1
|
Zhang W, Yao Y, Zhou H, He J, Wang J, Li L, Gao M, Liu X, Shi Y, Lin J, Liu J, Chen H, Feng Y, Zhou Z, Yu Y, Hua X. Interactions between host epithelial cells and Acinetobacter baumannii promote the emergence of highly antibiotic resistant and highly mucoid strains. Emerg Microbes Infect 2022; 11:2556-2569. [PMID: 36227610 PMCID: PMC9621264 DOI: 10.1080/22221751.2022.2136534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Acinetobacter baumannii is an important nosocomial pathogen. Upon colonizing a host, A. baumannii are subjected to selective pressure by immune defenses as they adapt to the host environment. However, the mechanism of this pathoadaptation is unknown. Here, we established an in vitro system to evolve A. baumannii driven by the continuous selective pressure exerted by epithelial cells, and we used a combination of experimental evolution, phenotypic characterization and multi-omics analysis to address the underlying mechanism. When continuously exposed to selective pressure by pulmonary epithelial cells, A. baumannii showed ptk mutation-mediated mucoid conversion (reduced adhesion and increased anti-phagocytic ability) by enhancement of capsular exopolysaccharide chain length; rsmG mutation-mediated deficiency of 7-methylguanosine modification in the 524th nucleotide of 16S rRNA, which increased ribosome translation efficiency; and rnaseI mutation-mediated changes in outer membrane permeability and efflux pump expression. Together, these mutations altered susceptibility to a variety of antimicrobial agents, including the novel antibiotic cefiderocol, by regulating siderophore and siderophore-receptor biosynthesis. In conclusion, pulmonary epithelial cells modulate A. baumannii pathoadaptation, implicating the host–microbe interaction in the survival and persistence of A. baumannii.
Collapse
Affiliation(s)
- Wang Zhang
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| | - Yue Yao
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| | - Hua Zhou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jintao He
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| | - Jingfen Wang
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| | - Li Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Minsong Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaochen Liu
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| | - Ya Shi
- Hangzhou Digital-Micro Biotech Co., Ltd., Hangzhou, Zhejiang, China
| | - Jinzhong Lin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianzhao Liu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, China.,Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huan Chen
- Hangzhou Digital-Micro Biotech Co., Ltd., Hangzhou, Zhejiang, China
| | - Yu Feng
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Biophysics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhihui Zhou
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| | - Yunsong Yu
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| | - Xiaoting Hua
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| |
Collapse
|
2
|
Fan Q, Wang C, Guo R, Jiang X, Li W, Chen X, Li K, Hong W. Step-by-step dual stimuli-responsive nanoparticles for efficient bacterial biofilm eradication. Biomater Sci 2021; 9:6889-6902. [PMID: 34519743 DOI: 10.1039/d1bm01038g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Biofilm-related bacterial infections are extremely resistant to antibiotics, mainly due to the impermeability of the intensive matrices, which allow the bacteria to survive antibiotic treatment. Herein, step-by-step dual stimuli-responsive azithromycin-loaded nanoparticles (CM/AZM@Tyr) was constructed for efficient biofilm eradication. CM/AZM@Tyr was prepared by the self-assembly of poly(ε-caprolactone)-polyethylene glycol-polyethylenimine (PCL-PEG-PEI) into cationic micelles and simultaneously encapsulated AZM into the hydrophobic core, which is further bound with cis-aconityl-D-tyrosine (CA-Tyr) through electrostatic interaction. Upon initial penetration, CM/AZM@Tyr could show step-by-step dual-response to the microenvironment of biofilms. Firstly, the CA-Tyr shell rapidly responded to the acidic microenvironment and released D-Tyr to disassemble the biofilm mass. Then, the exposed cationic CM/AZM micelles could bind firmly to the negatively-charged bacteria cell membrane. With the enzymolysis of the PCL core, the rapidly releasing AZM could kill the bacteria over the depth of biofilms. Massive accumulation was observed in the infected lungs of biofilms-associated lung infection mice after the i.v. injection of CM/Cy5.5@Tyr under the 3D mode of the in vivo Imaging System. Reduced bacterial burden and alleviated fibrosis in the infected lungs were also obtained after treatment with CM/AZM@Tyr mainly due to its intensive penetration in the biofilm and the orderly release of the biofilm dispersant and antimicrobial agents. In summary, this research developed an effective strategy for the treatment of blood-accessible biofilm-induced infections.
Collapse
Affiliation(s)
- Qing Fan
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, P. R. China.
| | - Changrong Wang
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, P. R. China.
| | - Rong Guo
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, P. R. China.
| | - Xinyu Jiang
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, P. R. China.
| | - Wenting Li
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, P. R. China.
| | - Xiangjun Chen
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, P. R. China.
| | - Keke Li
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, P. R. China.
| | - Wei Hong
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, P. R. China.
| |
Collapse
|
3
|
Chanjitwiriya K, Roytrakul S, Kunthalert D. Quercetin negatively regulates IL-1β production in Pseudomonas aeruginosa-infected human macrophages through the inhibition of MAPK/NLRP3 inflammasome pathways. PLoS One 2020; 15:e0237752. [PMID: 32817626 PMCID: PMC7446918 DOI: 10.1371/journal.pone.0237752] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 07/31/2020] [Indexed: 01/02/2023] Open
Abstract
Pseudomonas aeruginosa remains a leading cause of nosocomial and serious life-threatening infections, and contributes to increased mortality in immunocompromised individuals. P. aeruginosa infection triggers host immune response and often provokes potent inflammatory mediators, which do not necessarily eradicate the causative pathogen. On the other hand, it causes severe airway damage and eventually decreased lung function. Such unfavorable outcomes of inflammatory injury have necessitated the development of novel effective agents that can combat with P. aeruginosa-mediated inflammation. Herein, we investigated the potential of quercetin in regulating P. aeruginosa-induced inflammation, with particular emphasized on the interleukin-1β (IL-1β). Our results showed that quercetin exerted the potent inhibitory activity against the production of IL-1β in macrophages infected by live P. aeruginosa PAO1, without exhibiting cytotoxicity. According to our settings, such the potent inhibitory activity of quercetin was clearly demonstrated through its ability to efficiently inhibit IL-1β during P. aeruginosa infection, pre- or even post-infection. In addition, quercetin strongly suppressed MAPK signaling pathway by inhibiting phosphorylation of the p38 MAPK and JNK2, and molecular docking study supported well with this observation. Moreover, quercetin reduced the NLRP3 expression and inhibited the P. aeruginosa-mediated cleavage of caspase-1 as well as mature IL-1β. These results thus indicated that quercetin inhibition of P. aeruginosa-induced IL-1β production is mediated by suppressing the initial priming step and by inhibiting the NLRP3 inflammasome activation. Taken together, our findings demonstrated the promising regulatory activity of quercetin against IL-1β production in P. aeruginosa-infected macrophages, and indicated that quercetin has the potential to be effective as a novel therapeutic agent for treatment of P. aeruginosa-induced inflammation.
Collapse
Affiliation(s)
- Kasem Chanjitwiriya
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Sittiruk Roytrakul
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Thailand Science Park, Pathumthani, Thailand
| | - Duangkamol Kunthalert
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
- Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
- * E-mail: ,
| |
Collapse
|
4
|
Chatterjee P, Sass G, Swietnicki W, Stevens DA. Review of Potential Pseudomonas Weaponry, Relevant to the Pseudomonas-Aspergillus Interplay, for the Mycology Community. J Fungi (Basel) 2020; 6:jof6020081. [PMID: 32517271 PMCID: PMC7345761 DOI: 10.3390/jof6020081] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/03/2020] [Accepted: 06/03/2020] [Indexed: 12/15/2022] Open
Abstract
Pseudomonas aeruginosa is one of the most prominent opportunistic bacteria in airways of cystic fibrosis patients and in immunocompromised patients. These bacteria share the same polymicrobial niche with other microbes, such as the opportunistic fungus Aspergillus fumigatus. Their inter-kingdom interactions and diverse exchange of secreted metabolites are responsible for how they both fare in competition for ecological niches. The outcomes of their contests likely determine persistent damage and degeneration of lung function. With a myriad of virulence factors and metabolites of promising antifungal activity, P. aeruginosa products or their derivatives may prove useful in prophylaxis and therapy against A. fumigatus. Quorum sensing underlies the primary virulence strategy of P. aeruginosa, which serves as cell–cell communication and ultimately leads to the production of multiple virulence factors. Understanding the quorum-sensing-related pathogenic mechanisms of P. aeruginosa is a first step for understanding intermicrobial competition. In this review, we provide a basic overview of some of the central virulence factors of P. aeruginosa that are regulated by quorum-sensing response pathways and briefly discuss the hitherto known antifungal properties of these virulence factors. This review also addresses the role of the bacterial secretion machinery regarding virulence factor secretion and maintenance of cell–cell communication.
Collapse
Affiliation(s)
- Paulami Chatterjee
- California Institute for Medical Research, San Jose, CA 95128, USA; (P.C.); (G.S.)
| | - Gabriele Sass
- California Institute for Medical Research, San Jose, CA 95128, USA; (P.C.); (G.S.)
| | - Wieslaw Swietnicki
- Department of Immunology of Infectious Diseases, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 50-114 Wroclaw, Poland;
| | - David A. Stevens
- California Institute for Medical Research, San Jose, CA 95128, USA; (P.C.); (G.S.)
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Correspondence: ; Tel.: +1-408-998-4554
| |
Collapse
|
5
|
Efficacy of Aerosolized Rifaximin versus Tobramycin for Treatment of Pseudomonas aeruginosa Pneumonia in Mice. Antimicrob Agents Chemother 2019; 63:AAC.02341-18. [PMID: 31010865 DOI: 10.1128/aac.02341-18] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 04/16/2019] [Indexed: 02/08/2023] Open
Abstract
Pseudomonas aeruginosa is a Gram-negative opportunistic bacterial pathogen that can cause chronic lung infections in patients with cystic fibrosis (CF). The current preferred treatment for CF lung infections includes inhaled tobramycin (TOB); however, studies suggest TOB cannot effectively inhibit biofilm formation. Using an NIH small compounds drug library approved for safe use in humans, we identified rifaximin (RFX), a semisynthetic, rifamycin family, nonsystemic antibiotic that inhibits alginate production and growth in P. aeruginosa Inhibition of alginate production was further analyzed using the uronic acid carbazole assay and a promoter reporter assay that measures the transcription of the alginate biosynthetic operon. Compared to TOB, RFX significantly reduced alginate production in laboratory and CF sputum isolates of P. aeruginosa In addition, RFX showed a narrow range of MICs when measured with multidrug-resistant bacterial species of clinical relevance, synergistic activities with TOB or amikacin against clinical isolates, as well as reduction toward in vitro preformed biofilms. In C57BL/6 mice, penetration of nebulized TOB into the lungs was shown at a higher level than that of RFX. Further, in vivo assessment using a DBA/2 mouse lung infection model found increased survival rates with a single-dose treatment of nebulized RFX and decreased P. aeruginosa PAO1 bioburden with a multiple-dose treatment of RFX plus TOB. In addition, mice treated with a single exposure to dimethyl sulfoxide (DMSO), a solvent that dissolves RFX, showed no apparent toxicity. In summary, RFX may be used to supplement TOB inhalation therapy to increase efficacy against P. aeruginosa biofilm infections.
Collapse
|
6
|
Malhotra S, Hayes D, Wozniak DJ. Cystic Fibrosis and Pseudomonas aeruginosa: the Host-Microbe Interface. Clin Microbiol Rev 2019; 32:e00138-18. [PMID: 31142499 PMCID: PMC6589863 DOI: 10.1128/cmr.00138-18] [Citation(s) in RCA: 258] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In human pathophysiology, the clash between microbial infection and host immunity contributes to multiple diseases. Cystic fibrosis (CF) is a classical example of this phenomenon, wherein a dysfunctional, hyperinflammatory immune response combined with chronic pulmonary infections wreak havoc upon the airway, leading to a disease course of substantial morbidity and shortened life span. Pseudomonas aeruginosa is an opportunistic pathogen that commonly infects the CF lung, promoting an accelerated decline of pulmonary function. Importantly, P. aeruginosa exhibits significant resistance to innate immune effectors and to antibiotics, in part, by expressing specific virulence factors (e.g., antioxidants and exopolysaccharides) and by acquiring adaptive mutations during chronic infection. In an effort to review our current understanding of the host-pathogen interface driving CF pulmonary disease, we discuss (i) the progression of disease within the primitive CF lung, specifically focusing on the role of host versus bacterial factors; (ii) critical, neutrophil-derived innate immune effectors that are implicated in CF pulmonary disease, including reactive oxygen species (ROS) and antimicrobial peptides (e.g., LL-37); (iii) P. aeruginosa virulence factors and adaptive mutations that enable evasion of the host response; and (iv) ongoing work examining the distribution and colocalization of host and bacterial factors within distinct anatomical niches of the CF lung.
Collapse
Affiliation(s)
- Sankalp Malhotra
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Don Hayes
- The Ohio State University College of Medicine, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
- Section of Pulmonary Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Daniel J Wozniak
- The Ohio State University College of Medicine, Columbus, Ohio, USA
- Section of Pulmonary Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
7
|
Zhao M, Li W, Liu K, Li H, Lan X. C4-HSL aptamers for blocking qurom sensing and inhibiting biofilm formation in Pseudomonas aeruginosa and its structure prediction and analysis. PLoS One 2019; 14:e0212041. [PMID: 30779754 PMCID: PMC6380626 DOI: 10.1371/journal.pone.0212041] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 01/25/2019] [Indexed: 12/13/2022] Open
Abstract
This study aimed to screen DNA aptamers against the signal molecule C4-HSL of the rhl system for the inhibition of biofilm formation of Pseudomonas aeruginosa using an improved systematic evolution of ligand by exponential enrichment (SELEX) method based on a structure-switching fluorescent activating bead. The aptamers against the C4-HSL with a high affinity and specifity were successfully obtained and evaluated in real-time by this method. Results of biofilm inhibition experiments in vitro showed that the biofilm formation of P. aeruginosa was efficiently reduced to about 1/3 by the aptamers compared with that of the groups without the aptamers. Independent secondary structure simulation and computer-aided tertiary structure prediction (3dRNA) showed that the aptamers contained a highly conserved Y-shaped structural unit. Therefore, this study benefits the search for new methods for the detection and treatment of P. aeruginosa biofilm formation.
Collapse
Affiliation(s)
- Meng Zhao
- Second Military Medical University, Shanghai, China
- Institute for Laboratory Medicine, The 900th Hospital of Joint Service Support Force, Fuzhou, Fujian, China
| | - Weibin Li
- Institute for Laboratory Medicine, The 900th Hospital of Joint Service Support Force, Fuzhou, Fujian, China
| | - Kuancan Liu
- Institute for Laboratory Medicine, The 900th Hospital of Joint Service Support Force, Fuzhou, Fujian, China
| | - Huiling Li
- Institute for Laboratory Medicine, The 900th Hospital of Joint Service Support Force, Fuzhou, Fujian, China
| | - Xiaopeng Lan
- Second Military Medical University, Shanghai, China
- Institute for Laboratory Medicine, The 900th Hospital of Joint Service Support Force, Fuzhou, Fujian, China
- * E-mail:
| |
Collapse
|
8
|
Pandey S, Delgado C, Kumari H, Florez L, Mathee K. Outer-membrane protein LptD (PA0595) plays a role in the regulation of alginate synthesis in Pseudomonas aeruginosa. J Med Microbiol 2018; 67:1139-1156. [PMID: 29923820 DOI: 10.1099/jmm.0.000752] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
PURPOSE The presence of alginate-overproducing (Alg+) strains of Pseudomonas aeruginosa in cystic fibrosis patients is indicative of chronic infection. The Alg+ phenotype is generally due to a mutation in the mucA gene, encoding an innermembrane protein that sequesters AlgT/U, the alginate-specific sigma factor. AlgT/U release from the anti-sigma factor MucA is orchestrated via a complex cascade called regulated intramembrane proteolysis. The goal of this study is to identify new players involved in the regulation of alginate production. METHODOLOGY Previously, a mutant with a second-site suppressor of alginate production (sap), sap27, was isolated from the constitutively Alg+ PDO300 that harbours the mucA22 allele. A cosmid from a P. aeruginosa minimum tiling path library was identified via en masse complementation of sap27. The cosmid was transposon mutagenized to map the contributing gene involved in the alginate production. The identified gene was sequenced in sap27 along with algT/U, mucA, algO and mucP. The role of the novel gene was explored using precise in-frame algO and algW deletion mutants of PAO1 and PDO300.Results/Key findings. The gene responsible for restoring the mucoid phenotype was mapped to lptD encoding an outer-membrane protein. However, the sequencing of sap27 revealed a mutation in algO, but not in lptD. In addition, we demonstrate that lipopolysaccharide transport protein D (LptD)-dependent alginate production requires AlgW in PAO1 and AlgO in PDO300. CONCLUSION LptD plays a specific role in alginate production. Our findings suggest that there are two pathways for the production of alginate in P. aeruginosa, one involving AlgW in the wild-type, and one involving AlgO in the mucA22 mutant.
Collapse
Affiliation(s)
- Sundar Pandey
- 1Department of Biological Sciences, College of Arts Sciences and Education, Florida International University, Miami, FL, USA
| | - Camila Delgado
- 2Department of Microbiology and Infectious Diseases, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.,†Present address: Langone Medical Center, New York University School of Medicine, New York, USA
| | - Hansi Kumari
- 2Department of Microbiology and Infectious Diseases, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.,3Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Laura Florez
- 2Department of Microbiology and Infectious Diseases, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Kalai Mathee
- 4Biomolecular Sciences Institute, Florida International University, Miami, FL, USA.,2Department of Microbiology and Infectious Diseases, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.,3Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| |
Collapse
|
9
|
Banaschewski BJH, Baer B, Arsenault C, Jazey T, Veldhuizen EJA, Delport J, Gooyers T, Lewis JF, Haagsman HP, Veldhuizen RAW, Yamashita C. The Antibacterial and Anti-inflammatory Activity of Chicken Cathelicidin-2 combined with Exogenous Surfactant for the Treatment of Cystic Fibrosis-Associated Pathogens. Sci Rep 2017; 7:15545. [PMID: 29138462 PMCID: PMC5686076 DOI: 10.1038/s41598-017-15558-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 10/30/2017] [Indexed: 01/12/2023] Open
Abstract
Cystic fibrosis (CF) is characterized by recurrent airway infections with antibiotic-resistant bacteria and chronic inflammation. Chicken cathelicin-2 (CATH-2) has been shown to exhibit antimicrobial activity against antibiotic-resistant bacteria and to reduce inflammation. In addition, exogenous pulmonary surfactant has been suggested to enhance pulmonary drug delivery. It was hypothesized that CATH-2 when combined with an exogenous surfactant delivery vehicle, bovine lipid extract surfactant (BLES), would exhibit antimicrobial activity against CF-derived bacteria and downregulate inflammation. Twelve strains of CF-pathogens were exposed to BLES+CATH-2 in vitro and killing curves were obtained to determine bactericidal activity. Secondly, heat-killed bacteria were administered in vivo to elicit a pro-inflammatory response with either a co-administration or delayed administration of BLES+CATH-2 to assess the antimicrobial-independent, anti-inflammatory properties of BLES+CATH-2. CATH-2 alone exhibited potent antimicrobial activity against all clinical strains of antibiotic-resistant bacteria, while BLES+CATH-2 demonstrated a reduction, but significant antimicrobial activity against bacterial isolates. Furthermore, BLES+CATH-2 reduced inflammation in vivo when either co-administered with killed bacteria or after delayed administration. The use of a host-defense peptide combined with an exogenous surfactant compound, BLES+CATH-2, is shown to exhibit antimicrobial activity against antibiotic-resistant CF bacterial isolates and reduce inflammation.
Collapse
Affiliation(s)
| | - Brandon Baer
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
| | - Christina Arsenault
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
| | - Teah Jazey
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
| | - Edwin J A Veldhuizen
- Department of Infectious Diseases and Immunology, Division of Molecular Host Defense, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Johan Delport
- London Health Sciences Centre, London, Ontario, Canada
| | | | - James F Lewis
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada.,Department of Medicine, Western University, London, Ontario, Canada
| | - Henk P Haagsman
- Department of Infectious Diseases and Immunology, Division of Molecular Host Defense, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Ruud A W Veldhuizen
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada.,Department of Medicine, Western University, London, Ontario, Canada
| | - Cory Yamashita
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada. .,Department of Medicine, Western University, London, Ontario, Canada.
| |
Collapse
|
10
|
Exploring the mechanisms of Yinlai Decoction intervening the mice model of FM1 virus infected compound with high-fat and protein-diet. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2017. [DOI: 10.1016/j.jtcms.2017.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
11
|
Dosunmu EF, Emeh RO, Dixit S, Bakeer MK, Coats MT, Owen DR, Pillai SR, Singh SR, Dennis VA. The anti-microbial peptide TP359 attenuates inflammation in human lung cells infected with Pseudomonas aeruginosa via TLR5 and MAPK pathways. PLoS One 2017; 12:e0176640. [PMID: 28467446 PMCID: PMC5415104 DOI: 10.1371/journal.pone.0176640] [Citation(s) in RCA: 17] [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: 02/13/2017] [Accepted: 04/13/2017] [Indexed: 11/19/2022] Open
Abstract
Pseudomonas aeruginosa infection induces vigorous inflammatory mediators secreted by epithelial cells, which do not necessarily eradicate the pathogen. Nonetheless, it reduces lung function due to significant airway damage, most importantly in cystic fibrosis patients. Recently, we published that TP359, a proprietary cationic peptide had potent bactericidal effects against P. aeruginosa, which were mediated by down-regulating its outer membrane biogenesis genes. Herein, we hypothesized that TP359 bactericidal effects could also serve to regulate P. aeruginosa-induced lung inflammation. We explored this hypothesis by infecting human A549 lung cells with live P. aeruginosa non-isogenic, mucoid and non-mucoid strains and assessed the capacity of TP359 to regulate the levels of elicited TNFα, IL-6 and IL-8 inflammatory cytokines. In all instances, the mucoid strain elicited higher concentrations of cytokines in comparison to the non-mucoid strain, and TP359 dose-dependently down-regulated their respective levels, suggesting its regulation of lung inflammation. Surprisingly, P. aeruginosa flagellin, and not its lipopolysaccharide moiety, was the primary inducer of inflammatory cytokines in lung cells, which were similarly down-regulated by TP359. Blocking of TLR5, the putative flagellin receptor, completely abrogated the capacity of infected lung cells to secrete cytokines, underscoring that TP359 regulates inflammation via the TLR5-dependent signaling pathway. Downstream pathway-specific inhibition studies further revealed that the MAPK pathway, essentially p38 and JNK are necessary for induction of P. aeruginosa elicited inflammatory cytokines and their down-regulation by TP359. Collectively, our data provides evidence to support exploring the relevancy of TP359 as an anti-microbial and anti-inflammatory agent against P. aeruginosa for clinical applications.
Collapse
Affiliation(s)
- Ejovwoke F. Dosunmu
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, Alabama, United States of America
| | - Robert O. Emeh
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, Alabama, United States of America
| | - Saurabh Dixit
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, Alabama, United States of America
| | - Mona K. Bakeer
- Lousiana State University Health Sciences Center, School of Allied Health Professions, New Orleans, Louisiana, United States of America
| | - Mamie T. Coats
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, Alabama, United States of America
| | - Donald R. Owen
- Therapeutic Peptides Inc., Baton Rouge, Louisiana, United States of America
| | - Shreekumar R. Pillai
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, Alabama, United States of America
| | - Shree R. Singh
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, Alabama, United States of America
| | - Vida A. Dennis
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, Alabama, United States of America
- * E-mail:
| |
Collapse
|
12
|
Abstract
The recent increase in genomic data is revealing an unexpected perspective of gene loss as a pervasive source of genetic variation that can cause adaptive phenotypic diversity. This novel perspective of gene loss is raising new fundamental questions. How relevant has gene loss been in the divergence of phyla? How do genes change from being essential to dispensable and finally to being lost? Is gene loss mostly neutral, or can it be an effective way of adaptation? These questions are addressed, and insights are discussed from genomic studies of gene loss in populations and their relevance in evolutionary biology and biomedicine.
Collapse
|
13
|
Metabolism and Pathogenicity of Pseudomonas aeruginosa Infections in the Lungs of Individuals with Cystic Fibrosis. Microbiol Spectr 2016; 3. [PMID: 26350318 DOI: 10.1128/microbiolspec.mbp-0003-2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Individuals with the genetic disease cystic fibrosis (CF) accumulate mucus or sputum in their lungs. This sputum is a potent growth substrate for a range of potential pathogens, and the opportunistic bacterium Pseudomonas aeruginosa is generally most difficult of these to eradicate. As a result, P. aeruginosa infections are frequently maintained in the CF lung throughout life, and are the leading cause of death for these individuals. While great effort has been expended to better understand and treat these devastating infections, only recently have researchers begun to rigorously examine the roles played by specific nutrients in CF sputum to cue P. aeruginosa pathogenicity. This chapter summarizes the current state of knowledge regarding how P. aeruginosa metabolism in CF sputum affects initiation and maintenance of these infections. It contains an overview of CF lung disease and the mechanisms of P. aeruginosa pathogenicity. Several model systems used to study these infections are described with emphasis on the challenge of replicating the chronic infections observed in humans with CF. Nutrients present in CF sputum are surveyed, and the impacts of these nutrients on the infection are discussed. The chapter concludes by addressing the future of this line of research including the use of next-generation technologies and the potential for metabolism-based therapeutics.
Collapse
|
14
|
Min KB, Lee KM, Oh YT, Yoon SS. Nonmucoid conversion of mucoid Pseudomonas aeruginosa induced by sulfate-stimulated growth. FEMS Microbiol Lett 2014; 360:157-66. [PMID: 25227776 DOI: 10.1111/1574-6968.12600] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 09/11/2014] [Indexed: 11/27/2022] Open
Abstract
Alginate-overproducing mucoid Pseudomonas aeruginosa, responsible for chronic airway infections in cystic fibrosis (CF) patients, is resistant to antibiotic treatments and host immune clearance. In this study, we performed a phenotype microarray screen and identified sulfate ion as a molecule that can suppress alginate production. When a mucoid P. aeruginosa strain CM21 and additional mucoid isolates were grown with 5% sodium sulfate, significantly decreased levels of alginate were produced. Suppression of alginate production was also induced by other sulfate salts. Expression of a reporter gene fused to the algD promoter was considerably decreased when grown with sulfate. Furthermore, bacterial cell shape was abnormally altered in CM21, but not in PAO1, a prototype nonmucoid strain, suggesting that sulfate-stimulated cell shape change is associated with transcriptional suppression of the alginate operon. Finally, a CM21 lpxC mutant defective in lipid A biosynthesis continued to produce alginate and maintained the correct cell shape when grown with sulfate. These results suggest a potential involvement of lipoploysaccharide biosynthesis in the sulfate-induced reversion to nonmucoid phenotype. This study proposes a novel strategy that can be potentially applied to treat persistent infection by recalcitrant mucoid P. aeruginosa.
Collapse
Affiliation(s)
- Kyung Bae Min
- Department of Microbiology and Immunology, Brain Korea PLUS Project for Medical Science, Seoul, Korea
| | | | | | | |
Collapse
|
15
|
Kukavica-Ibrulj I, Facchini M, Cigana C, Levesque RC, Bragonzi A. Assessing Pseudomonas aeruginosa virulence and the host response using murine models of acute and chronic lung infection. Methods Mol Biol 2014; 1149:757-71. [PMID: 24818948 DOI: 10.1007/978-1-4939-0473-0_58] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Murine models of acute and chronic lung infection have been used in studying Pseudomonas aeruginosa for assessing in vivo behavior and for monitoring of the host response. These models provide an important resource for studies of the initiation and maintenance of bacterial infection, identify bacterial genes essential for in vivo maintenance and for the development and testing of new therapies. The rat has been used extensively as a model of chronic lung infection, whereas the mouse has been a model of acute and chronic infection. Intratracheal administration of planktonic bacterial cells in the mouse provides a model of acute pneumonia. Bacteria enmeshed in agar beads can be used in the rat and mouse to reproduce the lung pathology of cystic fibrosis patients with advanced chronic pulmonary disease. Here, we describe the methods to assess virulence of P. aeruginosa using prototype and clinical strains in the Sprague-Dawley rat and the C57BL/6NCrlBR mouse by monitoring several measurable read-outs including weight loss, mortality, in vivo growth curves, the competitive index of infectivity, and the inflammatory response.
Collapse
Affiliation(s)
- Irena Kukavica-Ibrulj
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, 1030 av. de la médecine, Québec, QC, Canada, G1V 0A6
| | | | | | | | | |
Collapse
|
16
|
Miskinyte M, Sousa A, Ramiro RS, de Sousa JAM, Kotlinowski J, Caramalho I, Magalhães S, Soares MP, Gordo I. The genetic basis of Escherichia coli pathoadaptation to macrophages. PLoS Pathog 2013; 9:e1003802. [PMID: 24348252 PMCID: PMC3861542 DOI: 10.1371/journal.ppat.1003802] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 10/14/2013] [Indexed: 12/25/2022] Open
Abstract
Antagonistic interactions are likely important driving forces of the evolutionary process underlying bacterial genome complexity and diversity. We hypothesized that the ability of evolved bacteria to escape specific components of host innate immunity, such as phagocytosis and killing by macrophages (MΦ), is a critical trait relevant in the acquisition of bacterial virulence. Here, we used a combination of experimental evolution, phenotypic characterization, genome sequencing and mathematical modeling to address how fast, and through how many adaptive steps, a commensal Escherichia coli (E. coli) acquire this virulence trait. We show that when maintained in vitro under the selective pressure of host MΦ commensal E. coli can evolve, in less than 500 generations, virulent clones that escape phagocytosis and MΦ killing in vitro, while increasing their pathogenicity in vivo, as assessed in mice. This pathoadaptive process is driven by a mechanism involving the insertion of a single transposable element into the promoter region of the E. coli yrfF gene. Moreover, transposition of the IS186 element into the promoter of Lon gene, encoding an ATP-dependent serine protease, is likely to accelerate this pathoadaptive process. Competition between clones carrying distinct beneficial mutations dominates the dynamics of the pathoadaptive process, as suggested from a mathematical model, which reproduces the observed experimental dynamics of E. coli evolution towards virulence. In conclusion, we reveal a molecular mechanism explaining how a specific component of host innate immunity can modulate microbial evolution towards pathogenicity.
Collapse
Affiliation(s)
| | - Ana Sousa
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | | | | | - Iris Caramalho
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
- Unidade de Imunologia Clínica, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Sara Magalhães
- Centro Biologia Ambiental, Faculdade de Ciências da Universidade de Lisboa, Lisboa, Portugal
| | | | - Isabel Gordo
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
- * E-mail:
| |
Collapse
|
17
|
Lukinskiene L, Liu Y, Reynolds SD, Steele C, Stripp BR, Leikauf GD, Kolls JK, Di YP. Antimicrobial activity of PLUNC protects against Pseudomonas aeruginosa infection. THE JOURNAL OF IMMUNOLOGY 2011; 187:382-90. [PMID: 21632717 DOI: 10.4049/jimmunol.1001769] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Epithelial antimicrobial activity may protect the lung against inhaled pathogens. The bactericidal/permeability-increasing protein family has demonstrated antimicrobial activity in vitro. PLUNC (palate, lung, and nasal epithelium associated) is a 25-kDa secreted protein that shares homology with bactericidal/permeability-increasing proteins and is expressed in nasopharyngeal and respiratory epithelium. The objective of this study was to determine whether PLUNC can limit Pseudomonas aeruginosa infection in mice. Transgenic mice (Scgb1a1-hPLUNC) were generated in which human PLUNC (hPLUNC) was directed to the airway epithelium with the Scgb1a1 promoter. The hPLUNC protein (hPLUNC) was detected in the epithelium throughout the trachea and bronchial airways and in bronchoalveolar lavage fluid. Bronchoalveolar lavage fluid from transgenic mice exhibited higher antibacterial activity than that from wild type littermates in vitro. After in vivo P. aeruginosa challenge, Scgb1a1-hPLUNC transgenic mice displayed enhanced bacterial clearance. This was accompanied by a decrease in neutrophil infiltration and cytokine levels. More importantly, the overexpressed hPLUNC in Scgb1a1-hPLUNC transgenic mouse airway significantly enhanced mouse survival against P. aeruginosa-induced respiratory infection. These data indicate that PLUNC is a novel antibacterial protein that likely plays a critical role in airway epithelium-mediated innate immune response.
Collapse
Affiliation(s)
- Lina Lukinskiene
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Li F, Wang W, Hu L, Li L, Yu J. Effect of Ambroxol on Pneumonia Caused by Pseudomonas aeruginosa with Biofilm Formation in an Endotracheal Intubation Rat Model. Chemotherapy 2011; 57:173-80. [DOI: 10.1159/000323622] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Accepted: 08/11/2010] [Indexed: 11/19/2022]
|
19
|
Zamorano L, Juan C, Fernández-Olmos A, Ge Y, Cantón R, Oliver A. Activity of the new cephalosporin CXA-101 (FR264205) against Pseudomonas aeruginosa isolates from chronically-infected cystic fibrosis patients. Clin Microbiol Infect 2010. [DOI: 10.1111/j.1469-0691.2010.03130.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
20
|
Hawdon NA, Aval PS, Barnes RJ, Gravelle SK, Rosengren J, Khan S, Ciofu O, Johansen HK, Høiby N, Ulanova M. Cellular responses of A549 alveolar epithelial cells to serially collected Pseudomonas aeruginosa from cystic fibrosis patients at different stages of pulmonary infection. ACTA ACUST UNITED AC 2010; 59:207-20. [PMID: 20528926 DOI: 10.1111/j.1574-695x.2010.00693.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Pseudomonas aeruginosa is the major cause of chronic pulmonary disease in cystic fibrosis (CF) patients. During chronic infection, P. aeruginosa lose certain virulence factors, transform into a mucoid phenotype, and develop antibiotic resistance. We hypothesized that these genetic and phenotypic alterations of P. aeruginosa affect the airway epithelial responses. A549 cells were infected with 27 well-characterized isolates of P. aeruginosa from CF patients obtained during longitudinal observation, or with P. aeruginosa mutant strains lacking flagella, pili, lipopolysaccharide, or pyocyanin. Pseudomonas aeruginosa isolates from the early stages of the infection exhibited high adherence to A549 cells, were readily internalized, and able to induce reactive oxygen species (ROS) production, apoptosis of infected cells, and the release of granulocyte macrophage colony-stimulating factor. Late P. aeruginosa isolates collected from patients with chronic lung infection were shown to have reduced adherence to and internalization into A549 cells compared with bacteria from patients with intermittent P. aeruginosa colonization, and induced lower production of ROS and apoptosis, but caused high proinflammatory cytokine and adhesion molecule expression. Our findings suggest that despite the loss of virulence factors during the adaptation process in the CF lung by late P. aeruginosa strains, they retain high proinflammatory abilities that likely contribute to the disease pathogenesis.
Collapse
|
21
|
IL-10 delivery by AAV5 vector attenuates inflammation in mice with Pseudomonas pneumonia. Gene Ther 2010; 17:567-76. [PMID: 20357828 DOI: 10.1038/gt.2010.28] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Lung infections with Pseudomonas aeruginosa and other pathogens in cystic fibrosis (CF) cause progressive airway obstruction and tissue damage, the predominant cause of morbidity and mortality in CF. We investigated whether a recombinant adeno-associated virus type 5 (AAV5) vector expressing murine interleukin (IL)-10 (AAV5.Cbeta-mIL-10), a regulatory/anti-inflammatory cytokine, could decrease airway inflammation in IL-10 knockout mice chronically infected with mucoid P. aeruginosa. Mice that received AAV5.Cbeta-mIL10 through intratracheal inoculation produced IL-10 at an average of 25 000 pg/ml in the epithelial lining fluid (ELF) and 12 000 pg/g-lung tissue 6 weeks post-vector delivery, significantly higher levels than in placebo-treated mice. At 3 days post-infection, proinflammatory cytokines (IL-1beta, tumor necrosis factor (TNF)-alpha, macrophage inhibitory protein (MIP)-1alpha and (KC) in the ELF and lung homogenate were decreased (1-9 folds) in the AAV5.Cbeta-mIL10-treated mice accompanied by less pronounced and more localized neutrophil infiltration in lung sections, when compared with placebo-treated mice. These results suggest that AAV5.Cbeta-mIL10 induces IL-10 levels in the lungs mediating a significant anti-inflammatory response and making AAV-IL-10 gene transfer a potentially useful therapy in the treatment of CF lung disease.
Collapse
|
22
|
Moser C, Van Gennip M, Bjarnsholt T, Jensen PØ, Lee B, Hougen HP, Calum H, Ciofu O, Givskov M, Molin S, Høiby N. Novel experimental Pseudomonas aeruginosa lung infection model mimicking long-term host-pathogen interactions in cystic fibrosis. APMIS 2009; 117:95-107. [PMID: 19239431 PMCID: PMC2774147 DOI: 10.1111/j.1600-0463.2008.00018.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The dominant cause of premature death in patients suffering from cystic fibrosis (CF) is chronic lung infection with Pseudomonas aeruginosa. The chronic lung infection often lasts for decades with just one clone. However, as a result of inflammation, antibiotic treatment and different niches in the lungs, the clone undergoes significant genetic changes, resulting in diversifying geno- and phenotypes. Such an adaptation may generate different host responses. To experimentally reflect the year-long chronic lung infection in CF, groups of BALB/c mice were infected with clonal isolates from different periods (1980, 1988, 1997, 1999 and 2003) of the chronic lung infection of one CF patient using the seaweed alginate embedment model. The results showed that the non-mucoid clones reduced their virulence over time, resulting in faster clearing of the bacteria from the lungs, improved pathology and reduced pulmonary production of macrophage inflammatory protein-2 (MIP-2) and granulocyte colony-stimulating factor (G-CSF). In contrast, the mucoid clones were more virulent and virulence increased with time, resulting in impaired pulmonary clearing of the latest clone, severe inflammation and increased pulmonary MIP-2 and G-CSF production. In conclusion, adaptation of P. aeruginosa in CF is reflected by changed ability to establish lung infection and results in distinct host responses to mucoid and non-mucoid phenotypes.
Collapse
Affiliation(s)
- Claus Moser
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Kukavica-Ibrulj I, Levesque RC. Animal models of chronic lung infection with Pseudomonas aeruginosa: useful tools for cystic fibrosis studies. Lab Anim 2008; 42:389-412. [PMID: 18782827 DOI: 10.1258/la.2007.06014e] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Cystic fibrosis (CF) is caused by a defect in the transmembrane conductance regulator (CFTR) protein that functions as a chloride channel. Dysfunction of the CFTR protein results in salty sweat, pancreatic insufficiency, intestinal obstruction, male infertility and severe pulmonary disease. In most patients with CF life expectancy is limited due to a progressive loss of functional lung tissue. Early in life a persistent neutrophylic inflammation can be demonstrated in the airways. The cause of this inflammation, the role of CFTR and the cause of lung morbidity by different CF-specific bacteria, mostly Pseudomonas aeruginosa, are not well understood. The lack of an appropriate animal model with multi-organ pathology having the characteristics of the human form of CF has hampered our understanding of the pathobiology and chronic lung infections of the disease for many years. This review summarizes the main characteristics of CF and focuses on several available animal models that have been frequently used in CF research. A better understanding of the chronic lung infection caused particularly by P. aeruginosa, the pathophysiology of lung inflammation and the pathogenesis of lung disease necessitates animal models to understand CF, and to develop and improve treatment.
Collapse
Affiliation(s)
- I Kukavica-Ibrulj
- Centre de Recherche sur la Fonction, Structure et Ingénierie des Protéines, Pavillon Charles-Eugène Marchand, Biologie Médicale, Faculté de Médecine, Université Laval, Québec G1K 7P4, Canada
| | | |
Collapse
|
24
|
Yu H, Buff SM, Baatz JE, Virella-Lowell I. Oral instillation with surfactant phospholipid: a reliable alternative to intratracheal injection in mouse studies. Lab Anim 2008; 42:294-304. [DOI: 10.1258/la.2007.007055] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The intratracheal (IT) injection technique has been widely used in the mouse studies of pulmonary diseases. Here, we describe a non-invasive technique using oral instillation challenge with the surfactant phospholipid that may advantageously replace the traditional IT technique. We performed comparative studies between oral instillation and IT injection of both vectors (adeno-associated virus, AAV vector) and bacteria ( Pseudomonas aeruginosa). Our results demonstrated that the oral instillation is a reliable alternative to IT injection. The administration of a fluorophore-labelled AAV vector demonstrated a similar pattern of distribution and quantity of vector delivered by oral instillation compared with IT injection. In addition, administration of AAV5-alpha-1 antitrypsin (AAT) to the lungs by oral instillation resulted in similar levels of AAT in both the lung homogenates and sera compared with the IT injection group. In our study of P. aeruginosa delivery, oral instillation resulted in similar mouse weight loss, cytokine levels in the epithelial lining fluid [interleukin (IL)-1β, IL-6, tumour necrosis factor-α, neutrophil chemokine and macrophage inflammatory protein-1α], lung histology/pathology and bacterial loads. Therefore, we conclude that oral instillation of materials mixed with surfactant phospholipid is an adequate and reproducible technique to replace the invasive IT injection procedure for the delivery of either vector or bacteria to the lungs. This procedure has the benefits of eliminating the discomfort, local inflammation and mortality associated with the more invasive IT surgical procedures.
Collapse
Affiliation(s)
| | - S M Buff
- Department of Pediatrics
- Department of Microbiology and Immunology
| | - J E Baatz
- Department of Pediatrics
- Department of Biochemistry and Molecular Biology, Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC 29403, USA
| | - I Virella-Lowell
- Department of Pediatrics
- Department of Microbiology and Immunology
| |
Collapse
|
25
|
|
26
|
Wilson KR, Napper JM, Denvir J, Sollars VE, Yu HD. Defect in early lung defence against Pseudomonas aeruginosa in DBA/2 mice is associated with acute inflammatory lung injury and reduced bactericidal activity in naive macrophages. MICROBIOLOGY (READING, ENGLAND) 2007; 153:968-979. [PMID: 17379707 PMCID: PMC2074882 DOI: 10.1099/mic.0.2006/002261-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that causes serious respiratory disease in the immune-compromised host. Using an aerosol infection model, 11 inbred mouse strains (129/Sv, A/J, BALB/c, C3H/HeN, C57BL/6, DBA/2, FVB, B10.D2/oSnJ, B10.D2/nSnJ, AKR/J and SWR/J) were tested for increased susceptibility to P. aeruginosa lung colonization. DBA/2 was the only mouse strain that had increased bacterial counts in the lung within 6 h post-infection. This deficiency incited a marked inflammatory response with reduced bacterial lung clearance and a mortality rate of 96.7 %. DBA/2 mice displayed progressive deterioration of lung pathology with extensive alveolar exudate and oedema formation at 48-72 h post-infection. The neutrophil-specific myeloperoxidase activity remained elevated throughout infection, suggesting that the increased leukocyte infiltration into alveoli caused acute inflammatory lung injury. DBA/2 mice lack the haemolytic complement; however, three additional mouse strains (AKR/J, SWR/J and A/J) with the same defect effectively cleared the infection, indicating that other host factors are involved in defence. Bone marrow-derived macrophages of DBA/2 showed an initial increase in phagocytosis, while their bactericidal activity was reduced compared to that of C57BL/6 macrophages. Comparison of pulmonary cytokine profiles of DBA/2 versus C57BL/6 or C3H/HeN indicated that DBA/2 had similar increases in tumour necrosis factor (TNF)-alpha, KC and interleukin (IL)-1a as C3H/HeN, but showed specific induction of IL-17, monocyte chemotactic protein (MCP)-1 and vascular endothelial growth factor (VEGF). Together, DBA/2 mice have a defect in the initial lung defence against P. aeruginosa colonization, which causes the host to produce a greater, but damaging, inflammatory response. Such a response may originate from the reduced antimicrobial activity of DBA/2 macrophages.
Collapse
Affiliation(s)
- Kari R. Wilson
- Department of Biochemistry and Microbiology, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25704-9330
| | - Jennifer M. Napper
- Department of Biochemistry and Microbiology, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25704-9330
| | - James Denvir
- Department of Biochemistry and Microbiology, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25704-9330
| | - Vincent E. Sollars
- Department of Biochemistry and Microbiology, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25704-9330
| | - Hongwei D. Yu
- Department of Biochemistry and Microbiology, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25704-9330
- Department of Pediatrics, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25704-9330
| |
Collapse
|
27
|
Jelsbak L, Johansen HK, Frost AL, Thøgersen R, Thomsen LE, Ciofu O, Yang L, Haagensen JAJ, Høiby N, Molin S. Molecular epidemiology and dynamics of Pseudomonas aeruginosa populations in lungs of cystic fibrosis patients. Infect Immun 2007; 75:2214-24. [PMID: 17261614 PMCID: PMC1865789 DOI: 10.1128/iai.01282-06] [Citation(s) in RCA: 174] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The ability to establish lifelong persistent infections is a fundamental aspect of the interactions between many pathogenic microorganisms and their mammalian hosts. One example is chronic lung infections by the opportunistic pathogen Pseudomonas aeruginosa in cystic fibrosis (CF) patients. This infection process is associated with extensive genetic adaptation and microevolution of the infecting bacteria. Through investigations of P. aeruginosa populations and infection dynamics in a group of CF patients followed at the Danish CF Clinic in Copenhagen, we have identified two distinct and dominant clones that have evolved into highly successful colonizers of CF patient airways. A significant component of the evolutionary success of these two clones has been their efficient transmissibility among the CF patients. The two clones have been present and transmitted among different CF patients for more than 2 decades. Our data also suggest that the P. aeruginosa population structure in the CF patient airways has been influenced by competition between different clones and that the two dominant clones have been particularly competitive within the lungs, which may add to their overall establishment success. In contrast, we show that adaptive traits commonly associated with establishment of chronic P. aeruginosa infections of CF patients, such as transition to the mucoid phenotype and production of virulence factors, play minor roles in the ability of the two dominant clones to spread among patients and cause long-term chronic infections. These findings suggest that hitherto-unrecognized evolutionary pathways may be involved in the development of successful and persistent P. aeruginosa colonizers of CF patient lungs.
Collapse
Affiliation(s)
- Lars Jelsbak
- Infection Microbiology Group, BioCentrum-DTU, Technical University of Denmark, Building 301, 2800 Lyngby, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
To understand the evolution of genetic diversity within species--bacterial and others--we must dissect the first steps of genetic adaptation to novel habitats, particularly habitats that are suboptimal for sustained growth where there is strong selection for adaptive changes. Here, we present the view that bacterial human pathogens represent an excellent model for understanding the molecular mechanisms of the adaptation of a species to alternative habitats. In particular, bacterial pathogens allow us to develop analytical methods to detect genetic adaptation using an evolutionary 'source-sink' model, with which the evolution of bacterial pathogens can be seen from the angle of continuous switching between permanent (source) and transient (sink) habitats. The source-sink model provides a conceptual framework for understanding the population dynamics and molecular mechanisms of virulence evolution.
Collapse
Affiliation(s)
- Evgeni V Sokurenko
- Department of Microbiology, University of Washington, Seattle, Washington 98105, USA.
| | | | | |
Collapse
|
29
|
Colasurdo GN, Fullmer JJ, Elidemir O, Atkins C, Khan AM, Stark JM. Respiratory syncytial virus infection in a murine model of cystic fibrosis. J Med Virol 2006; 78:651-8. [PMID: 16555274 DOI: 10.1002/jmv.20589] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Viral respiratory infections play an important role in the development and progression of pulmonary disease in cystic fibrosis (CF). The CF mouse model provides a tool to examine the relationship between the cystic fibrosis transmembrane conductance regulator (CFTR) defect and lung disease. This work investigates the cellular response to a common viral pathogen, respiratory syncytial virus (RSV) in the lung of CF mice. RSV was administered by intranasal inoculation of CFTR(tm1Unc)-Tg(FABPCFTR)1Jaw/J (CFTR-/-) and control mice. At day 5 post infection, viral titers, bronchoalveolar fluid nitrate levels (BALF) cell and differential counts, histology and studies on airway mechanics were performed. CFTR-/- mice had an impaired ability to clear RSV. This was associated with an exaggerated inflammatory response (increased lymphocytes and neutrophils) in BALF of RSV-infected CFTR-/- mice and a decreased ability to generate nitric oxide (NO) (measured as BAL nitrate). Lung histopathology of RSV-infected CFTR-/- mice demonstrated increased inflammation compared to RSV (-) CFTR-/- and control mice (regardless of RSV treatment). The airway response to methacholine was increased by RSV infection in CF mice when compared to controls. The CFTR-/- mouse exhibits an aberrant response to RSV infection. This model should be useful in providing further mechanistic information on the biology of respiratory viruses in mammalian models, and provide new insights into the pathogenesis of airway inflammation in patients with CF.
Collapse
Affiliation(s)
- Giuseppe N Colasurdo
- Department of Pediatrics, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | | | | | | | | | | |
Collapse
|
30
|
Bylund J, Burgess LA, Cescutti P, Ernst RK, Speert DP. Exopolysaccharides from Burkholderia cenocepacia inhibit neutrophil chemotaxis and scavenge reactive oxygen species. J Biol Chem 2005; 281:2526-32. [PMID: 16316987 DOI: 10.1074/jbc.m510692200] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Bacteria belonging to the Burkholderia cepacia complex are important opportunistic pathogens in compromised hosts, particularly patients with cystic fibrosis or chronic granulomatous disease. Isolates of B. cepacia complex may produce large amounts of exopolysaccharides (EPS) that endow the bacteria with a mucoid phenotype and appear to facilitate bacterial persistence during infection. We showed that EPS from a clinical B. cenocepacia isolate interfered with the function of human neutrophils in vitro; it inhibited chemotaxis and production of reactive oxygen species (ROS), both essential components of innate neutrophil-mediated host defenses. These inhibitory effects were not due to cytotoxicity or interference with intracellular calcium signaling. EPS also inhibited enzymatic generation of ROS in cell-free systems, indicating that it scavenges these bactericidal products. B. cenocepacia EPS is structurally distinct from Pseudomonas aeruginosa alginate, yet they share the capacity to scavenge ROS and inhibit chemotaxis. These properties could explain why the two bacterial species resist clearance from the infected cystic fibrosis lung.
Collapse
Affiliation(s)
- Johan Bylund
- Department of Pediatrics, University of British Columbia, Child and Family Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | | | | | | | | |
Collapse
|
31
|
Lee B, Haagensen JAJ, Ciofu O, Andersen JB, Høiby N, Molin S. Heterogeneity of biofilms formed by nonmucoid Pseudomonas aeruginosa isolates from patients with cystic fibrosis. J Clin Microbiol 2005; 43:5247-55. [PMID: 16207991 PMCID: PMC1248443 DOI: 10.1128/jcm.43.10.5247-5255.2005] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Biofilms are thought to play a key role in the occurrence of lung infections by Pseudomonas aeruginosa in patients with cystic fibrosis (CF). In this study, 20 nonmucoid P. aeruginosa isolates collected during different periods of chronic infection from eight CF patients were assessed with respect to phenotypic changes and in vitro biofilm formation. The physiological alterations were associated with a loss of motility (35% were nonmotile) and with decreased production of virulence factors (pyocyanin, proteases) and quorum-sensing molecules (45% of the isolates were unable to produce 3-O-C(12)-homoserine lactone quorum-sensing molecules). Compared with wild-type strain PAO 1, most P. aeruginosa isolates demonstrated different degrees of reduction of adherence on polystyrene surfaces. The in vitro biofilm formation of isolates was investigated in a hydrodynamic flow system. Confocal laser scanning microscope analysis showed that the biofilm structures of the P. aeruginosa isolates were highly variable in biomass and morphology. Biofilm development of six genotypically identical sequential isolates recovered from a particular patient at different time points of chronic infection (20 years) and after lung transplantation demonstrated significant changes in biofilm architectures. P. aeruginosa biofilm formation followed a trend of decreased adherence with progression of the chronic lung infection. The results suggest that the adherent characteristic of in vitro biofilm development was not essential for the longitudinal survival of nonmucoid P. aeruginosa during chronic lung colonization.
Collapse
Affiliation(s)
- Baoleri Lee
- Department of Clinical Microbiology, Rigshospitalet, Institute for Medical Microbiology and Immunology, University of Copenhagen, DK-2100 Copenhagen, Denmark.
| | | | | | | | | | | |
Collapse
|
32
|
Sedlak-Weinstein E, Cripps AW, Kyd JM, Foxwell AR. Pseudomonas aeruginosa: the potential to immunise against infection. Expert Opin Biol Ther 2005; 5:967-82. [PMID: 16018741 DOI: 10.1517/14712598.5.7.967] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Pseudomonas aeruginosa remains a serious pathogen for specific cohorts of patients where chronic infection is a poor prognostic indicator, such as those with cystic fibrosis, burn wounds or those who are immunocompromised. Significant disease burden is associated with a diverse spectrum of both nosocomial and community-acquired infections. To date, vaccines against P. aeruginosa have shown limited and often conflicting efficacy data, especially against heterologous strains, which are increasingly identified as co-colonisers of biofilms. While few studies have gone beyond Phase II clinical trials, a particular concern is the ability of P. aeruginosa to evade the immune system while provoking an immune response that contributes to the destructive nature of infection. Therefore, vaccine development needs to focus on preventing attachment and colonisation, as well as preventing conversion to a mucoid phenotype that is characteristic of the chronic condition that promotes pathology.
Collapse
Affiliation(s)
- E Sedlak-Weinstein
- Griffith University Gold Coast Campus, School of Medicine, PMB 50, Gold Coast Mail Centre, Queensland 9726, Australia
| | | | | | | |
Collapse
|
33
|
Hoffmann N, Rasmussen TB, Jensen PØ, Stub C, Hentzer M, Molin S, Ciofu O, Givskov M, Johansen HK, Høiby N. Novel mouse model of chronic Pseudomonas aeruginosa lung infection mimicking cystic fibrosis. Infect Immun 2005; 73:2504-14. [PMID: 15784597 PMCID: PMC1087399 DOI: 10.1128/iai.73.4.2504-2514.2005] [Citation(s) in RCA: 138] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pseudomonas aeruginosa causes a chronic infection in the lungs of cystic fibrosis (CF) patients by establishing an alginate-containing biofilm. The infection has been studied in several animal models; however, most of the models required artificial embedding of the bacteria. We present here a new pulmonary mouse model without artificial embedding. The model is based on a stable mucoid CF sputum isolate (NH57388A) with hyperproduction of alginate due to a deletion in mucA and functional N-acylhomoserine lactone (AHL)-based quorum-sensing systems. Chronic lung infection could be established in both CF mice (Cftr(tmlUnc-/-)) and BALB/c mice, as reflected by the detection of a high number of P. aeruginosa organisms in the lung homogenates at 7 days postinfection and alginate biofilms, surrounded by polymorphonuclear leukocytes in the alveoli. In comparison, both an AHL-producing nonmucoid revertant (NH57388C) from the mucoid isolate (NH57388A) and a nonmucoid isolate (NH57388B) deficient in AHL were almost cleared from the lungs of the mice. This model, in which P. aeruginosa is protected against the defense system of the lung by alginate, is similar to the clinical situation. Therefore, the mouse model provides an improved method for evaluating the interaction between mucoid P. aeruginosa, the host, and antibacterial therapy.
Collapse
Affiliation(s)
- Nadine Hoffmann
- Department of Bacteriology, Institute for Medical Microbiology and Immunology, Panum Institute, University of Copenhagen, Copenhagen DK-2200, Denmark.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Saadane A, Soltys J, Berger M. Role of IL-10 deficiency in excessive nuclear factor-κB activation and lung inflammation in cystic fibrosis transmembrane conductance regulator knockout mice. J Allergy Clin Immunol 2005; 115:405-11. [PMID: 15696103 DOI: 10.1016/j.jaci.2004.10.044] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND Patients with cystic fibrosis (CF) and CF transmembrane conductance regulator knockout (CF-KO) mice are deficient in pulmonary IL-10 and have excessive inflammatory response to Pseudomonas aeruginosa infection. OBJECTIVE We hypothesized that local IL-10 deficiency in the lung was responsible for prolonged and excessive inflammatory responses and observations of inflammation in the absence of infection. METHODS To determine whether IL-10 deficiency could account for persistent inflammation in CF mice independent of interactions of bacteria with epithelial cells, we challenged IL-10-knockout (IL-10-KO), CF-KO, and wild-type (WT) mice intratracheally with LPS and determined the effects of IL-10 replacement in CF-KO mice. RESULTS In response to LPS, IL-10-KO and CF-KO mice had more neutrophils and proinflammatory cytokines in bronchoalveolar lavage than WT mice. Both types of knockout mice had more profound and prolonged consumption of I-kappaB and increased activation of nuclear factor kappaB (NF-kappaB). Activated NF-kappaB persisted for 6 to 8 hours in CF-KO and IL-10-KO mice but was not detected beyond 2 hours in WT mice. IL-10 treatment of CF-KO mice attenuated the reduction in I-kappaBalpha and activation of NF-kappaB and reduced the excessive inflammation. CONCLUSION Similarities in the responses of CF-KO and IL-10-KO mice and correction of excessive responses in CF mice by exogenous IL-10 suggest that deficiency of IL-10 may be responsible for prolonged and excessive inflammatory responses in CF. Because LPS was used as the stimulus, these excessive responses are independent of any possible differences in the interactions of bacteria with CF epithelial cells.
Collapse
Affiliation(s)
- Aicha Saadane
- Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | | | | |
Collapse
|
35
|
Turkay C, Saba R, Sahin N, Pahin N, Altunbas H, Altunbap H, Ozbudak O, Akkaya B, Ozbilim G, Cölbasi I, Gölbapý Y, Turkay M, Ogünc D, Odünç D, Bayezid O. Effect of chronic Pseudomonas aeruginosa infection on the development of atherosclerosis in a rat model. Clin Microbiol Infect 2004; 10:705-8. [PMID: 15301672 DOI: 10.1111/j.1469-0691.2004.00920.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In order to investigate the possible relationship between atherosclerosis and chronic Pseudomonas aeruginosa infection, 66 Wistar rats were given five separate intratracheal inoculations of either P. aeruginosa or sterile saline at 4-week intervals. The rats were divided into four groups: group 1 was infected with P. aeruginosa and fed a diet containing cholesterol 1% w/v; group 2 was infected with P. aeruginosa and fed a normal diet; group 3 was not infected and was fed a diet containing cholesterol 1% w/v; and group 4 (the control group) was not infected and was fed a normal diet. One month after the final inoculation, the rats were killed humanely; computerised image analysis was used to evaluate sections of the aorta and heart, and the maximal wall thickness of the aorta and coronary artery. The aortic wall thickness was significantly greater for group 1 (329.53 +/- 58.06 microm) compared to groups 2 (190.59 +/- 27.81 microm; p < 0.0001), 3 (262.90 +/- 61.12 microm; p < 0.0004) and 4 (158.00 +/- 30.30 microm; p < 0.0001). Similarly, the coronary artery wall thickness was significantly greater for group 1 (72.96 +/- 10.67 microm) compared to groups 2 (35.07 +/- 8.53 microm; p < 0.0001), 3 (41.45 +/- 10.22 microm; p < 0.0001) and 4 (32.30 +/- 5.27 microm; p < 0.0001). These findings strengthen the hypothesis that chronic infection plays a role in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- C Turkay
- Department of Cardiovascular Surgery, Akdeniz University, Antalya, Turkey.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Cobb LM, Mychaleckyj JC, Wozniak DJ, López-Boado YS. Pseudomonas aeruginosaFlagellin and Alginate Elicit Very Distinct Gene Expression Patterns in Airway Epithelial Cells: Implications for Cystic Fibrosis Disease. THE JOURNAL OF IMMUNOLOGY 2004; 173:5659-70. [PMID: 15494517 DOI: 10.4049/jimmunol.173.9.5659] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Infection with the opportunistic pathogen Pseudomonas aeruginosa remains a major health concern. Two P. aeruginosa phenotypes relevant in human disease include motility and mucoidy. Motility is characterized by the presence of flagella and is essential in the establishment of acute infections, while mucoidy, defined by the production of the exopolysaccharide alginate, is critical in the development of chronic infections, such as the infections seen in cystic fibrosis patients. Indeed, chronic infection of the lung by mucoid P. aeruginosa is a major cause of morbidity and mortality in cystic fibrosis patients. We have used Calu-3 human airway epithelial cells to investigate global responses to infection with motile and mucoid P. aeruginosa. The response of airway epithelial cells to exposure to P. aeruginosa motile strains is characterized by a specific increase in gene expression in pathways controlling inflammation and host defense. By contrast, the response of airway epithelia to the stimuli presented by mucoid P. aeruginosa is not proinflammatory and, hence, may not be conducive to the effective elimination of the pathogen. The pattern of gene expression directed by flagellin, but not alginate, includes innate host defense genes, proinflammatory cytokines, and chemokines. By contrast, infection with alginate-producing P. aeruginosa results in an overall attenuation of host responses and an antiapoptotic effect.
Collapse
Affiliation(s)
- Laura M Cobb
- Department of Internal Medicine (Molecular Medicine), Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | |
Collapse
|
37
|
Stapper AP, Narasimhan G, Ohman DE, Barakat J, Hentzer M, Molin S, Kharazmi A, Høiby N, Mathee K. Alginate production affects Pseudomonas aeruginosa biofilm development and architecture, but is not essential for biofilm formation. J Med Microbiol 2004; 53:679-690. [PMID: 15184541 DOI: 10.1099/jmm.0.45539-0] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Extracellular polymers can facilitate the non-specific attachment of bacteria to surfaces and hold together developing biofilms. This study was undertaken to qualitatively and quantitatively compare the architecture of biofilms produced byPseudomonas aeruginosastrain PAO1 and its alginate-overproducing (mucA22) and alginate-defective (algD) variants in order to discern the role of alginate in biofilm formation. These strains, PAO1, Alg+PAOmucA22and Alg−PAOalgD, tagged with green fluorescent protein, were grown in a continuous flow cell system to characterize the developmental cycles of their biofilm formation using confocal laser scanning microscopy. Biofilm Image Processing (bip) and Community Statistics (comstat) software programs were used to provide quantitative measurements of the two-dimensional biofilm images. All three strains formed distinguishable biofilm architectures, indicating that the production of alginate is not critical for biofilm formation. Observation over a period of 5 days indicated a three-stage development pattern consisting of initiation, establishment and maturation. Furthermore, this study showed that phenotypically distinguishable biofilms can be quantitatively differentiated.
Collapse
Affiliation(s)
- Andres Plata Stapper
- Department of Biological Sciences1 and School of Computer Science2, Florida International University, Miami, FL 33199, USA 3Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA 4Section of Molecular Microbiology, The Technical University of Denmark, DK-2800 Lyngby, Denmark 5Department of Clinical Microbiology, University Hospital of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Giri Narasimhan
- Department of Biological Sciences1 and School of Computer Science2, Florida International University, Miami, FL 33199, USA 3Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA 4Section of Molecular Microbiology, The Technical University of Denmark, DK-2800 Lyngby, Denmark 5Department of Clinical Microbiology, University Hospital of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Dennis E Ohman
- Department of Biological Sciences1 and School of Computer Science2, Florida International University, Miami, FL 33199, USA 3Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA 4Section of Molecular Microbiology, The Technical University of Denmark, DK-2800 Lyngby, Denmark 5Department of Clinical Microbiology, University Hospital of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Johnny Barakat
- Department of Biological Sciences1 and School of Computer Science2, Florida International University, Miami, FL 33199, USA 3Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA 4Section of Molecular Microbiology, The Technical University of Denmark, DK-2800 Lyngby, Denmark 5Department of Clinical Microbiology, University Hospital of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Morten Hentzer
- Department of Biological Sciences1 and School of Computer Science2, Florida International University, Miami, FL 33199, USA 3Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA 4Section of Molecular Microbiology, The Technical University of Denmark, DK-2800 Lyngby, Denmark 5Department of Clinical Microbiology, University Hospital of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Søren Molin
- Department of Biological Sciences1 and School of Computer Science2, Florida International University, Miami, FL 33199, USA 3Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA 4Section of Molecular Microbiology, The Technical University of Denmark, DK-2800 Lyngby, Denmark 5Department of Clinical Microbiology, University Hospital of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Arsalan Kharazmi
- Department of Biological Sciences1 and School of Computer Science2, Florida International University, Miami, FL 33199, USA 3Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA 4Section of Molecular Microbiology, The Technical University of Denmark, DK-2800 Lyngby, Denmark 5Department of Clinical Microbiology, University Hospital of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Niels Høiby
- Department of Biological Sciences1 and School of Computer Science2, Florida International University, Miami, FL 33199, USA 3Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA 4Section of Molecular Microbiology, The Technical University of Denmark, DK-2800 Lyngby, Denmark 5Department of Clinical Microbiology, University Hospital of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Kalai Mathee
- Department of Biological Sciences1 and School of Computer Science2, Florida International University, Miami, FL 33199, USA 3Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA 4Section of Molecular Microbiology, The Technical University of Denmark, DK-2800 Lyngby, Denmark 5Department of Clinical Microbiology, University Hospital of Copenhagen, DK-2100 Copenhagen, Denmark
| |
Collapse
|
38
|
Head NE, Yu H. Cross-sectional analysis of clinical and environmental isolates of Pseudomonas aeruginosa: biofilm formation, virulence, and genome diversity. Infect Immun 2004; 72:133-44. [PMID: 14688090 PMCID: PMC343948 DOI: 10.1128/iai.72.1.133-144.2004] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Chronic lung infections with Pseudomonas aeruginosa biofilms are associated with refractory and fatal pneumonia in cystic fibrosis (CF). In this study, a group of genomically diverse P. aeruginosa isolates were compared with the reference strain PAO1 to assess the roles of motility, twitching, growth rate, and overproduction of a capsular polysaccharide (alginate) in biofilm formation. In an in vitro biofilm assay system, P. aeruginosa displayed strain-specific biofilm formation that was not solely dependent on these parameters. Compared with non-CF isolates, CF isolates expressed two opposing growth modes: reduced planktonic growth versus efficient biofilm formation. Planktonic cells of CF isolates showed elevated sensitivity to hydrogen peroxide, a reactive oxygen intermediate, and decreased lung colonization in an aerosol infection mouse model. Despite having identical genomic profiles, CF sequential isolates produced different amounts of biofilm. While P. aeruginosa isolates exhibited genomic diversity, the genome size of these isolates was estimated to be 0.4 to 19% (27 to 1,184 kb) larger than that of PAO1. To identify these extra genetic materials, random amplification of polymorphic DNA was coupled with PAO1-subtractive hybridization. Three loci were found within the genomes of two CF isolates encoding one novel homolog involved in retaining a Shigella virulence plasmid (mvpTA) and two divergent genes that function in removing negative supercoiling (topA) and biosynthesis of pyoverdine (PA2402). Together, P. aeruginosa biodiversity could provide one cause for the variation of morbidity and mortality in CF. P. aeruginosa may possess undefined biofilm adhesins that are important to the development of an antibiofilm therapeutic target.
Collapse
Affiliation(s)
- Nathan E Head
- Department of Microbiology, Immunology and Molecular Genetics, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia 25704-9330, USA
| | | |
Collapse
|
39
|
Song Z, Wu H, Ciofu O, Kong KF, Høiby N, Rygaard J, Kharazmi A, Mathee K. Pseudomonas aeruginosa alginate is refractory to Th1 immune response and impedes host immune clearance in a mouse model of acute lung infection. J Med Microbiol 2003; 52:731-740. [PMID: 12909647 DOI: 10.1099/jmm.0.05122-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic respiratory pathogen that accounts for most of the morbidity and mortality in cystic fibrosis (CF) patients. In CF-affected lungs, the bacteria undergo conversion from a non-mucoid to a non-tractable mucoid phenotype, due to overproduction of alginate. The effect of alginate production on pathogenicity was investigated by using an acute lung infection mouse model that compared a non-mucoid P. aeruginosa strain, PAO1, to its constitutive alginate-overproducing derivative, Alg(+) PAOmucA22, and an alginate-defective strain, Alg(-) PAOalgD. Bacterial suspensions were instilled into the left bronchus and examined 24 and 48 h post-infection. The highest bacterial loads and the most severe lung pathology were observed with strain Alg(-) PAOalgD at 24 h post-infection, which may have been due to an increase in expression of bacterial elastase by the mutant. Significantly lower lung and spleen bacterial loads were found in the two non-mucoid (PAO1 and Alg(-) PAOalgD) groups, compared to the mucoid Alg(+) PAOmucA22 group, between 24 and 48 h post-infection. The positive correlation between lung bacteriology and lung macroscopic pathology in the Alg(+) PAOmucA22 group suggests that alginate production not only impedes pulmonary clearing, but also results in severe lung damage. Positive correlations between IL12 levels and lung macroscopic pathology, and between IL12 and IFN-gamma levels in the Alg(+) PAOmucA22 group, suggested a possible contribution of these pro-inflammatory cytokines to tissue damage. No significant differences were found between the three groups in lung cytokine responses at 24 or 48 h post-infection. However, on comparison within each group at 24 and 48 h post-infection, a significant increase in the pro-inflammatory cytokine IFN-gamma was observed. Higher ratios of IFN-gamma/IL4 and IFN-gamma/IL10, but lower IL10 levels, were also found in all three groups. These results indicate a Th1-predominated immune response in these animals. Such cytokine responses could have aided the clearance of non-mucoid P. aeruginosa, but were not sufficient to alleviate infection by the mucoid variants. Alginate production may promote survival and persistence of this pathogenic micro-organism in the lung.
Collapse
Affiliation(s)
- Zhijun Song
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA 2Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark 3Bartholin Institute, Kommunehospitalet, Copenhagen, Denmark
| | - Hong Wu
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA 2Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark 3Bartholin Institute, Kommunehospitalet, Copenhagen, Denmark
| | - Oana Ciofu
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA 2Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark 3Bartholin Institute, Kommunehospitalet, Copenhagen, Denmark
| | - Kok-Fai Kong
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA 2Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark 3Bartholin Institute, Kommunehospitalet, Copenhagen, Denmark
| | - Niels Høiby
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA 2Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark 3Bartholin Institute, Kommunehospitalet, Copenhagen, Denmark
| | - Jørgen Rygaard
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA 2Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark 3Bartholin Institute, Kommunehospitalet, Copenhagen, Denmark
| | - Arsalan Kharazmi
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA 2Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark 3Bartholin Institute, Kommunehospitalet, Copenhagen, Denmark
| | - Kalai Mathee
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA 2Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark 3Bartholin Institute, Kommunehospitalet, Copenhagen, Denmark
| |
Collapse
|
40
|
Guilbault C, Stotland P, Lachance C, Tam M, Keller A, Thompson-Snipes L, Cowley E, Hamilton TA, Eidelman DH, Stevenson MM, Radzioch D. Influence of gender and interleukin-10 deficiency on the inflammatory response during lung infection with Pseudomonas aeruginosa in mice. Immunology 2002; 107:297-305. [PMID: 12423305 PMCID: PMC1782799 DOI: 10.1046/j.1365-2567.2002.01508.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cystic fibrosis females have a worse prognosis compared to male patients. Furthermore, cystic fibrosis patients infected with Pseudomonas aeruginosa have been shown to have dysregulated cytokine profiles, as higher levels of tumour necrosis factor alpha (TNF-alpha), interleukin (IL)-8, and lower levels of IL-10 are found in the bronchoalveolar lavage fluid compared to healthy controls. The present study was aimed at investigating the importance of gender and IL-10 in the susceptibility of C57BL/6 mice to pulmonary infection with Pseudomonas aeruginosa. We found that wildtype females were more susceptible than males to infection, as we observed greater weight loss, higher bacterial load, and inflammatory mediators in their lungs. IL-10 knockout mice, both females and males, had higher levels of TNF-alpha in the lungs compared to wildtype mice and maintained higher levels of polymorphonuclear cells and lower levels of macrophages for a longer period of time. Our results demonstrate that the number of bacteria recovered from the lungs of IL-10 knockout male mice was significantly higher than that observed in their wildtype male counterparts and we show that neutralization of IL-10 in infected female mice for a prolonged period of time leads to increased susceptibility to infection. Results reported in this study clearly demonstrate that females, both wildtype and IL-10 knockout mice are more susceptible to Pseudomonas aeruginosa infection than males, and that they mount a stronger inflammatory response in the lungs.
Collapse
Affiliation(s)
- Claudine Guilbault
- Departments of Experimental Medicine and Urology, McGill University Health Center, Montreal General Hospital Research Institute, 1650 Cedar Avenue, Montreal, Quebec H3G 1A4, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Lizewski SE, Lundberg DS, Schurr MJ. The transcriptional regulator AlgR is essential for Pseudomonas aeruginosa pathogenesis. Infect Immun 2002; 70:6083-93. [PMID: 12379685 PMCID: PMC130412 DOI: 10.1128/iai.70.11.6083-6093.2002] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Chronic Pseudomonas aeruginosa lung infection is the major cause of morbidity and mortality in cystic fibrosis (CF) patients. One P. aeruginosa virulence factor unique to CF isolates is overproduction of alginate, phenotypically termed mucoidy. Mucoidy is the result of increased transcription from the algD gene and is activated by the transcriptional regulator AlgR. Mutations in algR result in a nonmucoid phenotype and loss of twitching motility. Additionally, AlgR controls transcription of algC, encoding a dual-function enzyme necessary for both lipopolysaccharide (LPS) and alginate production. Therefore, to determine the effect of algR on P. aeruginosa virulence, an algR mutant was examined for sensitivity to reactive oxygen intermediates, killing by phagocytes, systemic virulence, and the ability to maintain a murine lung infection. We found that P. aeruginosa PAO700 (algR::Gm(r)) was less lethal than PAO1, as tested in an acute septicemia infection mouse model, and was cleared more efficiently in a mouse pneumonia model. Additionally, the algR mutant (PAO700) was more sensitive to hypochlorite. However, PAO700 was more resistant to hydrogen peroxide and killed less readily in an acellular myeloperoxidase assay than PAO1. There was little difference in killing between PAO1 and PAO700 with macrophage-like J774 cells and human polymorhonuclear leukocytes. Two-dimensional gel analysis of P. aeruginosa algR mutant and wild-type protein extracts revealed 47 differentially regulated proteins, suggesting that AlgR plays both a positive role and a negative role in gene expression. Together, these results imply that AlgR is necessary for virulence and regulates genes in addition to the genes associated with alginate and LPS production and pilus function.
Collapse
Affiliation(s)
- Stephen E Lizewski
- Department of Microbiology and Immunology, Program in Molecular Pathogenesis and Immunity, Tulane University Health Sciences Center, New Orleans, Louisiana 70112-2699, USA
| | | | | |
Collapse
|
42
|
Fullner KJ, Boucher JC, Hanes MA, Haines GK, Meehan BM, Walchle C, Sansonetti PJ, Mekalanos JJ. The contribution of accessory toxins of Vibrio cholerae O1 El Tor to the proinflammatory response in a murine pulmonary cholera model. J Exp Med 2002; 195:1455-62. [PMID: 12045243 PMCID: PMC2193536 DOI: 10.1084/jem.20020318] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The contribution of accessory toxins to the acute inflammatory response to Vibrio cholerae was assessed in a murine pulmonary model. Intranasal administration of an El Tor O1 V. cholerae strain deleted of cholera toxin genes (ctxAB) caused diffuse pneumonia characterized by infiltration of PMNs, tissue damage, and hemorrhage. By contrast, the ctxAB mutant with an additional deletion in the actin-cross-linking repeats-in-toxin (RTX) toxin gene (rtxA) caused a less severe pathology and decreased serum levels of proinflammatory molecules interleukin (IL)-6 and murine macrophage inflammatory protein (MIP)-2. These data suggest that the RTX toxin contributes to the severity of acute inflammatory responses. Deletions within the genes for either hemagglutinin/protease (hapA) or hemolysin (hlyA) did not significantly affect virulence in this model. Compound deletion of ctxAB, hlyA, hapA, and rtxA created strain KFV101, which colonized the lung but induced pulmonary disease with limited inflammation and significantly reduced serum titers of IL-6 and MIP-2. 100% of mice inoculated with KFV101 survive, compared with 20% of mice inoculated with the ctxAB mutant. Thus, the reduced virulence of KFV101 makes it a prototype for multi-toxin deleted vaccine strains that could be used for protection against V. cholerae without the adverse effects of the accessory cholera toxins.
Collapse
Affiliation(s)
- Karla Jean Fullner
- Departments of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave., Morton 6-626, Chicago, IL 60611, USA.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Haug T, Søstrand P, Langård S. Exposure to culturable microorganisms in paper mills and presence of symptoms associated with infections. Am J Ind Med 2002; 41:498-505. [PMID: 12173375 DOI: 10.1002/ajim.10065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Previous studies of paper machine operators have to a large extent focused on endotoxins as a possible health hazard, but not culturable micro-organisms (MOs). METHODS Based on exposure assessment in 11 paper mills workers exposed to culturable bio-aerosols were grouped in three exposure groups. 781 exposed and 285 unexposed workers completed a questionnaire that provided data pertaining to infections and associated symptoms. RESULTS Concentrations of culturable bacteria in process waters varied in the range 10(4)-10(6) colony forming units (cfu)/ml, and in bio-aerosols concentrations varied typically in the range 10(4)-->10(5) cfu/m3. Operators exposed to bio-aerosols reported higher cumulative incidence of symptoms associated with infections compared to the reference population (ORs = 1.7-5.9), and the group of highest exposed workers reported higher cumulative incidence than the group of lowest exposed (ORs = 1.2-3.6). CONCLUSION Exposure to bio-aerosols containing culturable MOs may induce symptoms associated with infections among operators in paper mills.
Collapse
Affiliation(s)
- Terje Haug
- Center for Occupational and Environmental Medicine, National Hospital, Oslo, N-0027 Norway.
| | | | | |
Collapse
|
44
|
Chmiel JF, Konstan MW, Saadane A, Krenicky JE, Lester Kirchner H, Berger M. Prolonged inflammatory response to acute Pseudomonas challenge in interleukin-10 knockout mice. Am J Respir Crit Care Med 2002; 165:1176-81. [PMID: 11956064 DOI: 10.1164/ajrccm.165.8.2107051] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Cystic fibrosis (CF) lung disease is characterized by a neutrophilic infiltrate that is excessive relative to the burden of infection. Decreased interleukin-10 in CF airways may impair proper termination of inflammation, leading to persistence of neutrophils after acute infections have been cleared. This could explain reports of lung inflammation in the absence of bacteria in infants with CF. We evaluated the kinetics of inflammation after transient Pseudomonas aeruginosa challenge in IL-10 knockout (KO) and wild-type (WT) mice. Both types of mice cleared the infection by Day 6 (p > or = 0.29). However, IL-10 KO mice had more neutrophils in bronchoalveolar lavage fluid than did WT mice on Days 4 (p < 0.0001), 6 (p < 0.0001), and 8 (p = 0.042). IL-10 KO mice had high concentrations of proinflammatory cytokines in BAL on Days 2 and 4, with some cytokines detectable on Days 6 and 8, whereas cytokines in BAL from WT mice were greatest on Day 2 and undetectable by Day 4. Moreover, IL-10 KO mice failed to regenerate IkappaBalpha once degraded and subsequently had prolonged activation of NF-kappaB. These data suggest that IL-10 deficiency contributes to prolonged inflammatory responses early in CF, when infection may be transient.
Collapse
Affiliation(s)
- James F Chmiel
- Department of Pediatrics, Rainbow Babies and Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Soltys J, Bonfield T, Chmiel J, Berger M. Functional IL-10 deficiency in the lung of cystic fibrosis (cftr(-/-)) and IL-10 knockout mice causes increased expression and function of B7 costimulatory molecules on alveolar macrophages. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:1903-10. [PMID: 11823525 DOI: 10.4049/jimmunol.168.4.1903] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Alveolar macrophages are poor APCs that only minimally express B7 costimulatory molecules. Because our previous data suggest that bronchial epithelial cells constitutively secrete IL-10, and IL-10 inhibits B7 expression in vitro, we hypothesized that this IL-10 is responsible for suppressing B7 expression on macrophages that enter the airways. Furthermore, because we have shown that cystic fibrosis (CF) lungs are deficient in IL-10, we hypothesized that bronchoalveolar macrophages (BALMs) from cystic fibrosis transmembrane conductance regulator (CFTR)(-/-) as well as IL-10(-/-) mice might express increased B7. Immunofluorescence for B7 was positive on BALMs from CF patients and CFTR(-/-) and IL-10(-/-) mice, but was negative on controls. FACS showed that 63.9% of BALMs from IL-10(-/-) mice were B7-1 positive, as were 67.4% of BALMs from CFTR(-/-) mice, whereas <7% of BALMs from wild-type controls were positive. Using BALMs to costimulate splenic T cells with anti-CD3 as a mitogen showed 9202 +/- 2107 cpm [(3)H]thymidine incorporation for BALMs from IL-10(-/-) mice and 4082 +/- 1036 cpm for BALMs from CFTR(-/-) mice, but <200 cpm with BALMs from either type of +/+ mouse. Treatment of CFTR(-/-) mice with recombinant mouse IL-10 reduced the B7 expression and costimulatory activity of the BALMs. These data suggest that the IL-10 secreted in the healthy lung may be responsible for the absence of B7 and poor costimulatory activity of BALMs and that reductions of pulmonary IL-10 in CF may enhance B7 expression and local immune responses.
Collapse
Affiliation(s)
- Jindrich Soltys
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | |
Collapse
|
46
|
Firoved AM, Boucher JC, Deretic V. Global genomic analysis of AlgU (sigma(E))-dependent promoters (sigmulon) in Pseudomonas aeruginosa and implications for inflammatory processes in cystic fibrosis. J Bacteriol 2002; 184:1057-64. [PMID: 11807066 PMCID: PMC134789 DOI: 10.1128/jb.184.4.1057-1064.2002] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The conversion of Pseudomonas aeruginosa to the mucoid phenotype coincides with the establishment of chronic respiratory infections in cystic fibrosis (CF). A major pathway of conversion to mucoidy in clinical strains of P. aeruginosa is dependent upon activation of the alternative sigma factor AlgU (P. aeruginosa sigma(E)). Here we initiated studies of AlgU-dependent global expression patterns in P. aeruginosa in order to assess whether additional genes, other than those involved in the production of the mucoid exopolysaccharide alginate, are turned on during conversion to mucoidy. Using genomic information and the consensus AlgU promoter sequence, we identified 35 potential AlgU (sigma(E)) promoter sites on the P. aeruginosa chromosome. Each candidate promoter was individually tested by reverse transcription and mRNA 5'-end mapping using RNA isolated from algU(+) and algU::Tc(r) mutant cells. A total of 10 new AlgU-dependent promoters were identified, and the corresponding mRNA start sites were mapped. Two of the 10 newly identified AlgU promoters were upstream of predicted lipoprotein genes. Since bacterial lipoproteins have been implicated as inducers of inflammatory pathways, we tested whether lipopeptides corresponding to the products of the newly identified AlgU-dependent lipoprotein genes, lptA and lptB, had proinflammatory activity. In human peripheral blood monocyte-derived macrophages the peptides caused production of interleukin-8, a proinflammatory chemokine typically present at excessively high levels in the CF lung. Our studies show how genomic information can be used to uncover on a global scale the genes controlled by a given sigma factor (collectively termed here sigmulon) using conventional molecular tools. In addition, our data suggest the existence of a previously unknown connection between conversion to mucoidy and expression of lipoproteins with potential proinflammatory activity. This link may be of significance for infections and inflammatory processes in CF.
Collapse
Affiliation(s)
- Aaron M Firoved
- Department of Microbiology and Immunology, University of Michigan Medical School Ann Arbor, Michigan 48109-0620, USA
| | | | | |
Collapse
|
47
|
Sokurenko EV, Tchesnokova V, Yeung AT, Oleykowski CA, Trintchina E, Hughes KT, Rashid RA, Brint JM, Moseley SL, Lory S. Detection of simple mutations and polymorphisms in large genomic regions. Nucleic Acids Res 2001; 29:E111. [PMID: 11713331 PMCID: PMC92577 DOI: 10.1093/nar/29.22.e111] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2001] [Revised: 09/19/2001] [Accepted: 09/19/2001] [Indexed: 11/14/2022] Open
Abstract
We have developed a novel technology that makes it possible to detect simple nucleotide polymorphisms directly within a sample of total genomic DNA. It allows, in a single Southern blot experiment, the determination of sequence identity of genomic regions with a combined length of hundreds of kilobases. This technology does not require PCR amplification of the target DNA regions, but exploits preparative size-fractionation of restriction-digested genomic DNA and a newly discovered property of the mismatch-specific endonuclease CEL I to cleave heteroduplex DNA with a very high specificity and sensitivity. We have used this technique to detect various simple mutations directly in the genomic DNA of isogenic pairs of recombinant Pseudomonas aeruginosa, Escherichia coli and Salmonella isolates. Also, by using a cosmid DNA library and genomic fractions as hybridization probes, we have compared total genomic DNA of two clinical P.aeruginosa clones isolated from the same patient, but exhibiting divergent phenotypes. The mutation scan correctly detected a GA insertion in the quorum-sensing regulator gene rhlR and, in addition, identified a novel intragenomic polymorphism in rrn operons, indicating very high stability of the bacterial genomes under natural non-mutator conditions.
Collapse
Affiliation(s)
- E V Sokurenko
- Department of Microbiology, University of Washington, Box 357242, Seattle, WA 98195, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Eggert FM, McLeod MH, Flowerdew G. Effects of smoking and treatment status on periodontal bacteria: evidence that smoking influences control of periodontal bacteria at the mucosal surface of the gingival crevice. J Periodontol 2001; 72:1210-20. [PMID: 11577953 DOI: 10.1902/jop.2000.72.9.1210] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND We examined whether smoking status could influence growth of potentially pathogenic bacteria in the periodontal environment of treated and untreated periodontal patients. METHODS We have previously reported effects of treatment status on marker bacteria in our patients. We established a history of any smoking during 6 months prior to microbiological sampling (F-ME, 16 smokers out of 64; MHM, 70 smokers out of 185). We used a commercial immunoassay to quantitate Porphyromonas gingivalis, Prevotella intermedia, and Actinobacillus actinomycetemcomitans in paper point samples from periodontal sites. RESULTS Logistic regression showed that in smokers, neither P. gingivalis nor A. actinomycetemcomitans was quantitatively increased, while P intermedia was somewhat increased. Multiple regression demonstrated that smoking disrupts the positive relationship between increasing probing depth and increasing bacterial growth that is found in non-smokers. In smokers, growth of marker bacteria at shallow sites (< or =5 mm) was significantly increased to the levels found at deeper sites (>5 mm) in both smokers and non-smokers. Supragingival plaque biofilm was identified as a reservoir for marker bacteria; smokers and nonsmokers had equal ranges of oral cleanliness. CONCLUSIONS Smoking-associated periodontitis is not simply a reflection of oral cleanliness. Smoking extends a favorable habitat for bacteria such as P. gingivalis, P. intermedia, and A. actinomycetemcomitans to shallow sites (< or =5 mm). Molecular byproducts of smoking interfere with mechanisms that normally contain growth of damaging bacteria at the surface of the oral mucosa in gingival crevices. In this way, smoking can promote early development of periodontal lesions.
Collapse
Affiliation(s)
- F M Eggert
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton.
| | | | | |
Collapse
|
49
|
Yorgey P, Rahme LG, Tan MW, Ausubel FM. The roles of mucD and alginate in the virulence of Pseudomonas aeruginosa in plants, nematodes and mice. Mol Microbiol 2001; 41:1063-76. [PMID: 11555287 DOI: 10.1046/j.1365-2958.2001.02580.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We are exploiting the broad host range of the human opportunistic pathogen Pseudomonas aeruginosa strain PA14 to elucidate the molecular basis of bacterial virulence in plants, nematodes, insects and mice. In this report, we characterize the role that two PA14 gene products, MucD and AlgD, play in virulence. MucD is orthologous to the Escherichia coli periplasmic protease and chaperone DegP. DegP homologues are known virulence factors that play a protective role in stress responses in various species. AlgD is an enzyme involved in the biosynthesis of the exopolysaccharide alginate, which is hyperinduced in mucD mutants. A PA14 mucD mutant was significantly impaired in its ability to cause disease in Arabidopsis thaliana and mice and to kill the nematode Caenorhabditis elegans. Moreover, MucD was found to be required for the production of an extracellular toxin involved in C. elegans killing. In contrast, a PA14 algD mutant was not impaired in virulence in plants, nematodes or mice. A mucDalgD double mutant had the same phenotype as the mucD single mutant in the plant and nematode pathogenesis models. However, the mucDalgD double mutant was synergistically reduced in virulence in mice, suggesting that alginate can partially compensate for the loss of MucD function in mouse pathogenesis.
Collapse
Affiliation(s)
- P Yorgey
- Department of Genetics, Harvard Medical School, Boston, MA 02114, USA
| | | | | | | |
Collapse
|
50
|
Poschet JF, Boucher JC, Firoved AM, Deretic V. Conversion to mucoidy in Pseudomonas aeruginosa infecting cystic fibrosis patients. Methods Enzymol 2001; 336:65-76. [PMID: 11398420 DOI: 10.1016/s0076-6879(01)36579-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- J F Poschet
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | | | | | | |
Collapse
|