disease progressed, but without overall differences compared to uninfected rats (P > 0.05). Areas of well-'perfused' muscle decreased with the progression of infection indicative of septicaemia (P = 0.05). CONCLUSION The evolution of bacterial meningitis was successfully followed in-vivo with MRI. Increasing BBB-breakdown and ventricle size was observed in rats with meningitis whereas changes in brain water distribution were heterogeneous. MRI will be a valuable technique for future studies aiming at evaluating or optimizing adjunctive treatments.
Collapse
Affiliation(s)
- Christian T Brandt
- National Center for Antimicrobials and Infection Control, Statens Serum Institut, Copenhagen, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Louis DN. Molecular pathology of malignant gliomas. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2007; 2:277-305. [PMID: 18039109 DOI: 10.1146/annurev.pathol.2.010506.091930] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Malignant gliomas, the most common type of primary brain tumor, are a spectrum of tumors of varying differentiation and malignancy grades. These tumors may arise from neural stem cells and appear to contain tumor stem cells. Early genetic events differ between astrocytic and oligodendroglial tumors, but all tumors have an initially invasive phenotype, which complicates therapy. Progression-associated genetic alterations are common to different tumor types, targeting growth-promoting and cell cycle control pathways and resulting in focal hypoxia, necrosis, and angiogenesis. Knowledge of malignant glioma genetics has already impacted clinical management of these tumors, and researchers hope that further knowledge of the molecular pathology of malignant gliomas will result in novel therapies.
Collapse
Affiliation(s)
- David N Louis
- Molecular Pathology Unit, Department of Pathology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA.
| |
Collapse
|
36
|
Abstract
Optical techniques, such as bioluminescence and fluorescence, are emerging as powerful new modalities for molecular imaging in disease and therapy. Combining innovative molecular biology and chemistry, researchers have developed optical methods for imaging a variety of cellular and molecular processes in vivo, including protein interactions, protein degradation, and protease activity. Whereas optical imaging has been used primarily for research in small-animal models, there are several areas in which optical molecular imaging will translate to clinical medicine. In this review, we summarize recent advances in optical techniques for molecular imaging and the potential impact for clinical medicine.
Collapse
Affiliation(s)
- Gary D Luker
- University of Michigan, Ann Arbor, Michigan, USA.
| | | |
Collapse
|
37
|
Enninga J, Sansonetti P, Tournebize R. Roundtrip explorations of bacterial infection: from single cells to the entire host and back. Trends Microbiol 2007; 15:483-90. [PMID: 17983749 DOI: 10.1016/j.tim.2007.10.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2007] [Revised: 09/04/2007] [Accepted: 10/24/2007] [Indexed: 01/05/2023]
Abstract
Host-pathogen interactions are highly regulated, dynamic processes that take place at the molecular, cellular and organ level. Innovative imaging technologies have emerged recently to investigate the underlying mechanisms of host-pathogen interactions. Innovations in fluorescence microscopy enable functional studies on the single-cell level. New light microscopes have been developed that improve the resolution to less than 100 nm. At the other extreme, intravital microscopy enables the correlation of cellular events on the organ level. This is also achieved by alternatives to microscopy such as bioluminescence, positron-emission tomography and magnetic resonance imaging. The methodologies described here will have a tremendous effect on our understanding of host-pathogen interactions.
Collapse
Affiliation(s)
- Jost Enninga
- Unité de Pathogénie Microbienne, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris cedex 15, France.
| | | | | |
Collapse
|
38
|
Abstract
Over the last thirteen years, the field of optical imaging has expanded from in vitro fluorescence microscopy of cells to in vivo imaging of living animals. Recent advances in optical imaging of bacterial infection have been propelled by the invention of genetic methods that produce fluorescent and bioluminescent bacteria, and also the discovery of synthetic fluorescent probes that selectively target bacterial cell surfaces. Optical imaging is an effective method of conducting longitudinal studies of bacterial infection in small animals such as nude mice. It can be used to address questions in medical microbiology concerning migration and colonization and it is an attractive method for determining the efficacy of antibiotic therapies.
Collapse
|
39
|
Abstract
Bioluminescence imaging (BLI) has emerged as a powerful new method to analyse infectious diseases in animal models. BLI offers real-time monitoring of spatial and temporal progression of infection in the same animal, as opposed to euthanizing a cohort of animals and quantifying colony or plaque forming units at multiple time points. Pathogens or mice are engineered to express genetically encoded luciferase enzymes from bacteria, insects, or the sea pansy. The seminal study showing the feasibility of detecting microbially generated luminescence within a living mouse was published by Contag and colleagues in 1995, using Salmonella typhimurium transformed with the lux operon from Photorhabdus luminescens. Following this, they and others performed many studies of infection by bioluminescent Gram-negative and Gram-positive bacteria. Viruses can also be engineered to encode luciferase. Our laboratory has used bioluminescent reporter viruses to follow HSV and vaccinia pathogenesis; others have used an alphavirus or novirhabdovirus. Recently, even eukaryotic parasites Plasmodium, Leishmania and Toxoplasma have been transformed with luciferase and yielded unique insights into their in vivo behaviour. We expect that both the range of organisms and the molecular events able to be studied by BLI will continue to expand, yielding important insights into mechanisms of pathogenesis.
Collapse
Affiliation(s)
- Martha Hutchens
- Immunology Program, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA
| | | |
Collapse
|
40
|
Correa-Cerro LS, Mandell JW. Molecular mechanisms of astrogliosis: new approaches with mouse genetics. J Neuropathol Exp Neurol 2007; 66:169-76. [PMID: 17356378 DOI: 10.1097/01.jnen.0000248555.53079.d5] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Astrocytes are increasingly being recognized as dynamic participants in many aspects of normal central nervous system function. In disease states, reactive astrocytes undergo complex phenotypic changes, generically referred to as astrogliosis. Unraveling the functions of reactive astrocytes and underlying molecular mechanisms is a difficult problem. The use of genetically modified mice is beginning to yield some answers to long-standing questions in the field. What are the functions of reactive astrocytes? What extracellular factors and intracellular signaling mechanisms are responsible for astrocyte activation in various forms of neural injury? In this review we will highlight studies using astrocyte reporter lines for cellular imaging and lineage tracing, as well as gain- and loss-of-function mutations that have begun to shed light on mechanisms of astrogliosis.
Collapse
Affiliation(s)
- Lina S Correa-Cerro
- Department of Pathology, Division of Neuropathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | | |
Collapse
|
41
|
Davidsen T, Koomey M, Tønjum T. Microbial genome dynamics in CNS pathogenesis. Neuroscience 2007; 145:1375-87. [PMID: 17367950 DOI: 10.1016/j.neuroscience.2007.01.059] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2006] [Revised: 01/19/2007] [Accepted: 01/19/2007] [Indexed: 10/23/2022]
Abstract
The balancing act between microbes and their host in commensal and disease states needs to be deciphered in order to fully treat and combat infectious diseases. The elucidation of microbial genome dynamics in each instance is therefore required. In this context, the major bacterial meningitis pathogens are Neisseria meningitidis, Haemophilus influenzae and Streptococcus pneumoniae. In prokaryotic CNS pathogenesis both the intact organism as well as its released components can elicit disease, often resulting in neurological sequelae, neurodegeneration or fatal outcome. The study of microbial virulence in CNS disease is expected to generate findings that yield new information on the general mechanisms of brain edema and excitatory neuronal disturbances due to meningitis, with significant potential for discoveries that can directly influence and inspire new strategies for prevention and treatment of this serious disease.
Collapse
Affiliation(s)
- T Davidsen
- Centre for Molecular Biology and Neuroscience, Institute of Microbiology, Rikshospitalet-Radiumhospitalet Medical Centre, Sognsvannsveien 20, NO-0027 Oslo, Norway
| | | | | |
Collapse
|
42
|
Contag CH. Molecular imaging using visible light to reveal biological changes in the brain. Neuroimaging Clin N Am 2007; 16:633-54, ix. [PMID: 17148024 DOI: 10.1016/j.nic.2006.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Advances in imaging have enabled the study of cellular and molecular processes in the context of the living body that include cell migration patterns, location and extent of gene expression, degree of protein-protein interaction, and levels of enzyme activity. These tools, which operate over a range of scales, resolutions, and sensitivities, have opened up broad new areas of investigation where the influence of organ systems and functional circulation is intact. There are a myriad of imaging modalities available, each with its own advantages and disadvantages, depending on the specific application. Among these modalities, optical imaging techniques, including in vivo bioluminescence imaging and fluorescence imaging, use visible light to interrogate biology in the living body. Optimal imaging with these modalities require that the appropriate marker be used to tag the process of interest to make it uniquely visible using a particular imaging technology. For each optical modality, there are various labels to choose from that range from dyes that permit tissue contrast and dyes that can be activated by enzymatic activity, to gene-encoding proteins with optical signatures that can be engineered into specific biological processes. This article provides and overview of optical imaging technologies and commonly used labels, focusing on bioluminescence and fluorescence, and describes several examples of how these tools are applied to biological questions relating to the central nervous system.
Collapse
Affiliation(s)
- Christopher H Contag
- Departments of Pediatrics, Microbiology & Immunology and Radiology, E150 Clark Center, MC 5427, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
43
|
Spreer A, Gerber J, Hanssen M, Schindler S, Hermann C, Lange P, Eiffert H, Nau R. Dexamethasone increases hippocampal neuronal apoptosis in a rabbit model of Escherichia coli meningitis. Pediatr Res 2006; 60:210-5. [PMID: 16864706 DOI: 10.1203/01.pdr.0000227553.47378.9f] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Mortality and long-term sequelae rates are high among adults and children with acute bacterial meningitis. Adjunctive treatment with dexamethasone has been shown to reduce systemic complications in bacterial meningitis patients, but corticosteroid treatment may have detrimental effects on hippocampal function. We evaluated the effect of dexamethasone treatment in addition to antibiotic therapy in a rabbit model of Escherichia coli meningitis. A moderate anti-inflammatory effect of dexamethasone could be demonstrated with respect to the inflammatory mediator prostaglandin E2, whereas no significant effect of dexamethasone on tumor necrosis factor-alpha, cerebrospinal fluid pleocytosis, protein, lactate, indicators of global neuronal damage, or blood gas analysis was found. Dexamethasone, however, increased the rate of apoptotic neurons in the granular layer of the hippocampal dentate gyrus. In view of the proapoptotic effect of adjunctive dexamethasone on hippocampal neuronal cells in animal models of Gram-positive and Gram-negative meningitis, the application of dexamethasone should be considered carefully in those forms of bacterial meningitis for which no evidence-based data of beneficial effect in humans are available, such as neonatal meningitis, bacillary Gram-negative meningitis or nosocomial forms of meningitis (e.g. following neurosurgery).
Collapse
Affiliation(s)
- Annette Spreer
- Department of Neurology, University Hospital, Georg-August-University of Göttingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
In vivo microscopy is an exciting tool for neurological research because it can reveal how single cells respond to damage of the nervous system. This helps us to understand how diseases unfold and how therapies work. Here, we review the optical imaging techniques used to visualize the different parts of the nervous system, and how they have provided fresh insights into the aetiology and therapeutics of neurological diseases. We focus our discussion on five areas of neuropathology (trauma, degeneration, ischaemia, inflammation and seizures) in which in vivo microscopy has had the greatest impact. We discuss the challenging issues in the field, and argue that the convergence of new optical and non-optical methods will be necessary to overcome these challenges.
Collapse
Affiliation(s)
- Thomas Misgeld
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, USA.
| | | |
Collapse
|