1
|
Yamaguchi D, Kamoshida G, Kawakubo S, Azuma S, Tsuji T, Kitada N, Saito-Moriya R, Yamada N, Tanaka R, Okuda A, Ueyama K, Isaka S, Tomita M, Nakano R, Morita Y, Yano H, Maki SA, Yahiro K, Kato S. Near-infrared in vivo imaging system for dynamic visualization of lung-colonizing bacteria in mouse pneumonia. Microbiol Spectr 2024; 12:e0082824. [PMID: 39287455 PMCID: PMC11537041 DOI: 10.1128/spectrum.00828-24] [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: 04/01/2024] [Accepted: 08/13/2024] [Indexed: 09/19/2024] Open
Abstract
In vivo imaging of bacterial infection models enables noninvasive and temporal analysis of individuals, enhancing our understanding of infectious disease pathogenesis. Conventional in vivo imaging methods for bacterial infection models involve the insertion of the bacterial luciferase LuxCDABE into the bacterial genome, followed by imaging using an expensive ultrasensitive charge-coupled device (CCD) camera. However, issues such as limited light penetration into the body and lack of versatility have been encountered. We focused on near-infrared (NIR) light, which penetrates the body effectively, and attempted to establish an in vivo imaging method to evaluate the number of lung-colonizing bacteria during the course of bacterial pneumonia. This was achieved by employing a novel versatile system that combines plasmid-expressing firefly luciferase bacteria, NIR substrate, and an inexpensive, scientific complementary metal-oxide semiconductor (sCMOS) camera. The D-luciferin derivative "TokeOni," capable of emitting NIR bioluminescence, was utilized in a mouse lung infection model of Acinetobacter baumannii, an opportunistic pathogen that causes pneumonia and is a concern due to drug resistance. TokeOni exhibited the highest sensitivity in detecting bacteria colonizing the mouse lungs compared with other detection systems such as LuxCDABE, enabling the monitoring of changes in bacterial numbers over time and the assessment of antimicrobial agent efficacy. Additionally, it was effective in detecting A. baumannii clinical isolates and Klebsiella pneumoniae. The results of this study are expected to be used in the analysis of animal models of infectious diseases for assessing the efficacy of therapeutic agents and understanding disease pathogenesis. IMPORTANCE Conventional animal models of infectious diseases have traditionally relied upon average assessments involving numerous individuals, meaning they do not directly reflect changes in the pathology of an individual. Moreover, in recent years, ethical concerns have resulted in the demand to reduce the number of animals used in such models. Although in vivo imaging offers an effective approach for longitudinally evaluating the pathogenesis of infectious diseases in individual animals, a standardized method has not yet been established. To our knowledge, this study is the first to develop a highly versatile in vivo pulmonary bacterial quantification system utilizing near-infrared luminescence, plasmid-mediated expression of firefly luciferase in bacteria, and a scientific complementary metal-oxide semiconductor camera. Our research holds promise as a useful tool for assessing the efficacy of therapeutic drugs and pathogenesis of infectious diseases.
Collapse
Affiliation(s)
- Daiki Yamaguchi
- Laboratory of Microbiology and Infection Control, Kyoto Pharmaceutical University, Kyoto, Japan
- Laboratory of Pharmacological and Experimental Therapeutics, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Go Kamoshida
- Laboratory of Microbiology and Infection Control, Kyoto Pharmaceutical University, Kyoto, Japan
- Department of Infection Control Science, Meiji Pharmaceutical University, Tokyo, Japan
| | - Syun Kawakubo
- Laboratory of Microbiology and Infection Control, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Saki Azuma
- Laboratory of Microbiology and Infection Control, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Takamitsu Tsuji
- Laboratory of Microbiology and Infection Control, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Nobuo Kitada
- Graduate School of Informatics and Engineering, The University of Electro-Communications, Chofu, Japan
| | - Ryohei Saito-Moriya
- Department of Chemical and Biological Sciences, Faculty of Science, Japan Women’s University, Tokyo, Japan
| | - Noriteru Yamada
- Laboratory of Microbiology and Infection Control, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Rentaro Tanaka
- Laboratory of Microbiology and Infection Control, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Ayane Okuda
- Laboratory of Microbiology and Infection Control, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Keisuke Ueyama
- Laboratory of Microbiology and Infection Control, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Shingo Isaka
- Laboratory of Microbiology and Infection Control, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Manaha Tomita
- Laboratory of Microbiology and Infection Control, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Ryuichi Nakano
- Department of Microbiology and Infectious Diseases, Nara Medical University, Nara, Japan
| | - Yuji Morita
- Department of Infection Control Science, Meiji Pharmaceutical University, Tokyo, Japan
| | - Hisakazu Yano
- Department of Microbiology and Infectious Diseases, Nara Medical University, Nara, Japan
| | - Shojiro A. Maki
- Graduate School of Informatics and Engineering, The University of Electro-Communications, Chofu, Japan
| | - Kinnosuke Yahiro
- Laboratory of Microbiology and Infection Control, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Shinichi Kato
- Laboratory of Pharmacological and Experimental Therapeutics, Kyoto Pharmaceutical University, Kyoto, Japan
| |
Collapse
|
2
|
Yamamoto K, Torigoe S, Tsujimura Y, Asaka MN, Okumura K, Ato M. In vivo imaging identified efficient antimicrobial treatment against Mycobacterium marinum infection in mouse footpads. Sci Rep 2024; 14:24343. [PMID: 39420066 PMCID: PMC11487254 DOI: 10.1038/s41598-024-75207-5] [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: 05/20/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
Mycobacterium marinum (M. marinum) is the most common causative bacteria of cutaneous non-tuberculous mycobacterial (NTM) infections, including fish tank granuloma. Treating M. marinum-caused infection takes longer than other NTM diseases because M. marinum is less susceptible to antimicrobial agents. A standard treatment regimen for M. marinum infection has not been established yet, and few in vivo experiments have been performed in mammals to evaluate the bactericidal effects of antimicrobials. In this study, we developed a noninvasive in vivo imaging method to assess the therapeutic efficacy of antimicrobials against M. marinum infection. The data obtained using fluorescent protein or bioluminescence from luciferase will offer valuable insights into bacteria visualization across various bacterial infections. Furthermore, through this imaging technique, we demonstrated that combining clarithromycin, rifampicin, ethambutol, and minocycline effectively cleared M. marinum from the footpad. Granulomas with necrotic abscesses formed on the footpad of M. marinum-infected mice, primarily due to neutrophils involved in the host's cell-mediated immune response. Inflammatory cytokine and chemokine levels significantly increased 7 days post-infection, aligning with the footpad swelling and granuloma formation observed in the untreated group. Interestingly, immune mediators and cells induced by M. marinum footpad infection were crucial factors associated with hypersensitivity and granuloma formation, as seen in pulmonary tuberculosis. This novel imaging analysis using a cutaneous NTM mouse model might be a powerful tool for the comprehensive analysis of mycobacterial infections.
Collapse
Affiliation(s)
- Kentaro Yamamoto
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Aoba-cho, Higashimurayama, Tokyo, Japan.
| | - Shota Torigoe
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Aoba-cho, Higashimurayama, Tokyo, Japan
- Research Center for Biosafety, Laboratory Animal and Pathogen Bank, National Institute of Infectious Diseases, Toyama, Shinjuku, Tokyo, Japan
| | - Yusuke Tsujimura
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Aoba-cho, Higashimurayama, Tokyo, Japan
| | - Masamitsu N Asaka
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Aoba-cho, Higashimurayama, Tokyo, Japan
| | - Kayo Okumura
- Research Center for Biosafety, Laboratory Animal and Pathogen Bank, National Institute of Infectious Diseases, Toyama, Shinjuku, Tokyo, Japan
| | - Manabu Ato
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Aoba-cho, Higashimurayama, Tokyo, Japan
| |
Collapse
|
3
|
Gaspar EB, dos Santos LR, do Egito AA, dos Santos MG, Mantovani C, Rieger JDSG, Abrantes GADS, Suniga PAP, Favacho JDM, Pinto IB, Nassar AFDC, dos Santos FL, de Araújo FR. Assessment of the Virulence of the Burkholderia mallei Strain BAC 86/19 in BALB/c Mice. Microorganisms 2023; 11:2597. [PMID: 37894255 PMCID: PMC10609534 DOI: 10.3390/microorganisms11102597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Burkholderia mallei is an aerobic, Gram-negative, non-motile bacillus. As an obligate mammalian pathogen, it primarily affects solipeds. Although rarely transmitted to humans, the disease it causes, glanders, is classified as a zoonosis. The bacterium was officially eradicated in Brazil in 1969; however, it reemerged after three decades. This study aims to assess the virulence of a specific B. mallei strain, isolated in Brazil, in BALB/c mice through intranasal infection. The strain, B. mallei BAC 86/19, was obtained from the tracheal secretion of a young mare displaying positive serology but no clinical signs of glanders. Post-mortem examinations revealed macroscopic lesions consistent with the disease, however. In mice, the LD50 was determined to be approximately 1.59 × 105 colony-forming units (CFU)/animal. Mice exposed to either 0.1 × LD50 or 1 × LD50 displayed transient weight loss, which resolved after three or five days, respectively. B. mallei persisted within the liver and lung for five days post-infection and in the spleen for seven days. These findings underscore the detectable virulence of the Brazilian B. mallei BAC 86/19 strain in mice, which are relatively resilient hosts. This research points to the importance of the continued investigation of the virulence mechanisms and potential countermeasures associated with B. mallei infections, including their Brazilian isolates.
Collapse
Affiliation(s)
- Emanuelle Baldo Gaspar
- Embrapa South Livestock, BR-153, Km 632, 9 Vila Industrial, Rural Area, Mailbox 242, Bagé 96401-970, RS, Brazil
| | - Lenita Ramires dos Santos
- Embrapa Beef Cattle, Rádio Maia Ave., 830, Campo Grande 79106-550, MS, Brazil; (L.R.d.S.); (A.A.d.E.); (M.G.d.S.); (F.R.d.A.)
| | - Andréa Alves do Egito
- Embrapa Beef Cattle, Rádio Maia Ave., 830, Campo Grande 79106-550, MS, Brazil; (L.R.d.S.); (A.A.d.E.); (M.G.d.S.); (F.R.d.A.)
| | - Maria Goretti dos Santos
- Embrapa Beef Cattle, Rádio Maia Ave., 830, Campo Grande 79106-550, MS, Brazil; (L.R.d.S.); (A.A.d.E.); (M.G.d.S.); (F.R.d.A.)
| | - Cynthia Mantovani
- Embrapa Beef Cattle/Ministry of Agriculture, Livestock and Food Supply Scholarship, Embrapa Beef Cattle, Rádio Maia Ave., 830, Campo Grande 79106-550, MS, Brazil; (C.M.); (J.d.S.G.R.); (G.A.d.S.A.); (I.B.P.)
| | - Juliana da Silva Gomes Rieger
- Embrapa Beef Cattle/Ministry of Agriculture, Livestock and Food Supply Scholarship, Embrapa Beef Cattle, Rádio Maia Ave., 830, Campo Grande 79106-550, MS, Brazil; (C.M.); (J.d.S.G.R.); (G.A.d.S.A.); (I.B.P.)
| | - Guilherme Augusto de Sousa Abrantes
- Embrapa Beef Cattle/Ministry of Agriculture, Livestock and Food Supply Scholarship, Embrapa Beef Cattle, Rádio Maia Ave., 830, Campo Grande 79106-550, MS, Brazil; (C.M.); (J.d.S.G.R.); (G.A.d.S.A.); (I.B.P.)
| | - Paula Adas Pereira Suniga
- MAI/DAI Scholarship, Federal University of Mato Grosso do Sul, Cidade Universitária, Costa e Silva Ave., Campo Grande 79070-900, MS, Brazil;
- Postgraduate Program in Animal Science, Faculty of Veterinary Medicine and Animal Science-FAMEZ/UFMS, Federal University of Mato Grosso do Sul, Senador Filinto Muller Ave., 2443, Campo Grande 79074-460, MS, Brazil
| | | | - Ingrid Batista Pinto
- Embrapa Beef Cattle/Ministry of Agriculture, Livestock and Food Supply Scholarship, Embrapa Beef Cattle, Rádio Maia Ave., 830, Campo Grande 79106-550, MS, Brazil; (C.M.); (J.d.S.G.R.); (G.A.d.S.A.); (I.B.P.)
| | | | - Fernando Leandro dos Santos
- UFPE Department of Veterinary Medicine, Federal Rural University of Pernambuco, Recife 52171-900, PE, Brazil;
| | - Flábio Ribeiro de Araújo
- Embrapa Beef Cattle, Rádio Maia Ave., 830, Campo Grande 79106-550, MS, Brazil; (L.R.d.S.); (A.A.d.E.); (M.G.d.S.); (F.R.d.A.)
| |
Collapse
|
4
|
Gregor C. Generation of bright autobioluminescent bacteria by chromosomal integration of the improved lux operon ilux2. Sci Rep 2022; 12:19039. [PMID: 36351939 PMCID: PMC9646698 DOI: 10.1038/s41598-022-22068-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/10/2022] [Indexed: 11/10/2022] Open
Abstract
The bacterial bioluminescence system enables the generation of light by living cells without the requirement of an external luciferin. Due to the relatively low light emission, many applications of bioluminescence imaging would benefit from an increase in brightness of this system. In this report, a new approach of mutagenesis and screening of the involved proteins is described that is based on the identification of mutants with improved properties under rate-limiting reaction conditions. Multiple rounds of screening in Escherichia coli resulted in the operon ilux2 that contains 26 new mutations in the fatty acid reductase complex which provides the aldehyde substrate for the bioluminescence reaction. Chromosomal integration of ilux2 yielded an autonomously bioluminescent E. coli strain with sixfold increased brightness compared to the previously described ilux operon. The ilux2 strain produces sufficient signal for the robust detection of individual cells and enables highly sensitive long-term imaging of bacterial propagation without a selection marker.
Collapse
Affiliation(s)
- Carola Gregor
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
- Department of Optical Nanoscopy, Institut für Nanophotonik Göttingen e.V., Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
5
|
Longitudinal monitoring of individual infection progression in Drosophila melanogaster. iScience 2022; 25:105378. [DOI: 10.1016/j.isci.2022.105378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/26/2022] [Accepted: 10/13/2022] [Indexed: 11/05/2022] Open
|
6
|
Korniakova V, Devinck A, Groleau MC, Déziel E, Perreault J. Fluoride-Controlled Riboswitch-Based Dampening of Gene Expression for Cloning Potent Promoters. Front Genet 2022; 12:591543. [PMID: 35126444 PMCID: PMC8814607 DOI: 10.3389/fgene.2021.591543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/15/2021] [Indexed: 11/29/2022] Open
Abstract
Bioreporter systems based on detectable enzyme activity, such as that of beta-galactosidase or luciferase, are key in novel bacterial promoter discovery and study. While these systems permit quantification of gene expression, their use is limited by the toxicity of the expressed reporter enzymes in a given host. Indeed, the most potent promoters may be overlooked if their activity causes a lethal overproduction of the reporter genes when screening for transcriptional activity of potential promoter sequences with the luxCDABE cassette. To overcome this limitation, a variation of the mini-CTX-lux plasmid has been designed which allows reduction of promoter activity via the addition of an adjacent fluoride riboswitch. The riboswitch adds a layer of regulation between the promoter and the reporter gene, allowing cloning of stronger promoters by weakening expression, while giving the potential to induce with fluoride to provide a good signal for weaker promoters, thus circumventing limitations associated with reporter toxicity. We noticed the riboswitch potential portability issues between species, suggesting caution when using riboswitches non-native to the species where it is being used. This study introduces a new molecular biology tool which will allow for the identification of previously unverifiable or uncharacterized potent promoters and also provides a cloning vector for translational fusion with luciferase in a plasmid compatible with many species such as from the genera Burkholderia and Pseudomonas.
Collapse
|
7
|
Park SB, Jung SH, Jin H, Kim SJ, Ryu Y, Lee KJ, Kim B, Shin HJ, Won KJ. Bioluminescence Imaging of Matrix Metalloproteinases-2 and -9 Activities in Ethanol-injured Cornea of Mice. In Vivo 2021; 35:1521-1528. [PMID: 33910830 DOI: 10.21873/invivo.12405] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND/AIM This study aimed to investigate the usefulness of in vivo bioluminescence imaging (BLI) to examine the role of matrix metalloproteinases (MMP)-2 and MMP-9 activation in the development and healing of ethanol-induced damage in the cornea of mice. MATERIALS AND METHODS Mouse corneal injury was induced by topical treatment with 20% ethanol. BLI was obtained from the ocular region of mice intravenously injected with an active-MMP-2/9 probe. In vivo results were validated in primary corneal epithelial cells. RESULTS BLI indicated that treatment of the eye with 20% ethanol elevated MMP-2/9 activity, which was inhibited by the application of eye drops (hyaluronic acid and serum). Treatment of corneal epithelial cells with 20% ethanol-increased the activities of MMP-2 and MMP-9, which were also inhibited by eye drops. CONCLUSION BLI can be applied in vivo in mice with corneal injury to examine the activity of MMPs and clarify the efficacy of eye drops.
Collapse
Affiliation(s)
- Seung-Bo Park
- Department of Physiology, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Seung Hyo Jung
- Department of Physiology, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Hengzhe Jin
- Department of Physiology, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Su Jung Kim
- Department of Physiology, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Yunkyoung Ryu
- Department of Physiology, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Kyung-Jin Lee
- Department of Physiology, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Bokyung Kim
- Department of Physiology, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Hyun Jin Shin
- Department of Ophthalmology, Konkuk Medical Center, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Kyung-Jong Won
- Department of Physiology, Konkuk University School of Medicine, Seoul, Republic of Korea;
| |
Collapse
|
8
|
Yang Z, Cui Q, Zhang M, Li Z, Wang M, Xu H. A lux-based Staphylococcus aureus bioluminescence screening assay for the detection/identification of antibiotics and prediction of antibiotic mechanisms. J Antibiot (Tokyo) 2020; 73:828-836. [PMID: 32678336 DOI: 10.1038/s41429-020-0349-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 06/09/2020] [Accepted: 06/30/2020] [Indexed: 11/09/2022]
Abstract
The need for the discovery of new antibiotics and solving the antibiotic resistance problem requires rapid detection of antibiotics, identification of known antibiotics, and prediction of antibiotic mechanisms. The bacterial lux genes encode proteins that convert chemical energy into photonic energy and lead to bioluminescence. Exploiting this phenomenon, we constructed a lux-based bioluminescence system in Staphylococcus aureus by expressing lux genes under the control of stress-inducible chaperon promoters. When experiencing antibiotic stress, these constructed reporter strains showed clear bioluminescence response. Therefore, this bioluminescence screening system can be used for the detection of antibiotics in unknown chemical mixtures. Further analysis of bioluminescence response patterns showed that: (1) these bioluminescence response patterns are highly antibiotic specific and therefore can be used for rapid and cheap identification of antibiotics; and that (2) antibiotics having the same mechanism of action have similar bioluminescence patterns and therefore these patterns can be used for the prediction of mechanism for an unknown antibiotic with good sensitivity and specificity. With this bioluminescence screening assay, the discovery and analysis of new antibiotics can be promoted, which benefits in solving the antibiotic resistance problem.
Collapse
Affiliation(s)
- Zhongjun Yang
- State Key Laboratory of Microbial Technology, Qilu Hospital, Shandong University, Qingdao, 266237, Shandong, China
| | - Qingyu Cui
- State Key Laboratory of Microbial Technology, Qilu Hospital, Shandong University, Qingdao, 266237, Shandong, China
| | - Mengge Zhang
- State Key Laboratory of Microbial Technology, Qilu Hospital, Shandong University, Qingdao, 266237, Shandong, China
| | - Zhiqiang Li
- Center for Optics Research and Engineering, Shandong University, Qingdao, 266237, Shandong, China
| | - Mingyu Wang
- State Key Laboratory of Microbial Technology, Qilu Hospital, Shandong University, Qingdao, 266237, Shandong, China.
| | - Hai Xu
- State Key Laboratory of Microbial Technology, Qilu Hospital, Shandong University, Qingdao, 266237, Shandong, China.
| |
Collapse
|
9
|
Norris MH, Khan MSR, Chirakul S, Schweizer HP, Tuanyok A. Outer Membrane Vesicle Vaccines from Biosafe Surrogates Prevent Acute Lethal Glanders in Mice. Vaccines (Basel) 2018; 6:E5. [PMID: 29320408 PMCID: PMC5874646 DOI: 10.3390/vaccines6010005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 01/04/2018] [Accepted: 01/06/2018] [Indexed: 01/13/2023] Open
Abstract
Burkholderia mallei is a host-adapted Gram-negative mammalian pathogen that causes the severe disease glanders. Glanders can manifest as a rapid acute progression or a chronic debilitating syndrome primarily affecting solipeds and humans in close association with infected animals. In USA, B. mallei is classified as one of the most important bacterial biothreat agents. Presently, there is no licensed glanders vaccine available for humans or animals. In this work, outer membrane vesicles (OMVs) were isolated from three attenuated biosafe bacterial strains, Burkholderia pseudomallei Bp82, B. thailandensis E555, and B. thailandensis TxDOH and used to vaccinate mice. B. thailandensis OMVs induced significantly higher antibody responses that were investigated. B. mallei specific serum antibody responses were of higher magnitude in mice vaccinated with B. thailandensis OMVs compared to levels in mice vaccinated with B. pseudomallei OMVs. OMVs derived from biosafe strains protected mice from acute lethal glanders with vesicles from the two B. thailandensis strains affording significant protection (>90%) up to 35 days post-infection with some up to 60 days. Organ loads from 35-day survivors indicated bacteria colonization of the lungs, liver, and spleen while those from 60 days had high CFUs in the spleens. The highest antibody producing vaccine (B. thailandensis E555 OMVs) also protected C57BL/6 mice from acute inhalational glanders with evidence of full protection.
Collapse
Affiliation(s)
- Michael H Norris
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine; University of Florida, Gainesville, FL 32608, USA.
- Emerging Pathogens Institute, University of Florida, Gainesville, FL 32610, USA.
| | - Mohammad S R Khan
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine; University of Florida, Gainesville, FL 32608, USA.
- Emerging Pathogens Institute, University of Florida, Gainesville, FL 32610, USA.
| | - Sunisa Chirakul
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine; University of Florida, Gainesville, FL 32608, USA.
- Emerging Pathogens Institute, University of Florida, Gainesville, FL 32610, USA.
| | - Herbert P Schweizer
- Emerging Pathogens Institute, University of Florida, Gainesville, FL 32610, USA.
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32603, USA.
| | - Apichai Tuanyok
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine; University of Florida, Gainesville, FL 32608, USA.
- Emerging Pathogens Institute, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
10
|
Zimmerman SM, Long ME, Dyke JS, Jelesijevic TP, Michel F, Lafontaine ER, Hogan RJ. Use of Immunohistochemistry to Demonstrate In Vivo Expression of the Burkholderia mallei Virulence Factor BpaB During Experimental Glanders. Vet Pathol 2017; 55:258-267. [PMID: 29145795 DOI: 10.1177/0300985817736113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Burkholderia mallei causes the highly contagious and debilitating zoonosis glanders, which infects via inhalation or percutaneous inoculation and often culminates in life-threatening pneumonia and sepsis. In humans, glanders is difficult to diagnose and requires prolonged antibiotic therapy with low success rates. No vaccine exists to protect against B. mallei, and there is concern regarding its use as a bioweapon. The authors previously identified the protein BpaB as a potential target for devising therapies due to its role in adherence to host cells and the formation of biofilms in vitro and its contribution to pathogenicity in a mouse model of glanders. In the present study, the authors developed an immunostaining approach to probe tissues of experimentally infected animals and demonstrated that BpaB is produced exclusively in vivo by wild-type B. mallei in target organs from mice and marmosets. They detected the expression of BpaB by B. mallei both extracellularly and within macrophages, neutrophils, and epithelial cells in respiratory tissues (7/10 marmoset; 2/2 mouse). The authors also noted the intracellular expression of BpaB by B. mallei in macrophages in the regional lymph nodes of mice (2/2 tissues) and MALT of marmosets (4/5 tissues). It is interesting that B. mallei bacteria infecting distal organs did not express BpaB (2/2 mice; 3/3 marmosets), suggesting that the protein is not necessary for bacterial fitness in these anatomic locations. These findings underscore the value of BpaB as a target for developing medical countermeasures and provide insight into its role in pathogenesis.
Collapse
Affiliation(s)
- Shawn M Zimmerman
- 1 Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens, GA, USA
| | - Mackenzie E Long
- 2 Veterinary Teaching Hospital, University of Georgia College of Veterinary Medicine, Athens, GA, USA
| | - Jeremy S Dyke
- 1 Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens, GA, USA
| | - Tomislav P Jelesijevic
- 1 Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens, GA, USA
| | - Frank Michel
- 3 Department of Veterinary Biosciences and Diagnostic Imaging, University of Georgia College of Veterinary Medicine, Athens, GA, USA
| | - Eric R Lafontaine
- 1 Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens, GA, USA
| | - Robert J Hogan
- 1 Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens, GA, USA.,3 Department of Veterinary Biosciences and Diagnostic Imaging, University of Georgia College of Veterinary Medicine, Athens, GA, USA
| |
Collapse
|
11
|
Morris JL, Fane A, Sarovich DS, Price EP, Rush CM, Govan BL, Parker E, Mayo M, Currie BJ, Ketheesan N. Increased Neurotropic Threat from Burkholderia pseudomallei Strains with a B. mallei-like Variation in the bimA Motility Gene, Australia. Emerg Infect Dis 2017; 23. [PMID: 28418830 PMCID: PMC5403032 DOI: 10.3201/eid2305.151417] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
These strains have heightened pathogenic potential for rapid dissemination to multiple tissues, including the central nervous system. Neurologic melioidosis is a serious, potentially fatal form of Burkholderia pseudomallei infection. Recently, we reported that a subset of clinical isolates of B. pseudomallei from Australia have heightened virulence and potential for dissemination to the central nervous system. In this study, we demonstrate that this subset has a B. mallei–like sequence variation of the actin-based motility gene, bimA. Compared with B. pseudomallei isolates having typical bimA alleles, isolates that contain the B. mallei–like variation demonstrate increased persistence in phagocytic cells and increased virulence with rapid systemic dissemination and replication within multiple tissues, including the brain and spinal cord, in an experimental model. These findings highlight the implications of bimA variation on disease progression of B. pseudomallei infection and have considerable clinical and public health implications with respect to the degree of neurotropic threat posed to human health.
Collapse
|
12
|
Mezzanotte L, van 't Root M, Karatas H, Goun EA, Löwik CWGM. In Vivo Molecular Bioluminescence Imaging: New Tools and Applications. Trends Biotechnol 2017; 35:640-652. [PMID: 28501458 DOI: 10.1016/j.tibtech.2017.03.012] [Citation(s) in RCA: 199] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/07/2017] [Accepted: 03/27/2017] [Indexed: 12/19/2022]
Abstract
in vivo bioluminescence imaging (BLi) is an optical molecular imaging technique used to visualize molecular and cellular processes in health and diseases and to follow the fate of cells with high sensitivity using luciferase-based gene reporters. The high sensitivity of this technique arises from efficient photon production, followed by the reaction between luciferase enzymes and luciferin substrates. Novel discoveries and developments of luciferase reporters, substrates, and gene-editing techniques, and emerging fields of applications, promise a new era of deeper and more sensitive molecular imaging.
Collapse
Affiliation(s)
- Laura Mezzanotte
- Optical Molecular imaging, Department of Radiology, Erasmus MC, Rotterdam, The Netherlands.
| | - Moniek van 't Root
- Optical Molecular imaging, Department of Radiology, Erasmus MC, Rotterdam, The Netherlands
| | - Hacer Karatas
- Laboratory of Bioorganic Chemistry and Molecular Imaging, Institute of Chemical Sciences and Engineering (ISIC), Swiss Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Elena A Goun
- Laboratory of Bioorganic Chemistry and Molecular Imaging, Institute of Chemical Sciences and Engineering (ISIC), Swiss Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Clemens W G M Löwik
- Optical Molecular imaging, Department of Radiology, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
13
|
A Bioluminescent Francisella tularensis SCHU S4 Strain Enables Noninvasive Tracking of Bacterial Dissemination and the Evaluation of Antibiotics in an Inhalational Mouse Model of Tularemia. Antimicrob Agents Chemother 2016; 60:7206-7215. [PMID: 27671061 DOI: 10.1128/aac.01586-16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 09/08/2016] [Indexed: 11/20/2022] Open
Abstract
Bioluminescence imaging (BLI) enables real-time, noninvasive tracking of infection in vivo and longitudinal infection studies. In this study, a bioluminescent Francisella tularensis strain, SCHU S4-lux, was used to develop an inhalational infection model in BALB/c mice. Mice were infected intranasally, and the progression of infection was monitored in real time using BLI. A bioluminescent signal was detectable from 3 days postinfection (3 dpi), initially in the spleen and then in the liver and lymph nodes, before finally becoming systemic. The level of bioluminescent signal correlated with bacterial numbers in vivo, enabling noninvasive quantification of bacterial burdens in tissues. Treatment with levofloxacin (commencing at 4 dpi) significantly reduced the BLI signal. Furthermore, BLI was able to distinguish noninvasively between different levofloxacin treatment regimens and to identify sites of relapse following treatment cessation. These data demonstrate that BLI and SCHU S4-lux are suitable for the study of F. tularensis pathogenesis and the evaluation of therapeutics for tularemia.
Collapse
|
14
|
Skare JT, Shaw DK, Trzeciakowski JP, Hyde JA. In Vivo Imaging Demonstrates That Borrelia burgdorferi ospC Is Uniquely Expressed Temporally and Spatially throughout Experimental Infection. PLoS One 2016; 11:e0162501. [PMID: 27611840 PMCID: PMC5017786 DOI: 10.1371/journal.pone.0162501] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 08/23/2016] [Indexed: 11/18/2022] Open
Abstract
Borrelia burgdorferi is a spirochetal bacterium transmitted by the Ixodes tick that causes Lyme disease in humans due to its ability to evade the host immune response and disseminate to multiple immunoprotective tissues. The pathogen undergoes dynamic genetic alterations important for adaptation from the tick vector to the mammalian host, but little is known regarding the changes at the transcriptional level within the distal tissues they colonize. In this study, B. burgdorferi infection and gene expression of the essential virulence determinant ospC was quantitatively monitored in a spatial and temporal manner utilizing reporter bioluminescent borrelial strains with in vivo and ex vivo imaging. Although expressed from a shuttle vector, the PospC-luc construct exhibited a similar expression pattern relative to native ospC. Bacterial burden in skin, inguinal lymph node, heart, bladder and tibiotarsal joint varied between tissues and fluctuated over the course of infection possibly in response to unique cues of each microenvironment. Expression of ospC, when normalized for changes in bacterial load, presented unique profiles in murine tissues at different time points. The inguinal lymph node was infected with a significant B. burgdorferi burden, but showed minimal ospC expression. B. burgdorferi infected skin and heart induced expression of ospC early during infection while the bladder and tibiotarsal joint continued to display PospC driven luminescence throughout the 21 day time course. Localized skin borrelial burden increased dramatically in the first 96 hours following inoculation, which was not paralleled with an increase in ospC expression, despite the requirement of ospC for dermal colonization. Quantitation of bioluminescence representing ospC expression in individual tissues was validated by qRT-PCR of the native ospC transcript. Taken together, the temporal regulation of ospC expression in distal tissues suggests a role for this virulence determinant beyond early infection.
Collapse
Affiliation(s)
- Jonathan T. Skare
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science Center, Bryan/College Station, Texas, United States of America
| | - Dana K. Shaw
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science Center, Bryan/College Station, Texas, United States of America
| | - Jerome P. Trzeciakowski
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan/College Station, Texas, United States of America
| | - Jenny A. Hyde
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science Center, Bryan/College Station, Texas, United States of America
- * E-mail:
| |
Collapse
|
15
|
Burkholderia mallei CLH001 Attenuated Vaccine Strain Is Immunogenic and Protects against Acute Respiratory Glanders. Infect Immun 2016; 84:2345-54. [PMID: 27271739 DOI: 10.1128/iai.00328-16] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 05/26/2016] [Indexed: 11/20/2022] Open
Abstract
Burkholderia mallei is the causative agent of glanders, an incapacitating disease with high mortality rates in respiratory cases. Its endemicity and ineffective treatment options emphasize its public health threat and highlight the need for a vaccine. Live attenuated vaccines are considered the most viable vaccine strategy for Burkholderia, but single-gene-deletion mutants have not provided complete protection. In this study, we constructed the select-agent-excluded B. mallei ΔtonB Δhcp1 (CLH001) vaccine strain and investigated its ability to protect against acute respiratory glanders. Here we show that CLH001 is attenuated, safe, and effective at protecting against lethal B. mallei challenge. Intranasal administration of CLH001 to BALB/c and NOD SCID gamma (NSG) mice resulted in complete survival without detectable colonization or abnormal organ histopathology. Additionally, BALB/c mice intranasally immunized with CLH001 in a prime/boost regimen were fully protected against lethal challenge with the B. mallei lux (CSM001) wild-type strain.
Collapse
|
16
|
Read HM, Mills G, Johnson S, Tsai P, Dalton J, Barquist L, Print CG, Patrick WM, Wiles S. The in vitro and in vivo effects of constitutive light expression on a bioluminescent strain of the mouse enteropathogen Citrobacter rodentium. PeerJ 2016; 4:e2130. [PMID: 27366640 PMCID: PMC4924136 DOI: 10.7717/peerj.2130] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 05/24/2016] [Indexed: 11/30/2022] Open
Abstract
Bioluminescent reporter genes, such as those from fireflies and bacteria, let researchers use light production as a non-invasive and non-destructive surrogate measure of microbial numbers in a wide variety of environments. As bioluminescence needs microbial metabolites, tagging microorganisms with luciferases means only live metabolically active cells are detected. Despite the wide use of bioluminescent reporter genes, very little is known about the impact of continuous (also called constitutive) light expression on tagged bacteria. We have previously made a bioluminescent strain of Citrobacter rodentium, a bacterium which infects laboratory mice in a similar way to how enteropathogenic Escherichia coli (EPEC) and enterohaemorrhagic E. coli (EHEC) infect humans. In this study, we compared the growth of the bioluminescent C. rodentium strain ICC180 with its non-bioluminescent parent (strain ICC169) in a wide variety of environments. To understand more about the metabolic burden of expressing light, we also compared the growth profiles of the two strains under approximately 2,000 different conditions. We found that constitutive light expression in ICC180 was near-neutral in almost every non-toxic environment tested. However, we also found that the non-bioluminescent parent strain has a competitive advantage over ICC180 during infection of adult mice, although this was not enough for ICC180 to be completely outcompeted. In conclusion, our data suggest that constitutive light expression is not metabolically costly to C. rodentium and supports the view that bioluminescent versions of microbes can be used as a substitute for their non-bioluminescent parents to study bacterial behaviour in a wide variety of environments.
Collapse
Affiliation(s)
- Hannah M Read
- Bioluminescent Superbugs Lab, University of Auckland, Auckland, New Zealand; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Grant Mills
- Bioluminescent Superbugs Lab, University of Auckland, Auckland, New Zealand; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Sarah Johnson
- Bioluminescent Superbugs Lab, University of Auckland, Auckland, New Zealand; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Peter Tsai
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; Bioinformatics Institute, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - James Dalton
- Bioluminescent Superbugs Lab, University of Auckland, Auckland, New Zealand; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand
| | - Lars Barquist
- Institute for Molecular Infection Biology, University of Würzburg , Würzburg , Germany
| | - Cristin G Print
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; Bioinformatics Institute, School of Biological Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand
| | - Wayne M Patrick
- Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand; Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Siouxsie Wiles
- Bioluminescent Superbugs Lab, University of Auckland, Auckland, New Zealand; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand
| |
Collapse
|
17
|
Bocan TM, Panchal RG, Bavari S. Applications of in vivo imaging in the evaluation of the pathophysiology of viral and bacterial infections and in development of countermeasures to BSL3/4 pathogens. Mol Imaging Biol 2015; 17:4-17. [PMID: 25008802 PMCID: PMC4544652 DOI: 10.1007/s11307-014-0759-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
While preclinical and clinical imaging have been applied to drug discovery/development and characterization of disease pathology, few examples exist where imaging has been used to evaluate infectious agents or countermeasures to biosafety level (BSL)3/4 threat agents. Viruses engineered with reporter constructs, i.e., enzymes and receptors, which are amenable to detection by positron emission tomography (PET), single photon emission tomography (SPECT), or magnetic resonance imaging (MRI) have been used to evaluate the biodistribution of viruses containing specific therapeutic or gene transfer payloads. Bioluminescence and nuclear approaches involving engineered reporters, direct labeling of bacteria with radiotracers, or tracking bacteria through their constitutively expressed thymidine kinase have been utilized to characterize viral and bacterial pathogens post-infection. Most PET, SPECT, CT, or MRI approaches have focused on evaluating host responses to the pathogens such as inflammation, brain neurochemistry, and structural changes and on assessing the biodistribution of radiolabeled drugs. Imaging has the potential when applied preclinically to the development of countermeasures against BSL3/4 threat agents to address the following: (1) presence, biodistribution, and time course of infection in the presence or absence of drug; (2) binding of the therapeutic to the target; and (3) expression of a pharmacologic effect either related to drug mechanism, efficacy, or safety. Preclinical imaging could potentially provide real-time dynamic tools to characterize the pathogen and animal model and for developing countermeasures under the U.S. FDA Animal Rule provision with high confidence of success and clinical benefit.
Collapse
Affiliation(s)
- Thomas M Bocan
- Molecular and Translational Sciences, US Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Ft. Detrick, MD, 21702, USA,
| | | | | |
Collapse
|
18
|
Characterization of the Burkholderia mallei tonB Mutant and Its Potential as a Backbone Strain for Vaccine Development. PLoS Negl Trop Dis 2015; 9:e0003863. [PMID: 26114445 PMCID: PMC4482651 DOI: 10.1371/journal.pntd.0003863] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 06/01/2015] [Indexed: 01/24/2023] Open
Abstract
Background In this study, a Burkholderia mallei tonB mutant (TMM001) deficient in iron acquisition was constructed, characterized, and evaluated for its protective properties in acute inhalational infection models of murine glanders and melioidosis. Methodology/Principal Findings Compared to the wild-type, TMM001 exhibits slower growth kinetics, siderophore hyper-secretion and the inability to utilize heme-containing proteins as iron sources. A series of animal challenge studies showed an inverse correlation between the percentage of survival in BALB/c mice and iron-dependent TMM001 growth. Upon evaluation of TMM001 as a potential protective strain against infection, we found 100% survival following B. mallei CSM001 challenge of mice previously receiving 1.5 x 104 CFU of TMM001. At 21 days post-immunization, TMM001-treated animals showed significantly higher levels of B. mallei-specific IgG1, IgG2a and IgM when compared to PBS-treated controls. At 48 h post-challenge, PBS-treated controls exhibited higher levels of serum inflammatory cytokines and more severe pathological damage to target organs compared to animals receiving TMM001. In a cross-protection study of acute inhalational melioidosis with B. pseudomallei, TMM001-treated mice were significantly protected. While wild type was cleared in all B. mallei challenge studies, mice failed to clear TMM001. Conclusions/Significance Although further work is needed to prevent chronic infection by TMM001 while maintaining immunogenicity, our attenuated strain demonstrates great potential as a backbone strain for future vaccine development against both glanders and melioidosis. Burkholderia mallei and B. pseudomallei are the causative agents of glanders and melioidosis, respectively. In addition to the recent rise in cases of glanders and the endemicity of melioidosis worldwide, these pathogens have gained attention as potential bioweapons. Further, these pathogens have huge potential for aerosol delivery and often produce fatal infection amongst untreated individuals. Both pathogens are difficult to treat, and even with antibiotic intervention, patients relapse or get re-infected. A big challenge for vaccine development against these pathogens includes identification of broadly protective antigens and a better understanding of the correlates of protection from both acute and chronic infections. Our study is the first to demonstrate significant protection against a lethal challenge with both Burkholderia species. Because TMM001 persists in immunized mice, we propose that this attenuated organism is a promising backbone-based strain from which a legitimate vaccine candidate can be generated.
Collapse
|
19
|
Recognition of Immune Response for the Early Diagnosis and Treatment of Osteoarthritis. J Immunol Res 2015; 2015:192415. [PMID: 26064995 PMCID: PMC4433702 DOI: 10.1155/2015/192415] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 12/02/2014] [Indexed: 12/15/2022] Open
Abstract
Osteoarthritis is a common and debilitating joint disease that affects up to 30 million Americans, leading to significant disability, reduction in quality of life, and costing the United States tens of billions of dollars annually. Classically, osteoarthritis has been characterized as a degenerative, wear-and-tear disease, but recent research has identified it as an immunopathological disease on a spectrum between healthy condition and rheumatoid arthritis. A systematic literature review demonstrates that the disease pathogenesis is driven by an early innate immune response which progressively catalyzes degenerative changes that ultimately lead to an altered joint microenvironment. It is feasible to detect this infiltration of cells in the early, and presumably asymptomatic, phase of the disease through noninvasive imaging techniques. This screening can serve to aid clinicians in potentially identifying high-risk patients, hopefully leading to early effective management, vast improvements in quality of life, and significant reductions in disability, morbidity, and cost related to osteoarthritis. Although the diagnosis and treatment of osteoarthritis routinely utilize both invasive and non-invasive strategies, imaging techniques specific to inflammatory cells are not commonly employed for these purposes. This review discusses this paradigm and aims to shift the focus of future osteoarthritis-related research towards early diagnosis of the disease process.
Collapse
|
20
|
Bruckbauer ST, Kvitko BH, Karkhoff-Schweizer RR, Schweizer HP. Tn5/7-lux: a versatile tool for the identification and capture of promoters in gram-negative bacteria. BMC Microbiol 2015; 15:17. [PMID: 25648327 PMCID: PMC4328036 DOI: 10.1186/s12866-015-0354-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 01/19/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The combination of imaging technologies and luciferase-based bioluminescent bacterial reporter strains provide a sensitive and simple non-invasive detection method (photonic bioimaging) for the study of diverse biological processes, as well as efficacy of therapeutic interventions, in live animal models of disease. The engineering of bioluminescent bacteria required for photonic bioimaging is frequently hampered by lack of promoters suitable for strong, yet stable luciferase gene expression. RESULTS We devised a novel method for identification of constitutive native promoters in Gram-negative bacteria. The method is based on a Tn5/7 transposon that exploits the unique features of Tn5 (random transposition) and Tn7 (site-specific transposition). The transposons are designed such that Tn5 transposition will allow insertion of a promoter-less bacterial luxCDABE operon downstream of a bacterial gene promoter. Cloning of DNA fragments from luminescent isolates results in a plasmid that replicates in pir (+) hosts. Sequencing of the lux-chromosomal DNA junctions on the plasmid reveals transposon insertion sites within genes or operons. The plasmid is also a mini-Tn7-lux delivery vector that can be used to introduce the promoter-lux operon fusion into other derivatives of the bacterium of interest in an isogenic fashion. Alternatively, promoter-containing sequences can be PCR-amplified from plasmid or chromosomal DNA and cloned into a series of accompanying mini-Tn7-lux vectors. The mini-Tn5/7-lux and mini-Tn7-lux vectors are equipped with diverse selection markers and thus applicable in numerous Gram-negative bacteria. Various mini-Tn5/7-lux vectors were successfully tested for transposition and promoter identification by imaging in Acinetobacter baumannii, Escherichia coli, and Burkholderia pseudomallei. Strong promoters were captured for lux expression in E. coli and A. baumannii. Some mini-Tn7-lux vectors are also equipped with attB sites for swapping of the lux operon with other reporter genes using Gateway technology. CONCLUSIONS Although mini-Tn5-lux and mini-Tn7-lux elements have previously been developed and used for bacterial promoter identification and chromosomal insertion of promoter-lux gene fusions, respectively, the newly developed mini-Tn5/7-lux and accompanying accessory plasmids streamline and accelerate the promoter discovery and bioluminescent strain engineering processes. Availability of vectors with diverse selection markers greatly extend the host-range of promoter probe and lux gene fusion vectors.
Collapse
Affiliation(s)
- Steven T Bruckbauer
- Department of Microbiology, Immunology and Pathology, and Rocky Mountain Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, Colorado State University, Fort Collins, 80523, CO, USA. .,Present Address: Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, 53706, WI, USA.
| | - Brian H Kvitko
- Department of Microbiology, Immunology and Pathology, and Rocky Mountain Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, Colorado State University, Fort Collins, 80523, CO, USA. .,Present Address: MSU-DOE Plant Research Laboratory, Michigan State University, East Lansing, 48824, MI, USA.
| | - RoxAnn R Karkhoff-Schweizer
- Department of Microbiology, Immunology and Pathology, and Rocky Mountain Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, Colorado State University, Fort Collins, 80523, CO, USA.
| | - Herbert P Schweizer
- Department of Microbiology, Immunology and Pathology, and Rocky Mountain Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, Colorado State University, Fort Collins, 80523, CO, USA. .,Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Emerging Pathogens Institute, PO Box 100266, Gainesville, 32610-0266, FL, USA.
| |
Collapse
|
21
|
Liu Y, Yang S, Xiao J, Yu L, Chen L, Zou J, Wang K, Tan S, Yu Z, Zeng Q. M13 phage peptide ZL4 exerts its targeted binding effect on schistosoma japonicum via alkaline phosphatase. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:1247-1258. [PMID: 25973009 PMCID: PMC4396255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 01/28/2015] [Indexed: 06/04/2023]
Abstract
The present study was to determine the targeting effect of M13 phage peptide ZL4 (MppZL4) on Schistosoma japonicum (S.j). Mice infected with S.j were injected with MppZL4. Real-time PCR was used to detect the distribution and metabolism of MppZL4 in the livers and lungs of mice. In vivo refusion test was performed to detect the targeting of MppZL4. Western blotting was employed to determine the expression of MppZL4. Live imaging was used to detect the distribution of oligopeptide MppZL4. Immunohistochemistry was employed to determine MppZL4 location on adult S.j body surface. Gomori method was employed to detect the influence of oligopeptide MppZL4 on alkaline phosphatase activity. The distribution and metabolism of MppZL4 and M13KE are not significantly different from each other at each time point. The abundance of MppZL4 is changed as S.j migrates in mice. The targeted binding effect of MppZL4 varies at different stages. ZL4 oligopeptide targets S.j in mice. The specific binding sites of MppZL4 on S.j body are mainly located in syncytial cells. The binding sites of MppZL4 on S.j body surface might be ALP or ALP-related proteins. MppZL4 had targeted binding effect on S.j with its binding site being associated with proteins related to S.j alkaline phosphatase. S.j tegument had a specifically binding site with exogenous peptides, offering new means to explore the interactions between hosts and parasites. Additionally, MppZL4 can possibly be used as targeting molecules in worm-resistant drugs or as tracing molecules in imaging diagnosis technologies.
Collapse
Affiliation(s)
- Yan Liu
- Department of Parasitology, School of Medicine, University of South ChinaHengyang, P.R. China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug StudyHengyang, P.R. China
| | - Shenghui Yang
- Department of Preventive Medicine and Pathogenic Biology, School of Medicine, Hunan University of Chinese MedicineChangsha, P.R. China
| | - Jianhua Xiao
- Department of Parasitology, School of Medicine, University of South ChinaHengyang, P.R. China
| | - Liang Yu
- Department of Parasitology, School of Medicine, University of South ChinaHengyang, P.R. China
| | - Li Chen
- Department of Medicine, Perelman School of Medicine, University of PennsylvaniaPhiladelphia, USA
| | - Ju Zou
- Department of Parasitology, School of Medicine, University of South ChinaHengyang, P.R. China
| | - Kegeng Wang
- Department of Parasitology, School of Medicine, University of South ChinaHengyang, P.R. China
| | - Sijie Tan
- Department of Histology and Embryology, School of Medicine, University of South ChinaHengyang, P.R. China
| | - Zhengyang Yu
- Surgical Oncology, First Affiliated Hospital, University of South ChinaHengyang, P.R. China
| | - Qingren Zeng
- Centre of Cell and Molecular Biology Experiment, Xiangya School of Medicine, Central South UniversityChangsha, P.R. China
| |
Collapse
|
22
|
Su S, Bangar H, Saldanha R, Pemberton A, Aronow B, Dean GE, Lamkin TJ, Hassett DJ. Construction and characterization of stable, constitutively expressed, chromosomal green and red fluorescent transcriptional fusions in the select agents, Bacillus anthracis, Yersinia pestis, Burkholderia mallei, and Burkholderia pseudomallei. Microbiologyopen 2014; 3:610-29. [PMID: 25044501 PMCID: PMC4234255 DOI: 10.1002/mbo3.192] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 05/23/2014] [Accepted: 05/29/2014] [Indexed: 11/09/2022] Open
Abstract
Here, we constructed stable, chromosomal, constitutively expressed, green and red fluorescent protein (GFP and RFP) as reporters in the select agents, Bacillus anthracis, Yersinia pestis, Burkholderia mallei, and Burkholderia pseudomallei. Using bioinformatic approaches and other experimental analyses, we identified P0253 and P1 as potent promoters that drive the optimal expression of fluorescent reporters in single copy in B. anthracis and Burkholderia spp. as well as their surrogate strains, respectively. In comparison, Y. pestis and its surrogate strain need two chromosomal copies of cysZK promoter (P2cysZK) for optimal fluorescence. The P0253-, P2cysZK-, and P1-driven GFP and RFP fusions were first cloned into the vectors pRP1028, pUC18R6KT-mini-Tn7T-Km, pmini-Tn7-gat, or their derivatives. The resultant constructs were delivered into the respective surrogates and subsequently into the select agent strains. The chromosomal GFP- and RFP-tagged strains exhibited bright fluorescence at an exposure time of less than 200 msec and displayed the same virulence traits as their wild-type parental strains. The utility of the tagged strains was proven by the macrophage infection assays and lactate dehydrogenase release analysis. Such strains will be extremely useful in high-throughput screens for novel compounds that could either kill these organisms, or interfere with critical virulence processes in these important bioweapon agents and during infection of alveolar macrophages.
Collapse
Affiliation(s)
- Shengchang Su
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of MedicineCincinnati, Ohio, 45267
| | - Hansraj Bangar
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of MedicineCincinnati, Ohio, 45267
| | | | | | - Bruce Aronow
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical CenterCincinnati, Ohio, 45229-3039
| | - Gary E Dean
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of MedicineCincinnati, Ohio, 45267
| | - Thomas J Lamkin
- Air Force Research Laboratory, 711th HPW/RHXBC, Molecular Signatures SectionWright-Patterson AFB, Ohio, 45433-7913
| | - Daniel J Hassett
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of MedicineCincinnati, Ohio, 45267
| |
Collapse
|
23
|
The impact of "omic" and imaging technologies on assessing the host immune response to biodefence agents. J Immunol Res 2014; 2014:237043. [PMID: 25333059 PMCID: PMC4182007 DOI: 10.1155/2014/237043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 07/23/2014] [Accepted: 08/05/2014] [Indexed: 01/08/2023] Open
Abstract
Understanding the interactions between host and pathogen is important for the development and assessment of medical countermeasures to infectious agents, including potential biodefence pathogens such as Bacillus anthracis, Ebola virus, and Francisella tularensis. This review focuses on technological advances which allow this interaction to be studied in much greater detail. Namely, the use of “omic” technologies (next generation sequencing, DNA, and protein microarrays) for dissecting the underlying host response to infection at the molecular level; optical imaging techniques (flow cytometry and fluorescence microscopy) for assessing cellular responses to infection; and biophotonic imaging for visualising the infectious disease process. All of these technologies hold great promise for important breakthroughs in the rational development of vaccines and therapeutics for biodefence agents.
Collapse
|
24
|
Yoshioka K, Ishii K, Kuramoto T, Nagai S, Funao H, Ishihama H, Shiono Y, Sasaki A, Aizawa M, Okada Y, Koyasu S, Toyama Y, Matsumoto M. A novel mouse model of soft-tissue infection using bioluminescence imaging allows noninvasive, real-time monitoring of bacterial growth. PLoS One 2014; 9:e106367. [PMID: 25184249 PMCID: PMC4153648 DOI: 10.1371/journal.pone.0106367] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 07/28/2014] [Indexed: 12/18/2022] Open
Abstract
Musculoskeletal infections, including surgical-site and implant-associated infections, often cause progressive inflammation and destroy areas of the soft tissue. Treating infections, especially those caused by multi-antibiotic resistant bacteria such as methicillin-resistant Staphylococcus aureus (MRSA) remains a challenge. Although there are a few animal models that enable the quantitative evaluation of infection in soft tissues, these models are not always reproducible or sustainable. Here, we successfully established a real-time, in vivo, quantitative mouse model of soft-tissue infection in the superficial gluteus muscle (SGM) using bioluminescence imaging. A bioluminescent strain of MRSA was inoculated into the SGM of BALB/c adult male mice, followed by sequential measurement of bacterial photon intensity and serological and histological analyses of the mice. The mean photon intensity in the mice peaked immediately after inoculation and remained stable until day 28. The serum levels of interleukin-6, interleukin-1 and C-reactive protein at 12 hours after inoculation were significantly higher than those prior to inoculation, and the C-reactive protein remained significantly elevated until day 21. Histological analyses showed marked neutrophil infiltration and abscesses containing necrotic and fibrous tissues in the SGM. With this SGM mouse model, we successfully visualized and quantified stable bacterial growth over an extended period of time with bioluminescence imaging, which allowed us to monitor the process of infection without euthanizing the experimental animals. This model is applicable to in vivo evaluations of the long-term efficacy of novel antibiotics or antibacterial implants.
Collapse
Affiliation(s)
- Kenji Yoshioka
- Department of Orthopedic Surgery, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Ken Ishii
- Department of Orthopedic Surgery, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
- Kanagawa Academy of Science and Technology (KAST), Kawasaki, Kanagawa, Japan
- * E-mail:
| | - Tetsuya Kuramoto
- Department of Orthopedic Surgery, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Shigenori Nagai
- Department of Microbiology and Immunology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo, Japan
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Haruki Funao
- Department of Orthopedic Surgery, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Hiroko Ishihama
- Department of Orthopedic Surgery, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Yuta Shiono
- Department of Orthopedic Surgery, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Aya Sasaki
- Department of Pathology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Mamoru Aizawa
- Department of Applied Chemistry, School of Science and Technology, Meiji University, Kawasaki, Kanagawa, Japan
- Kanagawa Academy of Science and Technology (KAST), Kawasaki, Kanagawa, Japan
| | - Yasunori Okada
- Department of Pathology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Shigeo Koyasu
- Department of Microbiology and Immunology, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
- Laboratory for Immune Cell System, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa, Japan
| | - Yoshiaki Toyama
- Department of Orthopedic Surgery, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | - Morio Matsumoto
- Department of Orthopedic Surgery, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| |
Collapse
|
25
|
Sarovich DS, Price EP, Webb JR, Ward LM, Voutsinos MY, Tuanyok A, Mayo M, Kaestli M, Currie BJ. Variable virulence factors in Burkholderia pseudomallei (melioidosis) associated with human disease. PLoS One 2014; 9:e91682. [PMID: 24618705 PMCID: PMC3950250 DOI: 10.1371/journal.pone.0091682] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Accepted: 02/14/2014] [Indexed: 11/18/2022] Open
Abstract
Burkholderia pseudomallei is a Gram-negative environmental bacterium that causes melioidosis, a potentially life-threatening infectious disease affecting mammals, including humans. Melioidosis symptoms are both protean and diverse, ranging from mild, localized skin infections to more severe and often fatal presentations including pneumonia, septic shock with multiple internal abscesses and occasionally neurological involvement. Several ubiquitous virulence determinants in B. pseudomallei have already been discovered. However, the molecular basis for differential pathogenesis has, until now, remained elusive. Using clinical data from 556 Australian melioidosis cases spanning more than 20 years, we identified a Burkholderia mallei-like actin polymerization bimA(Bm) gene that is strongly associated with neurological disease. We also report that a filamentous hemagglutinin gene, fhaB3, is associated with positive blood cultures but is negatively correlated with localized skin lesions without sepsis. We show, for the first time, that variably present virulence factors play an important role in the pathogenesis of melioidosis. Collectively, our study provides a framework for assessing other non-ubiquitous bacterial virulence factors and their association with disease, such as candidate loci identified from large-scale microbial genome-wide association studies.
Collapse
Affiliation(s)
- Derek S. Sarovich
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
| | - Erin P. Price
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
| | - Jessica R. Webb
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
| | - Linda M. Ward
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
| | - Marcos Y. Voutsinos
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
| | - Apichai Tuanyok
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, University of Hawai’i, Honolulu, Hawai’i, United States of America
| | - Mark Mayo
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
| | - Mirjam Kaestli
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
| | - Bart J. Currie
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
- Infectious Diseases Department, Royal Darwin Hospital, Darwin, Northern Territory, Australia
| |
Collapse
|
26
|
Comparative Burkholderia pseudomallei natural history virulence studies using an aerosol murine model of infection. Sci Rep 2014; 4:4305. [PMID: 24603493 PMCID: PMC3945929 DOI: 10.1038/srep04305] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 02/13/2014] [Indexed: 12/24/2022] Open
Abstract
Melioidosis is an endemic disease caused by the bacterium Burkholderia pseudomallei. Concerns exist regarding B. pseudomallei use as a potential bio-threat agent causing persistent infections and typically manifesting as severe pneumonia capable of causing fatal bacteremia. Development of suitable therapeutics against melioidosis is complicated due to high degree of genetic and phenotypic variability among B. pseudomallei isolates and lack of data establishing commonly accepted strains for comparative studies. Further, the impact of strain variation on virulence, disease presentation, and mortality is not well understood. Therefore, this study evaluate and compare the virulence and disease progression of B. pseudomallei strains K96243 and HBPUB10303a, following aerosol challenge in a standardized BALB/c mouse model of infection. The natural history analysis of disease progression monitored conditions such as weight, body temperature, appearance, activity, bacteremia, organ and tissue colonization (pathological and histological analysis) and immunological responses. This study provides a detailed, direct comparison of infection with different B. pseudomallei strains and set up the basis for a standardized model useful to test different medical countermeasures against Burkholderia species. Further, this protocol serves as a guideline to standardize other bacterial aerosol models of infection or to define biomarkers of infectious processes caused by other intracellular pathogens.
Collapse
|
27
|
Webb SE, Karplus E, Miller AL. Retrospective on the development of aequorin and aequorin-based imaging to visualize changes in intracellular free [Ca2+]. Mol Reprod Dev 2014; 82:563-86. [DOI: 10.1002/mrd.22298] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 12/26/2013] [Indexed: 12/17/2022]
Affiliation(s)
- Sarah E. Webb
- Division of Life Science and State Key Laboratory of Molecular Neuroscience; The Hong Kong University of Science and Technology; Kowloon Hong Kong
| | | | - Andrew L. Miller
- Division of Life Science and State Key Laboratory of Molecular Neuroscience; The Hong Kong University of Science and Technology; Kowloon Hong Kong
- Marine Biological Laboratory; Woods Hole Massachusetts
| |
Collapse
|
28
|
Lafontaine ER, Zimmerman SM, Shaffer TL, Michel F, Gao X, Hogan RJ. Use of a safe, reproducible, and rapid aerosol delivery method to study infection by Burkholderia pseudomallei and Burkholderia mallei in mice. PLoS One 2013; 8:e76804. [PMID: 24098563 PMCID: PMC3788738 DOI: 10.1371/journal.pone.0076804] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 08/26/2013] [Indexed: 11/24/2022] Open
Abstract
Burkholderia pseudomallei, the etiologic agent of melioidosis, is a saprophytic bacterium readily isolated from wet soils of countries bordering the equator. Burkholderia mallei is a host-adapted clone of B. pseudomallei that does not persist outside of its equine reservoir and causes the zoonosis glanders, which is endemic in Asia, Africa, the Middle East and South America. Infection by these organisms typically occurs via percutaneous inoculation or inhalation of aerosols, and the most common manifestation is severe pneumonia leading to fatal bacteremia. Glanders and melioidosis are difficult to diagnose and require prolonged antibiotic therapy with low success rates. There are no vaccines available to protect against either Burkholderia species, and there is concern regarding their use as biological warfare agents given that B. mallei has previously been utilized in this manner. Hence, experiments were performed to establish a mouse model of aerosol infection to study the organisms and develop countermeasures. Using a hand-held aerosolizer, BALB/c mice were inoculated intratracheally with strains B. pseudomallei 1026b and B. mallei ATCC23344 and growth of the agents in the lungs, as well as dissemination to the spleen, were examined. Mice infected with 102, 103 and 104 organisms were unable to control growth of B. mallei in the lungs and bacteria rapidly disseminated to the spleen. Though similar results were observed in mice inoculated with 103 and 104B. pseudomallei cells, animals infected with 102 organisms controlled bacterial replication in the lungs, dissemination to the spleen, and the extent of bacteremia. Analysis of sera from mice surviving acute infection revealed that animals produced antibodies against antigens known to be targets of the immune response in humans. Taken together, these data show that small volume aerosol inoculation of mice results in acute disease, dose-dependent chronic infection, and immune responses that correlate with those seen in human infections.
Collapse
Affiliation(s)
- Eric R. Lafontaine
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, United States of America
| | - Shawn M. Zimmerman
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, United States of America
| | - Teresa L. Shaffer
- Department of Microbiology, University of Georgia, Athens, Georgia, United States of America
| | - Frank Michel
- Department of Veterinary Biosciences and Diagnostic Imaging, University of Georgia, Athens, Georgia, United States of America
| | - Xiudan Gao
- Department of Veterinary Biosciences and Diagnostic Imaging, University of Georgia, Athens, Georgia, United States of America
| | - Robert J. Hogan
- Department of Veterinary Biosciences and Diagnostic Imaging, University of Georgia, Athens, Georgia, United States of America
- * E-mail:
| |
Collapse
|
29
|
Mott TM, Johnston RK, Vijayakumar S, Estes DM, Motamedi M, Sbrana E, Endsley JJ, Torres AG. Monitoring Therapeutic Treatments against Burkholderia Infections Using Imaging Techniques. Pathogens 2013; 2. [PMID: 24349761 PMCID: PMC3859531 DOI: 10.3390/pathogens2020383] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Burkholderia mallei, the etiologic agent of glanders, are Category B select agents with biothreat potential, and yet effective therapeutic treatments are lacking. In this study, we showed that CpG administration increased survival, demonstrating protection in the murine glanders model. Bacterial recovery from infected lungs, liver and spleen was significantly reduced in CpG-treated animals as compared with non-treated mice. Reciprocally, lungs of CpG-treated infected animals were infiltrated with higher levels of neutrophils and inflammatory monocytes, as compared to control animals. Employing the B. mallei bioluminescent strain CSM001 and the Neutrophil-Specific Fluorescent Imaging Agent, bacterial dissemination and neutrophil trafficking were monitored in real-time using multimodal in vivo whole body imaging techniques. CpG-treatment increased recruitment of neutrophils to the lungs and reduced bioluminescent bacteria, correlating with decreased bacterial burden and increased protection against acute murine glanders. Our results indicate that protection of CpG-treated animals was associated with recruitment of neutrophils prior to infection and demonstrated, for the first time, simultaneous real time in vivo imaging of neutrophils and bacteria. This study provides experimental evidence supporting the importance of incorporating optimized in vivo imaging methods to monitor disease progression and to evaluate the efficacy of therapeutic treatment during bacterial infections.
Collapse
Affiliation(s)
- Tiffany M Mott
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (T.M.M.); (R.K.J.); (S.V.); (E.S.); (J.J.E.)
| | - R Katie Johnston
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (T.M.M.); (R.K.J.); (S.V.); (E.S.); (J.J.E.)
| | - Sudhamathi Vijayakumar
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (T.M.M.); (R.K.J.); (S.V.); (E.S.); (J.J.E.)
| | - D Mark Estes
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA;
| | - Massoud Motamedi
- Center for Biomedical Engineering, University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - Elena Sbrana
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (T.M.M.); (R.K.J.); (S.V.); (E.S.); (J.J.E.) ; Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Janice J Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (T.M.M.); (R.K.J.); (S.V.); (E.S.); (J.J.E.)
| | - Alfredo G Torres
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (T.M.M.); (R.K.J.); (S.V.); (E.S.); (J.J.E.) ; Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
30
|
Stundick MV, Albrecht MT, Houchens CR, Smith AP, Dreier TM, Larsen JC. Animal models for Francisella tularensis and Burkholderia species: scientific and regulatory gaps toward approval of antibiotics under the FDA Animal Rule. Vet Pathol 2013; 50:877-92. [PMID: 23628693 DOI: 10.1177/0300985813486812] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The development and regulatory approval of medical countermeasures (MCMs) for the treatment and prevention of bacterial threat agent infections will require the evaluation of products in animal models. To obtain regulatory approval, these models must accurately recapitulate aspects of human disease, including, but not necessarily limited to, route of exposure, time to disease onset, pathology, immune response, and mortality. This article focuses on the state of animal model development for 3 agents for which models are largely immature: Francisella tularensis, Burkholderia mallei, and Burkholderia pseudomallei. An overview of available models and a description of scientific and regulatory gaps are provided.
Collapse
Affiliation(s)
- M V Stundick
- US Department of Health and Human Services, Office of the Assistant Secretary for Preparedness and Response, Biomedical Advanced Research and Development Authority, 375 E. St, SW- 12th Floor, Washington, DC 20024, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Construction of mobilizable mini-Tn7 vectors for bioluminescent detection of gram-negative bacteria and single-copy promoter lux reporter analysis. Appl Environ Microbiol 2013; 79:4149-53. [PMID: 23584769 DOI: 10.1128/aem.00640-13] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
We describe the construction of mini-Tn7-based broad-host-range vectors encoding lux genes as bioluminescent reporters. These constructs can be mobilized into the desired host(s) by conjugation for chromosomal mini-Tn7-lux integration and are useful for localization of bacteria during infections or for characterizing regulation of promoters of interest in Gram-negative bacteria.
Collapse
|
32
|
Revelli DA, Boylan JA, Gherardini FC. A non-invasive intratracheal inoculation method for the study of pulmonary melioidosis. Front Cell Infect Microbiol 2012; 2:164. [PMID: 23267442 PMCID: PMC3526731 DOI: 10.3389/fcimb.2012.00164] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 12/06/2012] [Indexed: 11/17/2022] Open
Abstract
Pulmonary melioidosis, a disease manifestation caused by the bacterium Burkholderia pseudomallei, has been studied using aerosols or intranasal (IN) inoculation in small animal models. Both have inherent disadvantages which may not accurately model primary pulmonary melioidosis in humans. Intratracheal inoculation (IT) by direct visualization of the tracheal opening offers an alternative technique for infection that overcomes the disadvantages of aerosol and IN challenge. In this study, we describe a method which requires relatively inexpensive equipment, little training, and is compliant with the operational constraints of a BSL3 laboratory. Results obtained using trypan blue demonstrated that an inoculum can be accurately delivered into the lungs of mice within a biosafety cabinet (BSC). Whole body imaging and histopathology confirmed that mice inoculated intratracheally with B. pseudomallei develop the primary focus of infection in the lungs, and not the nasal passages which can lead to invasion of the central nervous system and potential neurologic complications. Further, based on colony counts and bioluminescent imaging, dissemination to secondary organs occurred as expected. Taken together, this intratracheal method of inoculation fulfills four goals: (1) to accurately deliver B. pseudomallei into the lungs of the animal model, (2) to avoid potentially confounding complications due to primary infections at sites other than the lung, (3) to maintain normal organ dissemination, and (4) to be BSL3 compliant.
Collapse
Affiliation(s)
- David A Revelli
- Laboratory of Zoonotic Pathogens, National Institute of Allergy and Infectious Diseases, Rocky Mountain Laboratories Hamilton, MT, USA
| | | | | |
Collapse
|
33
|
Sun Y, Connor MG, Pennington JM, Lawrenz MB. Development of bioluminescent bioreporters for in vitro and in vivo tracking of Yersinia pestis. PLoS One 2012; 7:e47123. [PMID: 23071730 PMCID: PMC3469486 DOI: 10.1371/journal.pone.0047123] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 09/10/2012] [Indexed: 12/21/2022] Open
Abstract
Yersinia pestis causes an acute infection known as the plague. Conventional techniques to enumerate Y. pestis can be labor intensive and do not lend themselves to high throughput assays. In contrast, bioluminescent bioreporters produce light that can be detected using plate readers or optical imaging platforms to monitor bacterial populations as a function of luminescence. Here, we describe the development of two Y. pestis chromosomal-based luxCDABE bioreporters, Lux(PtolC) and Lux(PcysZK). These bioreporters use constitutive promoters to drive expression of luxCDABE that allow for sensitive detection of bacteria via bioluminescence in vitro. Importantly, both bioreporters demonstrate a direct correlation between bacterial numbers and bioluminescence, which allows for bioluminescence to be used to compare bacterial numbers. We demonstrate the use of these bioreporters to test antimicrobial inhibitors (Lux(PtolC)) and monitor intracellular survival (Lux(PtolC) and Lux(PcysZK)) in vitro. Furthermore, we show that Y. pestis infection of the mouse model can be monitored using whole animal optical imaging in real time. Using optical imaging, we observed Y. pestis dissemination and differentiated between virulence phenotypes in live animals via bioluminescence. Finally, we demonstrate that whole animal optical imaging can identify unexpected colonization patterns in mutant-infected animals.
Collapse
Affiliation(s)
| | | | | | - Matthew B. Lawrenz
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
34
|
A non-invasive in vivo imaging system to study dissemination of bioluminescent Yersinia pestis CO92 in a mouse model of pneumonic plague. Microb Pathog 2012; 55:39-50. [PMID: 23063826 DOI: 10.1016/j.micpath.2012.09.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 09/25/2012] [Accepted: 09/27/2012] [Indexed: 01/25/2023]
Abstract
The gold standard in microbiology for monitoring bacterial dissemination in infected animals has always been viable plate counts. This method, despite being quantitative, requires sacrificing the infected animals. Recently, however, an alternative method of in vivo imaging of bioluminescent bacteria (IVIBB) for monitoring microbial dissemination within the host has been employed. Yersinia pestis is a Gram-negative bacterium capable of causing bubonic, septicemic, and pneumonic plague. In this study, we compared the conventional counting of bacterial colony forming units (cfu) in the various infected tissues to IVIBB in monitoring Y. pestis dissemination in a mouse model of pneumonic plague. By using a transposon mutagenesis system harboring the luciferase (luc) gene, we screened approximately 4000 clones and obtained a fully virulent, luc-positive Y. pestis CO92 (Y. pestis-luc2) reporter strain in which transposition occurred within the largest pMT1 plasmid which possesses murine toxin and capsular antigen encoding genes. The aforementioned reporter strain and the wild-type CO92 exhibited similar growth curves, formed capsule based on immunofluorescence microscopy and flow cytometry, and had a similar LD(50). Intranasal infection of mice with 15 LD(50) of CO92-luc2 resulted in animal mortality by 72 h, and an increasing number of bioluminescent bacteria were observed in various mouse organs over a 24-72 h period when whole animals were imaged. However, following levofloxacin treatment (10 mg/kg/day) for 6 days 24 h post infection, no luminescence was observed after 72 h of infection, indicating that the tested antimicrobial killed bacteria preventing their detection in host peripheral tissues. Overall, we demonstrated that IVIBB is an effective and non-invasive way of monitoring bacterial dissemination in animals following pneumonic plague having strong correlation with cfu, and our reporter CO92-luc2 strain can be employed as a useful tool to monitor the efficacy of antimicrobial countermeasures in real time.
Collapse
|
35
|
Torres AG, Cieza RJ, Rojas-Lopez M, Blumentritt CA, Souza CS, Johnston RK, Strockbine N, Kaper JB, Sbrana E, Popov VL. In vivo bioluminescence imaging of Escherichia coli O104:H4 and role of aerobactin during colonization of a mouse model of infection. BMC Microbiol 2012; 12:112. [PMID: 22716772 PMCID: PMC3438087 DOI: 10.1186/1471-2180-12-112] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 06/06/2012] [Indexed: 12/21/2022] Open
Abstract
Background A major outbreak of bloody diarrhea associated with Shiga toxin-producing Escherichia coli O104:H4 occurred early in 2011, to which an unusual number of hemolytic uremic syndrome cases were linked. Due to limited information regarding pathogenesis and/or virulence properties of this particular serotype, we investigated the contribution of the aerobactin iron transport system during in vitro and in vivo conditions. Results A bioluminescent reporter construct was used to perform real-time monitoring of E. coli O104:H4 in a mouse model of infection. We verified that our reporter strain maintained characteristics and growth kinetics that were similar to those of the wild-type E. coli strain. We found that the intestinal cecum of ICR (CD-1) mice was colonized by O104:H4, with bacteria persisting for up to 7 days after intragastric inoculation. MALDI-TOF analysis of heat-extracted proteins was performed to identify putative surface-exposed virulence determinants. A protein with a high similarity to the aerobactin iron receptor was identified and further demonstrated to be up-regulated in E. coli O104:H4 when grown on MacConkey agar or during iron-depleted conditions. Because the aerobactin iron acquisition system is a key virulence factor in Enterobacteriaceae, an isogenic aerobactin receptor (iutA) mutant was created and its intestinal fitness assessed in the murine model. We demonstrated that the aerobactin mutant was out-competed by the wild-type E. coli O104:H4 during in vivo competition experiments, and the mutant was unable to persist in the cecum. Conclusion Our findings demonstrate that bioluminescent imaging is a useful tool to monitor E. coli O104:H4 colonization properties, and the murine model can become a rapid way to evaluate bacterial factors associated with fitness and/or colonization during E. coli O104:H4 infections.
Collapse
Affiliation(s)
- Alfredo G Torres
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555-1070, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|