1
|
Fallah A, Sedighian H, Behzadi E, Havaei SA, Kachuei R, Imani Fooladi AA. The role of serum circulating microbial toxins in severity and cytokine storm of COVID positive patients. Microb Pathog 2023; 174:105888. [PMID: 36402345 PMCID: PMC9671676 DOI: 10.1016/j.micpath.2022.105888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 11/13/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
Abstract
The emergence of Coronavirus disease 2019 (Covid-19) is a global problem nowadays, causing health difficulty with increasing mortality rates, which doesn't have a verified treatment. SARS-CoV-2 infection has various pathological and epidemiological characteristics, one of them is increased amounts of cytokine production, which in order activate an abnormal unrestricted response called "cytokine storm". This event contributes to severe acute respiratory distress syndrome (ARDS), which results in respiratory failure and pneumonia and is the great cause of death associated with Covid-19. Endotoxemia and the release of bacterial lipopolysaccharides (endotoxins) from the lumen into the bloodstream enhance proinflammatory cytokines. SARS-CoV-2 can straightly interplay with endotoxins via its S protein, leading to the extremely elevating release of cytokines and consequently increase the harshness of Covid-19. In this review, we will discuss the possible role of viral-bacterial interaction that occurs through the transfer of bacterial products such as lipopolysaccharide (LPS) from the intestine into the bloodstream, exacerbating the severity of Covid-19 and cytokine storms.
Collapse
Affiliation(s)
- Arezoo Fallah
- Department of Bacteriology and Virology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Sedighian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Elham Behzadi
- Academy of Medical Sciences of the I.R. of Iran, Tehran, Iran
| | - Seyed Asghar Havaei
- Department of Microbiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Kachuei
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran,Corresponding author. 14359-44711, Tehran, Iran
| |
Collapse
|
2
|
Ha J, Kim S, Lee J, Lee H, Choi Y, Oh H, Yoon Y, Choi KH. The role of Pseudomonas aeruginosa DesB in pathogen-host interaction. Int Microbiol 2020; 23:549-555. [PMID: 32323095 DOI: 10.1007/s10123-020-00130-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/19/2020] [Accepted: 04/13/2020] [Indexed: 11/25/2022]
Abstract
Pseudomonas aeruginosa, commonly found in environments, can cause chronic lung disease in immunocompromised patients. In previous study, an aerobic desaturase (DesB) in P. aeruginosa exerted considerable effects on virulence factor production. The objective of this study was to analyze the role of DesB on the virulence traits of P. aeruginosa in the host. For the in vitro experiments, cells and supernatants from wild-type (WT) P. aeruginosa and its desB mutant were collected. The diluted cells were added to the A549 cell monolayer in order to determine cell viability, invasion ability, and/or immune response. For the in vivo experiments, 6-week-old ICR mice were infected with 6-7 log CFU bacterial cells using endotracheal intubation. The ratio of lung weight to body weight and survival rate of each bacterial strain in the lung were measured. The histopathology of lung tissue was also studied. desB mutants exhibited lower cytotoxicity in A549 cells. In addition, more pro-inflammatory cytokines and chemokines were present in desB mutant-treated. In the lungs of mouse model, WT survived longer than desB mutant, and the WT migrated from the lung to the liver and spleen. The results suggest that P. aeruginosa DesB affects the pathogenicity of the organism in the host.
Collapse
Affiliation(s)
- Jimyeong Ha
- Risk Analysis Research Center, Sookmyung Women's University, Seoul, 04310, South Korea
| | - Sejeong Kim
- Risk Analysis Research Center, Sookmyung Women's University, Seoul, 04310, South Korea
| | - Jeeyeon Lee
- Department of Food and Nutrition, Dong-eui University, Busan, 47340, South Korea
| | - Heeyoung Lee
- Food Standard Research Center, Korea Food Research Institute, Wanju, Jeollabuk-do, 55365, South Korea
| | - Yukyung Choi
- Department of Food and Nutrition, Sookmyung Women's University, Seoul, 04310, South Korea
| | - Hyemin Oh
- Department of Food and Nutrition, Sookmyung Women's University, Seoul, 04310, South Korea
| | - Yohan Yoon
- Risk Analysis Research Center, Sookmyung Women's University, Seoul, 04310, South Korea
- Department of Food and Nutrition, Sookmyung Women's University, Seoul, 04310, South Korea
| | - Kyoung-Hee Choi
- Department of Oral Microbiology, College of Dentistry, Wonkwang University, Iksan, 54538, South Korea.
| |
Collapse
|
3
|
Liang X, Gupta K, Quintero JR, Cernadas M, Kobzik L, Christou H, Pier GB, Owen CA, Çataltepe S. Macrophage FABP4 is required for neutrophil recruitment and bacterial clearance in Pseudomonas aeruginosa pneumonia. FASEB J 2019; 33:3562-3574. [PMID: 30462529 PMCID: PMC6988858 DOI: 10.1096/fj.201802002r] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 10/15/2018] [Indexed: 01/29/2023]
Abstract
Fatty acid binding protein 4 (FABP4), an intracellular lipid chaperone and adipokine, is expressed by lung macrophages, but the function of macrophage-FABP4 remains elusive. We investigated the role of FABP4 in host defense in a murine model of Pseudomonas aeruginosa pneumonia. Compared with wild-type (WT) mice, FABP4-deficient (FABP4-/-) mice exhibited decreased bacterial clearance and increased mortality when challenged intranasally with P. aeruginosa. These findings in FABP4-/- mice were associated with a delayed neutrophil recruitment into the lungs and were followed by greater acute lung injury and inflammation. Among leukocytes, only macrophages expressed FABP4 in WT mice with P. aeruginosa pneumonia. Chimeric FABP4-/- mice with WT bone marrow were protected from increased mortality seen in chimeric WT mice with FABP4-/- bone marrow during P. aeruginosa pneumonia, thus confirming the role of macrophages as the main source of protective FABP4 against that infection. There was less production of C-X-C motif chemokine ligand 1 (CXCL1) in FABP4-/- alveolar macrophages and lower airway CXCL1 levels in FABP4-/- mice. Delivering recombinant CXCL1 to the airways protected FABP4-/- mice from increased susceptibility to P. aeruginosa pneumonia. Thus, macrophage-FABP4 has a novel role in pulmonary host defense against P. aeruginosa infection by facilitating crosstalk between macrophages and neutrophils via regulation of macrophage CXCL1 production.-Liang, X., Gupta, K., Rojas Quintero, J., Cernadas, M., Kobzik, L., Christou, H., Pier, G. B., Owen, C. A., Çataltepe, S. Macrophage FABP4 is required for neutrophil recruitment and bacterial clearance in Pseudomonas aeruginosa pneumonia.
Collapse
Affiliation(s)
- Xiaoliang Liang
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kushagra Gupta
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Joselyn Rojas Quintero
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Manuela Cernadas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lester Kobzik
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Helen Christou
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gerald B. Pier
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA; and
| | - Caroline A. Owen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- The Lovelace Respiratory Research Institute, Albuquerque, New Mexico, USA
| | - Sule Çataltepe
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Pompilio A, Crocetta V, Di Bonaventura G. Stenotrophomonas maltophilia mutant lacking flagella remains virulent in DBA/2N mice but is less efficient in stimulating TNF-α expression. FEMS Microbiol Lett 2018; 365:5075579. [DOI: 10.1093/femsle/fny205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 08/15/2018] [Indexed: 12/17/2022] Open
Affiliation(s)
- Arianna Pompilio
- Department of Medical, Oral and Biotechnological Sciences, ‘G. d’Annunzio’ University of Chieti-Pescara, Via Vestini 31, Chieti 66100, Italy
- Center of Excellence on Aging, “G. d'Annunzio” University Foundation, Via L. Polacchi 11, Chieti 66100, Italy
| | - Valentina Crocetta
- Department of Medical, Oral and Biotechnological Sciences, ‘G. d’Annunzio’ University of Chieti-Pescara, Via Vestini 31, Chieti 66100, Italy
- Center of Excellence on Aging, “G. d'Annunzio” University Foundation, Via L. Polacchi 11, Chieti 66100, Italy
| | - Giovanni Di Bonaventura
- Department of Medical, Oral and Biotechnological Sciences, ‘G. d’Annunzio’ University of Chieti-Pescara, Via Vestini 31, Chieti 66100, Italy
- Center of Excellence on Aging, “G. d'Annunzio” University Foundation, Via L. Polacchi 11, Chieti 66100, Italy
| |
Collapse
|
5
|
The impact of host genetic background in the Pseudomonas aeruginosa respiratory infections. Mamm Genome 2018; 29:550-557. [PMID: 29947963 PMCID: PMC7087806 DOI: 10.1007/s00335-018-9753-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/05/2018] [Indexed: 12/27/2022]
Abstract
Understanding the significance of human genetic diversity in modulating host susceptibility to opportunistic infections is an emerging challenge in the field of respiratory illnesses. While it is recognized that diverse bacterial strains account for differential disease manifestations, emerging data indicate that host genetic diversity is an important determinant factor that influences the severity of opportunistic infections. With particular regard to respiratory illnesses mediated by the gram-negative bacterium Pseudomonas aeruginosa, diverse genetic background is also emerging as a key contributor. Human-genome-wide association studies are a common approach for determining the inter-individual genetic variation associated with variability of the pulmonary infections. Historically, diverse murine inbred mouse strains and ex-vivo cellular models were considered complementary to human studies for establishing the contribution of genetic background to P. aeruginosa respiratory infections. More recently, the development of a new mouse model of infection, mirroring human airway diseases, combined with innovative murine resource populations, modelling human genetic variation, provides additional insights into the mechanisms of genetic susceptibility. In this review, we cover the recent state of the art of human and animal studies and we discuss future potential challenges in the field of P. aeruginosa respiratory infections.
Collapse
|
6
|
The host genetic background defines diverse immune-reactivity and susceptibility to chronic Pseudomonas aeruginosa respiratory infection. Sci Rep 2016; 6:36924. [PMID: 27848994 PMCID: PMC5111113 DOI: 10.1038/srep36924] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 10/21/2016] [Indexed: 01/31/2023] Open
Abstract
Patients with P. aeruginosa airways infection show markedly variable clinical phenotypes likely influenced by genetic backgrounds. Here, we investigated the cellular events involved in resistance and susceptibility to P. aeruginosa chronic infection using genetically distinct inbred mouse strains. As for patients, different murine genotypes revealed variable susceptibility to infection. When directly compared, resistant C3H/HeOuJ and susceptible A/J strains revealed distinct immune responsiveness to the pathogen. In C3H/HeOuJ resistant mice, IL17-producing cells rapidly and transiently infiltrated the infected lung, and this was paralleled by the acute accumulation of alveolar macrophages, bacterial clearance and resolution of infection. In contrast, A/J susceptible mice revealed a more delayed and prolonged lung infiltration by IL17+ and IFNγ+ cells, persistence of innate inflammatory cells and establishment of chronic infection. We conclude that the host genetic background confers diverse immunoreactivity to P. aeruginosa and IL17-producing cells might contribute to the progress of chronic lung infection.
Collapse
|
7
|
De Simone M, Spagnuolo L, Lorè NI, Cigana C, De Fino I, Broman KW, Iraqi FA, Bragonzi A. Mapping genetic determinants of host susceptibility to Pseudomonas aeruginosa lung infection in mice. BMC Genomics 2016; 17:351. [PMID: 27169516 PMCID: PMC4866434 DOI: 10.1186/s12864-016-2676-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 04/28/2016] [Indexed: 12/16/2022] Open
Abstract
Background P. aeruginosa is one of the top three causes of opportunistic human bacterial infections. The remarkable variability in the clinical outcomes of this infection is thought to be associated with genetic predisposition. However, the genes underlying host susceptibility to P. aeruginosa infection are still largely unknown. Results As a step towards mapping these genes, we applied a genome wide linkage analysis approach to a mouse model. A large F2 intercross population, obtained by mating P. aeruginosa-resistant C3H/HeOuJ, and susceptible A/J mice, was used for quantitative trait locus (QTL) mapping. The F2 progenies were challenged with a P. aeruginosa clinical strain and monitored for the survival time up to 7 days post-infection, as a disease phenotype associated trait. Selected phenotypic extremes of the F2 distribution were genotyped with high-density single nucleotide polymorphic (SNP) markers, and subsequently QTL analysis was performed. A significant locus was mapped on chromosome 6 and was named P. aeruginosa infection resistance locus 1 (Pairl1). The most promising candidate genes, including Dok1, Tacr1, Cd207, Clec4f, Gp9, Gata2, Foxp1, are related to pathogen sensing, neutrophils and macrophages recruitment and inflammatory processes. Conclusions We propose a set of genes involved in the pathogenesis of P. aeruginosa infection that may be explored to complement human studies. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2676-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maura De Simone
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenza Spagnuolo
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicola Ivan Lorè
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Cigana
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ida De Fino
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Karl W Broman
- Department of Biostatistics & Medical Informatics, University of Wisconsin, Madison, WI, USA
| | - Fuad A Iraqi
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, 69978, Tel Aviv, Israel
| | - Alessandra Bragonzi
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
8
|
De Simone M, Spagnuolo L, Lorè NI, Rossi G, Cigana C, De Fino I, Iraqi FA, Bragonzi A. Host genetic background influences the response to the opportunistic Pseudomonas aeruginosa infection altering cell-mediated immunity and bacterial replication. PLoS One 2014; 9:e106873. [PMID: 25268734 PMCID: PMC4182038 DOI: 10.1371/journal.pone.0106873] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 08/02/2014] [Indexed: 12/22/2022] Open
Abstract
Pseudomonas aeruginosa is a common cause of healthcare-associated infections including pneumonia, bloodstream, urinary tract, and surgical site infections. The clinical outcome of P. aeruginosa infections may be extremely variable among individuals at risk and patients affected by cystic fibrosis. However, risk factors for P. aeruginosa infection remain largely unknown. To identify and track the host factors influencing P. aeruginosa lung infections, inbred immunocompetent mouse strains were screened in a pneumonia model system. A/J, BALB/cJ, BALB/cAnNCrl, BALB/cByJ, C3H/HeOuJ, C57BL/6J, C57BL/6NCrl, DBA/2J, and 129S2/SvPasCRL mice were infected with P. aeruginosa clinical strain and monitored for body weight and mortality up to seven days. The most deviant survival phenotypes were observed for A/J, 129S2/SvPasCRL and DBA/2J showing high susceptibility while BALB/cAnNCrl and C3H/HeOuJ showing more resistance to P. aeruginosa infection. Next, one of the most susceptible and resistant mouse strains were characterized for their deviant clinical and immunological phenotype by scoring bacterial count, cell-mediated immunity, cytokines and chemokines profile and lung pathology in an early time course. Susceptible A/J mice showed significantly higher bacterial burden, higher cytokines and chemokines levels but lower leukocyte recruitment, particularly neutrophils, when compared to C3H/HeOuJ resistant mice. Pathologic scores showed lower inflammatory severity, reduced intraluminal and interstitial inflammation extent, bronchial and parenchymal involvement and diminished alveolar damage in the lungs of A/J when compared to C3H/HeOuJ. Our findings indicate that during an early phase of infection a prompt inflammatory response in the airways set the conditions for a non-permissive environment to P. aeruginosa replication and lock the spread to other organs. Host gene(s) may have a role in the reduction of cell-mediated immunity playing a critical role in the control of P. aeruginosa infection. These results now provide a basis for mapping genomic regions underlying host susceptibility to P. aeruginosa infection.
Collapse
Affiliation(s)
- Maura De Simone
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Lorenza Spagnuolo
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Nicola Ivan Lorè
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Giacomo Rossi
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Cristina Cigana
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Ida De Fino
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Fuad A. Iraqi
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alessandra Bragonzi
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| |
Collapse
|
9
|
Sharma S, Chhibber S, Mohan H, Sharma S. Dietary supplementation with omega-3 polyunsaturated fatty acids ameliorates acute pneumonia induced by Klebsiella pneumoniae in BALB/c mice. Can J Microbiol 2013; 59:503-10. [PMID: 23826960 DOI: 10.1139/cjm-2012-0521] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The immune benefits associated with the optimal intake of dietary fatty acids are widely known. The objective of the present investigation was to elucidate the role of omega-3 polyunsaturated fatty acids (n-3 PUFA) food source on acute pneumonia induced by Klebsiella pneumoniae. Three different n-3 PUFA preparations (cod liver oil, Maxigard, and flaxseed oil) were orally supplemented and infection was induced in different groups of experimental mice. Mice fed olive oil and normal saline served as oil and saline controls, respectively. After 2 weeks of fatty acid feeding, no effect on the establishment of infection was observed when acute pneumonia was induced in animals. On the other hand, 6 weeks of n-3 PUFA administration was found to improve resistance in mice, as reduced lung bacterial load coupled with significant improvement in pathology was seen in infected mice. Alveolar macrophages collected from all 3 groups of mice fed n-3 PUFA exhibited a significant decrease in the level of apoptosis following infection with K. pneumoniae and an enhanced in vitro phagocytic potential for the pathogen. Lower lung levels of nitric oxide, malondialdehyde, and lactate dehydrogenase were associated with a decrease in the severity of tissue damage. There was a significant increase in the lung levels of pro-inflammatory cytokines (tumor necrosis factor alpha (TNF-α) and interleukin-1 beta (IL-1β)). No significant change was observed in the levels of interleukin-10 (IL-10). This study highlights that dietary n-3 PUFA supplementation exerts an overall beneficial effect against acute experimental pneumonia. This mechanism is operative through upregulation of nonspecific and specific immune defenses of the host.
Collapse
Affiliation(s)
- Sonica Sharma
- Department of Microbiology, Panjab University, Chandigarh, India.
| | | | | | | |
Collapse
|
10
|
Saini A, Harjai K, Chhibber S. Inhibitory effect of polyunsaturated fatty acids on apoptosis induced by Streptococcus pneumoniae in alveolar macrophages. Indian J Med Res 2013; 137:1193-8. [PMID: 23852301 PMCID: PMC3734725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
BACKGROUND & OBJECTIVES Apoptosis is considered as a major defense mechanism of the body. Multiple pathogens induce macrophage apoptosis as a mode of immune evasion. In earlier studies, n-3 polyunsaturated fatty acids (PUFA) have been reported to be protective against neuronal apoptosis and neuronal degeneration, seen after spinal cord injury. In this study, we tried to evaluate the role of n-3 polyunsaturated fatty acids on the process of macrophage phagocytic activity and apoptosis in mice. METHODS Mice were divided into three groups (n=60); Group I was fed on sea cod oil; Group II on flaxseed oil supplementation for 9 wk along with standard laboratory chow diet. Group III was fed on standard diet and served as control. After supplementation, phagocytic and apoptotic (morphological staining: acridine orange plus ethidium bromide; H-33342 plus propidium iodide staining and DNA ladder formation) activities of mouse alveolar macrophages were assessed. RESULTS Alveolar macrophages (obtained from sea cod oil and flaxseed oil fed group mice) showed significant increase in bacterial uptake as well as intracellular killing (P 0.05) of Streptococcus pneumoniae. Significant decrease (P<0.05) in apoptotic cells was observed among alveolar macrophages from sea cod and flaxseed oil fed mice whereas maximum apoptosis was observed in control alveolar macrophages on interaction with bacteria in vitro which was confirmed by DNA laddering. INTERPRETATION & CONCLUSIONS These findings suggest that dietary supplementation with n-3 polyunsaturated fatty acids to mice led to enhanced phagocytic capability of their alveolar macrophages as well as provided protection against apoptosis upon challenge with S. pneumoniae.
Collapse
Affiliation(s)
- Archana Saini
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Kusum Harjai
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Sanjay Chhibber
- Department of Microbiology, Panjab University, Chandigarh, India,Reprint requests: Prof. Sanjay Chhibber, Department of Microbiology, Panjab University, Chandigarh 160 014, India e-mail:
| |
Collapse
|
11
|
Glucksam-Galnoy Y, Sananes R, Silberstein N, Krief P, Kravchenko VV, Meijler MM, Zor T. The bacterial quorum-sensing signal molecule N-3-oxo-dodecanoyl-L-homoserine lactone reciprocally modulates pro- and anti-inflammatory cytokines in activated macrophages. THE JOURNAL OF IMMUNOLOGY 2013; 191:337-44. [PMID: 23720811 DOI: 10.4049/jimmunol.1300368] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The bacterial molecule N-3-oxo-dodecanoyl-l-homoserine lactone (C12) has critical roles in both interbacterial communication and interkingdom signaling. The ability of C12 to downregulate production of the key proinflammatory cytokine TNF-α in stimulated macrophages was suggested to contribute to the establishment of chronic infections by opportunistic Gram-negative bacteria, such as Pseudomonas aeruginosa. We show that, in contrast to TNF-α suppression, C12 amplifies production of the major anti-inflammatory cytokine IL-10 in LPS-stimulated murine RAW264.7 macrophages, as well as peritoneal macrophages. Furthermore, C12 increased IL-10 mRNA levels and IL-10 promoter reporter activity in LPS-stimulated RAW264.7 macrophages, indicating that C12 modulates IL-10 expression at the transcriptional level. Finally, C12 substantially potentiated LPS-stimulated NF-κB DNA-binding levels and prolonged p38 MAPK phosphorylation in RAW264.7 macrophages, suggesting that increased transcriptional activity of NF-κB and/or p38-activated transcription factors serves to upregulate IL-10 production in macrophages exposed to both LPS and C12. These findings reveal another part of the complex array of host transitions through which opportunistic bacteria downregulate immune responses to flourish and establish a chronic infection.
Collapse
Affiliation(s)
- Yifat Glucksam-Galnoy
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, Tel Aviv University, Tel Aviv 69978, Israel
| | | | | | | | | | | | | |
Collapse
|
12
|
Gellatly SL, Hancock RE. Pseudomonas aeruginosa: new insights into pathogenesis and host defenses. Pathog Dis 2013; 67:159-73. [DOI: 10.1111/2049-632x.12033] [Citation(s) in RCA: 877] [Impact Index Per Article: 73.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 01/24/2013] [Accepted: 02/15/2013] [Indexed: 01/15/2023] Open
Affiliation(s)
- Shaan L. Gellatly
- Centre for Microbial Diseases and Immunity Research; University of British Columbia; Vancouver; BC; Canada
| | - Robert E.W. Hancock
- Centre for Microbial Diseases and Immunity Research; University of British Columbia; Vancouver; BC; Canada
| |
Collapse
|
13
|
Kishta OA, Iskandar M, Dauletbaev N, Kubow S, Lands LC. Pressurized whey protein can limit bacterial burden and protein oxidation in Pseudomonas aeruginosa lung infection. Nutrition 2013; 29:918-24. [PMID: 23395602 DOI: 10.1016/j.nut.2012.11.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 10/06/2012] [Accepted: 11/18/2012] [Indexed: 12/27/2022]
Abstract
BACKGROUND Lung infection caused by Pseudomonas aeruginosa is associated with an exuberant inflammatory response, oxidative stress, and lung damage. Whey protein is a rich source of cysteine, and anti-inflammatory and immune-enhancing peptides. Anti-inflammatory and antioxidant properties of whey are augmented by hyperbaric pressure treatment. In this study, we tested whether dietary supplementation with pressurized whey protein enhances the host ability to clear P. aeruginosa infection compared with native (i.e., unpressurized) whey. METHODS Using a minimally invasive, non-lethal model of murine (female C57Bl/6) model of P. aeruginosa infection (mucoid strain embedded in agar beads), we studied kinetics of infection, inflammation, and oxidative stress at d 1, 3, and 7 postinfection. A parallel set of mice were fed for 4 wk a semipurified diet containing either native or pressurized whey and subsequently infected with P. aeruginosa. In these mice, the parameters mentioned previously were studied at d 1 and 3 postinfection. RESULTS Infection with P. aeruginosa resulted in inflammation and protein oxidation sustained beyond bacterial clearance. Animals that were fed pressurized whey had fewer bacteria at day 3 than mice on native whey. Weight loss or broncho-alveolar lavage cell content were comparable. Airway protein oxidation was attenuated, whereas airway leukocyte bacterial killing ability and oxidative burst in response to opsonized bacteria were increased in the pressurized whey-fed animals. CONCLUSIONS Use of nutritionally derived substances with anti-inflammatory and antioxidant properties, such as pressurized whey, aids in limiting airway bacterial infection, particularly, under conditions of ongoing oxidative stress.
Collapse
Affiliation(s)
- Osama A Kishta
- Division of Pediatric Respiratory Medicine, Montreal Children's Hospital-McGill University Health Centre, Montreal, QC, Canada
| | | | | | | | | |
Collapse
|
14
|
Unger BL, Faris AN, Ganesan S, Comstock AT, Hershenson MB, Sajjan US. Rhinovirus attenuates non-typeable Hemophilus influenzae-stimulated IL-8 responses via TLR2-dependent degradation of IRAK-1. PLoS Pathog 2012; 8:e1002969. [PMID: 23055935 PMCID: PMC3464227 DOI: 10.1371/journal.ppat.1002969] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 08/28/2012] [Indexed: 11/24/2022] Open
Abstract
Bacterial infections following rhinovirus (RV), a common cold virus, are well documented, but pathogenic mechanisms are poorly understood. We developed animal and cell culture models to examine the effects of RV on subsequent infection with non-typeable Hemophilus influenzae (NTHi). We focused on NTHI-induced neutrophil chemoattractants expression that is essential for bacterial clearance. Mice infected with RV1B were superinfected with NTHi and lung bacterial density, chemokines and neutrophil counts determined. Human bronchial epithelial cells (BEAS-2B) or mouse alveolar macrophages (MH-S) were infected with RV and challenged with NHTi, TLR2 or TLR5 agonists. Chemokine levels were measured by ELISA and expression of IRAK-1, a component of MyD88-dependent TLR signaling, assessed by immunoblotting. While sham-infected mice cleared all NTHi from the lungs, RV-infected mice showed bacteria up to 72 h post-infection. However, animals in RV/NTHi cleared bacteria by day 7. Delayed bacterial clearance in RV/NTHi animals was associated with suppressed chemokine levels and neutrophil recruitment. RV-infected BEAS-2B and MH-S cells showed attenuated chemokine production after challenge with either NTHi or TLR agonists. Attenuated chemokine responses were associated with IRAK-1 protein degradation. Inhibition of RV-induced IRAK-1 degradation restored NTHi-stimulated IL-8 expression. Knockdown of TLR2, but not other MyD88-dependent TLRs, also restored IRAK-1, suggesting that TLR2 is required for RV-induced IRAK-1 degradation. In conclusion, we demonstrate for the first time that RV infection delays bacterial clearance in vivo and suppresses NTHi-stimulated chemokine responses via degradation of IRAK-1. Based on these observations, we speculate that modulation of TLR-dependent innate immune responses by RV may predispose the host to secondary bacterial infection, particularly in patients with underlying chronic respiratory disorders. Rhinovirus (RV) is responsible for the majority of common colds. RV infection is also associated with hospitalizations for lower respiratory tract illness, a significant proportion of which are accompanied by bacterial infections including acute otitis media, sinusitis and pneumonia. However, the mechanisms by which RV increases susceptibility to secondary bacterial infections are not understood. In this report, we demonstrate for the first time that RV infection promotes bacterial persistence of non-typeable Hemophilus influenzae (NTHi) in vivo, which was associated with reduced expression of neutrophil-attracting chemokines and neutrophil infiltration into the lungs. Further, RV infection attenuated NTHi or TLR2 or −5 agonist-stimulated chemokine responses in cultured bronchial epithelial cells and alveolar macrophages, suggesting that RV interferes with TLR-related innate immune responses. Next, we found that RV infection caused rapid degradation of IRAK-1, a key adaptor protein in the MyD88-dependent signaling. Inhibition of IRAK-1 degradation restored NTHi-stimulated chemokine responses in RV-infected bronchial epithelial cells. Finally, reductions in IRAK-1 were dependent on TLR2. Together, our results suggest that RV may increase the risk of acquiring secondary bacterial infection by attenuating TLR-dependent innate immune responses.
Collapse
Affiliation(s)
- Benjamin L. Unger
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Andrea N. Faris
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Shyamala Ganesan
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Adam T. Comstock
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Marc B. Hershenson
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Umadevi S. Sajjan
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
15
|
Lorè NI, Cigana C, De Fino I, Riva C, Juhas M, Schwager S, Eberl L, Bragonzi A. Cystic fibrosis-niche adaptation of Pseudomonas aeruginosa reduces virulence in multiple infection hosts. PLoS One 2012; 7:e35648. [PMID: 22558188 PMCID: PMC3338451 DOI: 10.1371/journal.pone.0035648] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 03/19/2012] [Indexed: 01/07/2023] Open
Abstract
The opportunistic pathogen Pseudomonas aeruginosa is able to thrive in diverse ecological niches and to cause serious human infection. P. aeruginosa environmental strains are producing various virulence factors that are required for establishing acute infections in several host organisms; however, the P. aeruginosa phenotypic variants favour long-term persistence in the cystic fibrosis (CF) airways. Whether P. aeruginosa strains, which have adapted to the CF-niche, have lost their competitive fitness in the other environment remains to be investigated. In this paper, three P. aeruginosa clonal lineages, including early strains isolated at the onset of infection, and late strains, isolated after several years of chronic lung infection from patients with CF, were analysed in multi-host model systems of acute infection. P. aeruginosa early isolates caused lethality in the three non-mammalian hosts, namely Caenorhabditis elegans, Galleria mellonella, and Drosophila melanogaster, while late adapted clonal isolates were attenuated in acute virulence. When two different mouse genetic background strains, namely C57Bl/6NCrl and Balb/cAnNCrl, were used as acute infection models, early P. aeruginosa CF isolates were lethal, while late isolates exhibited reduced or abolished acute virulence. Severe histopathological lesions, including high leukocytes recruitment and bacterial load, were detected in the lungs of mice infected with P. aeruginosa CF early isolates, while late isolates were progressively cleared. In addition, systemic bacterial spread and invasion of epithelial cells, which were detected for P. aeruginosa CF early strains, were not observed with late strains. Our findings indicate that niche-specific selection in P. aeruginosa reduced its ability to cause acute infections across a broad range of hosts while maintaining the capacity for chronic infection in the CF host.
Collapse
Affiliation(s)
- Nicola Ivan Lorè
- Infection and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Cigana
- Infection and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Ida De Fino
- Infection and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Camilla Riva
- Infection and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Mario Juhas
- Department of Microbiology, Institute of Plant Biology, University of Zurich, Zurich, Switzerland
| | - Stephan Schwager
- Department of Microbiology, Institute of Plant Biology, University of Zurich, Zurich, Switzerland
| | - Leo Eberl
- Department of Microbiology, Institute of Plant Biology, University of Zurich, Zurich, Switzerland
| | - Alessandra Bragonzi
- Infection and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
- * E-mail:
| |
Collapse
|
16
|
Sharma S, Mohan H, Sharma S, Chhibber S. A comparative study of induction of pneumonia in mice with planktonic and biofilm cells of Klebsiella pneumoniae. Microbiol Immunol 2011; 55:295-303. [PMID: 21272064 DOI: 10.1111/j.1348-0421.2011.00317.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In the present study, the course of acute pneumonia in normal BALB/c mice infected by intranasal inoculation of planktonic and preformed biofilm cells (3 days old) of Klebsiella pneumoniae B5055 was studied and compared. With both cell forms the peak of infection was observed on the third post infection day, as assessed on the basis of lung bacterial load and corresponding pathology. There was an intense neutrophil infiltration in bronchoalveolar lavage fluid. Tissue damage was assessed on the basis of increased amounts of nitrite, malondialdehyde and lactate dehydrogenase in lung homogenates. The phagocytic potential of alveolar macrophages was lower in biofilm cell-induced infection than in that induced by planktonic cells. Biofilm cell induced infection generated significantly greater production of tumor necrosis factor-α and interleukin-1β on the third and fifth days of infection, respectively. Production of interleukin-10 was, however, variable. There was no significant difference in the ability of planktonic and biofilm cell forms of K. pneumoniae to induce acute pneumonia in mice in terms of bacterial counts and histopathological changes. However, biofilm cell-induced infection showed delayed clearance as compared to infection induced with the planktonic form.
Collapse
Affiliation(s)
- Sonica Sharma
- Department of Microbiology, Panjab University Department of Pathology, Government Medical College, Chandigarh, India.
| | | | | | | |
Collapse
|
17
|
Goldberg JB, Ganesan S, Comstock AT, Zhao Y, Sajjan US. Cable pili and the associated 22 kDa adhesin contribute to Burkholderia cenocepacia persistence in vivo. PLoS One 2011; 6:e22435. [PMID: 21811611 PMCID: PMC3141045 DOI: 10.1371/journal.pone.0022435] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Accepted: 06/27/2011] [Indexed: 01/02/2023] Open
Abstract
Background Infection by Burkholderia cenocepacia in cystic fibrosis (CF) patients is associated with poor clinical prognosis. Previously, we demonstrated that one of the highly transmissible strains, BC7, expresses cable pili and the associated 22 kDa adhesin, both of which contribute to BC7 binding to airway epithelial cells. However, the contribution of these factors to induce inflammation and bacterial persistence in vivo is not known. Methodology/Principal Findings Wild-type BC7 stimulated higher IL-8 responses than the BC7 cbl and BC7 adhA mutants in both CF and normal bronchial epithelial cells. To determine the role of cable pili and the associated adhesin, we characterized a mouse model of B. cenocepacia, where BC7 are suspended in Pseudomonas aeruginosa alginate. C57BL/6 mice were infected intratracheally with wild-type BC7 suspended in either alginate or PBS and were monitored for lung bacterial load and inflammation. Mice infected with BC7 suspended in PBS completely cleared the bacteria by 3 days and resolved the inflammation. In contrast, mice infected with BC7 suspended in alginate showed persistence of bacteria and moderate lung inflammation up to 5 days post-infection. Using this model, mice infected with the BC7 cbl and BC7 adhA mutants showed lower bacterial loads and mild inflammation compared to mice infected with wild-type BC7. Complementation of the BC7 cblS mutation in trans restored the capacity of this strain to persist in vivo. Immunolocalization of bacteria revealed wild-type BC7 in both airway lumen and alveoli, while the BC7 cbl and BC7 adhA mutants were found mainly in airway lumen and peribronchiolar region. Conclusions and Significance B. cenocepacia suspended in alginate can be used to determine the capacity of bacteria to persist and cause lung inflammation in normal mice. Both cable pili and adhesin contribute to BC7-stimulated IL-8 response in vitro, and BC7 persistence and resultant inflammation in vivo.
Collapse
Affiliation(s)
- Joanna B. Goldberg
- Department of Microbiology, University of Virginia Health System, Charlottesville, Virginia, United States of America
| | - Shyamala Ganesan
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Adam T. Comstock
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ying Zhao
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Uma S. Sajjan
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
18
|
Wangdi T, Mijares LA, Kazmierczak BI. In vivo discrimination of type 3 secretion system-positive and -negative Pseudomonas aeruginosa via a caspase-1-dependent pathway. Infect Immun 2010; 78:4744-53. [PMID: 20823203 PMCID: PMC2976309 DOI: 10.1128/iai.00744-10] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Revised: 08/23/2010] [Accepted: 08/29/2010] [Indexed: 01/10/2023] Open
Abstract
Microbe-associated molecular patterns are recognized by Toll-like receptors of the innate immune system. This recognition enables a rapid response to potential pathogens but does not clearly provide a way for the innate immune system to discriminate between virulent and avirulent microbes. We find that pulmonary infection of mice with type 3 translocation-competent Pseudomonas aeruginosa triggers a rapid inflammatory response, while infection with isogenic translocation-deficient mutants does not. Discrimination between translocon-positive and -negative bacteria requires caspase-1 activity in bone marrow-derived cells and interleukin-1 receptor signaling. Thus, the activation of caspase-1 by bacteria expressing type 3 secretion systems allows for rapid recognition of bacteria expressing conserved functions associated with virulence.
Collapse
Affiliation(s)
- Tamding Wangdi
- Department of Internal Medicine (Infectious Diseases), Section of Microbial Pathogenesis, Graduate Program in Microbiology, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520
| | - Lilia A. Mijares
- Department of Internal Medicine (Infectious Diseases), Section of Microbial Pathogenesis, Graduate Program in Microbiology, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520
| | - Barbara I. Kazmierczak
- Department of Internal Medicine (Infectious Diseases), Section of Microbial Pathogenesis, Graduate Program in Microbiology, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520
| |
Collapse
|
19
|
Abstract
Lung infections caused by the opportunistic pathogen Pseudomonas aeruginosa can present as a spectrum of clinical entities from a rapidly fatal pneumonia in a neutropenic patient to a multi-decade bronchitis in patients with cystic fibrosis. P. aeruginosa is ubiquitous in our environment, and one of the most versatile pathogens studied, capable of infecting a number of diverse life forms and surviving harsh environmental factors. It is also able to quickly adapt to new environments, including the lung, where it orchestrates virulence factors to acquire necessary nutrients, and if necessary, turn them off to prevent immune recognition. Despite these capabilities, P. aeruginosa rarely infects healthy human lungs. This is secondary to a highly evolved host defence mechanism that efficiently removes inhaled or aspirated pseudomonads. Many arms of the respiratory host defence have been elucidated using P. aeruginosa as a model pathogen. Human infections with P. aeruginosa have demonstrated the importance of the mechanical barrier functions including mucus clearance, and the innate immune system, including the critical role of the neutrophilic response. As more models of persistent or biofilm P. aeruginosa infections are developed, the role of the adaptive immune response will likely become more evident. Understanding the pathogenesis of P. aeruginosa, and the respiratory host defence response to it has, and will continue to, lead to novel therapeutic strategies to help patients.
Collapse
Affiliation(s)
- Bryan J Williams
- Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | | | |
Collapse
|
20
|
Dubin PJ, Kolls JK. Pseudomonas aeruginosa and the host pulmonary immune response. Expert Rev Respir Med 2010; 1:121-37. [PMID: 20477272 DOI: 10.1586/17476348.1.1.121] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Pseudomonas aeruginosa is a highly adaptable, opportunistic pathogen that is commonly found in the environment. It can infect a number of sites in the body and disseminate. It can cause both acute and chronic pulmonary infection and the acuity of infection and accompanying inflammatory phenotype is determined, for the most part, by the host. Although P. aeruginosa has been a successful opportunist in the context of a number of different disease states, it has been best studied in the context of cystic fibrosis (CF). The adaptability of P. aeruginosa has enabled it to adjust quickly to the CF airway, transitioning from initial colonization to chronic infection. The organism quickly expresses virulence factors that allow it to circumvent some elements of the host immune response and, even more importantly, quickly develops antimicrobial resistance. In the case of CF, chronic infection resulting in progressive lung damage, coupled with antimicrobial resistance, becomes an increasingly important issue as individuals with CF live longer. It is for these reasons that both organism- and host-targeted immunotherapies are being increasingly explored.
Collapse
Affiliation(s)
- Patricia J Dubin
- Children's Hospital of Pittsburgh, Suite 3765, 3705 Fifth Avenue, Pittsburgh, PA 15213, USA.
| | | |
Collapse
|
21
|
Role of excessive inflammatory response to Stenotrophomonas maltophilia lung infection in DBA/2 mice and implications for cystic fibrosis. Infect Immun 2010; 78:2466-76. [PMID: 20308302 DOI: 10.1128/iai.01391-09] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Stenotrophomonas maltophilia is a pathogen that causes infections mainly in immunocompromised patients. Despite increased S. maltophilia isolation from respiratory specimens of patients with cystic fibrosis (CF), the real contribution of the microorganism to CF pathogenesis still needs to be clarified. The aim of the present study was to evaluate the pathogenic role of S. maltophilia in CF patients by using a model of acute respiratory infection in DBA/2 mice following a single exposure to aerosolized bacteria. The pulmonary bacterial load was stable until day 3 and then decreased significantly from day 3 through day 14, when the bacterial load became undetectable in all infected mice. Infection disseminated in most mice, although at a very low level. Severe effects (swollen lungs, large atelectasis, pleural adhesion, and hemorrhages) of lung pathology were observed on days 3, 7, and 14. The clearance of S. maltophilia observed in DBA/2 mouse lungs was clearly associated with an early and intense bronchial and alveolar inflammatory response, which is mediated primarily by neutrophils. Significantly higher levels of interleukin-1beta (IL-1beta), IL-6, IL-12, gamma interferon (IFN-gamma), tumor necrosis factor alpha (TNF-alpha), GROalpha/KC, MCP-1/JE, MCP-5, macrophage inflammatory protein 1alpha (MIP-1alpha), MIP-2, and TARC were observed in infected mice on day 1 with respect to controls. Excessive pulmonary infection and inflammation caused systemic effects, manifested by weight loss, and finally caused a high mortality rate. Taken together, our results show that S. maltophilia is not just a bystander in CF patients but has the potential to contribute to the inflammatory process that compromises respiratory function.
Collapse
|
22
|
Pseudomonas aeruginosa alginate promotes Burkholderia cenocepacia persistence in cystic fibrosis transmembrane conductance regulator knockout mice. Infect Immun 2010; 78:984-93. [PMID: 20048042 DOI: 10.1128/iai.01192-09] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pseudomonas aeruginosa, a major respiratory pathogen in cystic fibrosis (CF) patients, facilitates infection by other opportunistic pathogens. Burkholderia cenocepacia, which normally infects adolescent patients, encounters alginate elaborated by mucoid P. aeruginosa. To determine whether P. aeruginosa alginate facilitates B. cenocepacia infection in mice, cystic fibrosis transmembrane conductance regulator knockout mice were infected with B. cenocepacia strain BC7 suspended in either phosphate-buffered saline (BC7/PBS) or P. aeruginosa alginate (BC7/alginate), and the pulmonary bacterial load and inflammation were monitored. Mice infected with BC7/PBS cleared all of the bacteria within 3 days, and inflammation was resolved by day 5. In contrast, mice infected with BC7/alginate showed persistence of bacteria and increased cytokine levels for up to 7 days. Histological examination of the lungs indicated that there was moderate to severe inflammation and pneumonic consolidation in isolated areas at 5 and 7 days postinfection in the BC7/alginate group. Further, alginate decreased phagocytosis of B. cenocepacia by professional phagocytes both in vivo and in vitro. P. aeruginosa alginate also reduced the proinflammatory responses of CF airway epithelial cells and alveolar macrophages to B. cenocepacia infection. The observed effects are specific to P. aeruginosa alginate, because enzymatically degraded alginate or other polyuronic acids did not facilitate bacterial persistence. These observations suggest that P. aeruginosa alginate may facilitate B. cenocepacia infection by interfering with host innate defense mechanisms.
Collapse
|
23
|
Zhou Z, Wu M, Barrett RP, McClellan SA, Zhang Y, Hazlett LD. Role of the Fas pathway in Pseudomonas aeruginosa keratitis. Invest Ophthalmol Vis Sci 2009; 51:2537-47. [PMID: 20019368 DOI: 10.1167/iovs.09-4152] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
PURPOSE The role of the Fas pathway was tested in Pseudomonas aeruginosa-infected mouse cornea by contrasting the responses of FasL(-/-) and wild-type (WT) mice. METHODS TUNEL staining, real-time RT-PCR, immunostaining, and ELISA assay were used. RESULTS Compared with WT (resistant) mice, BALB/c FasL(-/-) exhibited significantly elevated bacterial counts and polymorphonuclear leukocyte numbers at 1 and 3 days postinfection (p.i.) and worse outcomes from disease. Similar bacterial challenges in C57BL/6 FasL(-/-) compared with WT mice also led to worsened disease as evidenced by earlier corneal perforation in the susceptible mouse strain. Intense TUNEL staining of apoptotic cells was seen earlier (1 day vs. 3 days) p.i. in BALB/c WT than in knockout mice, This earlier apoptotic pattern correlated with increased expression of caspases 3, 8, and 9 and BAX and with decreased expression of the antiapoptotic molecule Bcl-2. Furthermore, expression levels of the proinflammatory molecule TNF-alpha and its receptor, MIP-2, inducible nitric oxide synthase (iNOS), and nitrite also were significantly elevated in the infected cornea of BALB/c FasL(-/-) compared with WT mice. In vitro, LPS-stimulated Mphi from BALB/c FasL(-/-) mice expressed significantly less caspase 3 and 9, BAX, and IL-10 and more TNF-alpha, MIP-2, and IL-1beta than did cells from WT mice. CONCLUSIONS Fas-FasL interaction in the cornea balances the host innate immune response to improve disease outcome by promoting earlier apoptosis and regulating proinflammatory cytokines/chemokines and nitric oxide (nitrite) production. Dysregulation of this interaction contributes to bystander tissue damage, enhancing nutrients for bacterial growth and worsened disease outcome after P. aeruginosa infection.
Collapse
Affiliation(s)
- Zimei Zhou
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | | | | | | | |
Collapse
|
24
|
McClellan SA, Lighvani S, Hazlett LD. IFN-γ: Regulation of Nitric Oxide in theP. aeruginosa-Infected Cornea. Ocul Immunol Inflamm 2009; 14:21-8. [PMID: 16507487 DOI: 10.1080/09273940500545650] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
PURPOSE BALB/c mice are resistant to Pseudomonas aeruginosa (P. aeruginosa) keratitis and bacterial killing/stasis requires nitric oxide (NO). NO regulation in the cornea is unknown and was tested in this model. METHODS Nitrite detection, IFN-gamma-knockout mice, TNF-alpha neutralization, ELISA, aminoguanidine (AG) treatment, MPO, and plate counts were done. RESULTS Evidence shows (i) without IFN-gamma, nitrite levels are elevated, (ii) neutralization of TNF-alpha does not change nitrite levels, and (iii) absence of IFN-alpha and reduced NO synergistically increases disease progress and upregulates pro-inflammatory cytokines, PMN number, and bacterial load. CONCLUSION IFN-gamma regulates NO levels, and synergistic interaction between the two regulates disease outcome in resistant mice.
Collapse
Affiliation(s)
- Sharon A McClellan
- Department of Anatomy & Cell Biology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | |
Collapse
|
25
|
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: 3.5] [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
|
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: 1.4] [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
|
Reiniger N, Lee MM, Coleman FT, Ray C, Golan DE, Pier GB. Resistance to Pseudomonas aeruginosa chronic lung infection requires cystic fibrosis transmembrane conductance regulator-modulated interleukin-1 (IL-1) release and signaling through the IL-1 receptor. Infect Immun 2007; 75:1598-608. [PMID: 17283089 PMCID: PMC1865697 DOI: 10.1128/iai.01980-06] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Innate immunity is critical for clearing Pseudomonas aeruginosa from the lungs. In response to P. aeruginosa infection, a central transcriptional regulator of innate immunity-NF-kappaB-is translocated within 15 min to the nuclei of respiratory epithelial cells expressing wild-type (WT) cystic fibrosis (CF) transmembrane conductance regulator (CFTR). P. aeruginosa clearance from lungs is impaired in CF, and rapid NF-kappaB nuclear translocation is defective in cells with mutant or missing CFTR. We used WT and mutant P. aeruginosa and strains of transgenic mice lacking molecules involved in innate immunity to identify additional mediators required for P. aeruginosa-induced rapid NF-kappaB nuclear translocation in lung epithelia. We found neither Toll-like receptor 2 (TLR2) nor TLR4 nor TLR5 were required for this response. However, both MyD88-deficient mice and interleukin-1 receptor (IL-1R)-deficient mice failed to rapidly translocate NF-kappaB to the nuclei of respiratory epithelial cells in response to P. aeruginosa. Cultured human bronchial epithelial cells rapidly released IL-1beta in response to P. aeruginosa; this process was maximized by expression of WT-CFTR and dramatically muted in cells with DeltaF508-CFTR. The IL-1R antagonist blocked P. aeruginosa-induced NF-kappaB nuclear translocation. Oral inoculation via drinking water of IL-1R knockout mice resulted in higher rates of lung colonization and elevated P. aeruginosa-specific antibody titers in a manner analogous to that of CFTR-deficient mice. Overall, rapid IL-1 release and signaling through IL-1R represent key steps in the innate immune response to P. aeruginosa infection, and this process is deficient in cells lacking functional CFTR.
Collapse
Affiliation(s)
- Nina Reiniger
- Channing Laboratory, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
28
|
Lee VT, Pukatzki S, Sato H, Kikawada E, Kazimirova AA, Huang J, Li X, Arm JP, Frank DW, Lory S. Pseudolipasin A is a specific inhibitor for phospholipase A2 activity of Pseudomonas aeruginosa cytotoxin ExoU. Infect Immun 2006; 75:1089-98. [PMID: 17178785 PMCID: PMC1828555 DOI: 10.1128/iai.01184-06] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A number of bacterial pathogens utilize the type III secretion pathway to deliver effector proteins directly into the host cell cytoplasm. Certain strains of Pseudomonas aeruginosa associated with acute infections express a potent cytotoxin, exoenzyme U (ExoU), that is delivered via the type III secretion pathway directly into contacting host cells. Once inside the mammalian cell, ExoU rapidly lyses the intoxicated cells via its phospholipase A(2) (PLA(2)) activity. A high-throughput cell-based assay was developed to screen libraries of compounds for those capable of protecting cells against the cytotoxic effects of ExoU. A number of compounds were identified in this screen, including one group that blocks the intracellular activity of ExoU. In addition, these compounds specifically inhibited the PLA(2) activity of ExoU in vitro, whereas eukaryotic secreted PLA(2) and cytosolic PLA(2) were not inhibited. This novel inhibitor of ExoU-specific PLA(2) activity, named pseudolipasin A, may provide a new lead for virulence factor-based therapeutic design.
Collapse
Affiliation(s)
- Vincent T Lee
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Guilbault C, Novak JP, Martin P, Boghdady ML, Saeed Z, Guiot MC, Hudson TJ, Radzioch D. Distinct pattern of lung gene expression in theCftr-KO mice developing spontaneous lung disease compared with their littermate controls. Physiol Genomics 2006; 25:179-93. [PMID: 16418321 DOI: 10.1152/physiolgenomics.00206.2005] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cystic fibrosis (CF) is caused by a defect in the CF 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. Most of the morbidity and mortality of CF patients results from pulmonary complications. Differences in susceptibility to bacterial infection and variable degree of CF lung disease among CF patients remain unexplained. Many phenotypic expressions of the disease do not directly correlate with the type of mutation in the Cftr gene. Using a unique CF mouse model that mimics aspects of human CF lung disease, we analyzed the differential gene expression pattern between the normal lungs of wild-type mice (WT) and the affected lungs of CFTR knockout mice (KO). Using microarray analysis followed by quantitation of candidate gene mRNA and protein expression, we identified many interesting genes involved in the development of CF lung disease in mice. These findings point to distinct mechanisms of gene expression regulation between mice with CF and control mice.
Collapse
|
30
|
Ballinger MN, Paine R, Serezani CHC, Aronoff DM, Choi ES, Standiford TJ, Toews GB, Moore BB. Role of granulocyte macrophage colony-stimulating factor during gram-negative lung infection with Pseudomonas aeruginosa. Am J Respir Cell Mol Biol 2006; 34:766-74. [PMID: 16474098 PMCID: PMC2644237 DOI: 10.1165/rcmb.2005-0246oc] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Granulocyte macrophage colony-stimulating factor (GM-CSF) stimulates survival, proliferation, differentiation, and function of myeloid cells. Recently, GM-CSF has been shown to be important for normal pulmonary homeostasis. We report that GM-CSF is induced in lung leukocytes during infection with Gram-negative bacteria. Therefore, we postulated that deficiencies in GM-CSF would increase susceptibility to Gram-negative infection in vivo. After an intratracheal inoculum with Pseudomonas aeruginosa, GM-CSF-/- mice show decreased survival compared with wild-type mice. GM-CSF-/- mice show increased lung, spleen, and blood bacterial CFU. GM-CSF-/- mice are defective in the production of cysteinyl leukotrienes, prostaglandin E2, macrophage inflammatory protein, and keratinocyte-derived chemokine in lung leukocytes postinfection. Despite these defects, inflammatory cell recruitment is not diminished at 6 or 24 h postinfection, and the functional activity of polymorphonuclear leukocytes from the lung and peritoneum against P. aeruginosa is enhanced in GM-CSF-/- mice. In contrast, alveolar macrophage (AM) phagocytosis, killing, and H2O2 production are defective in GM-CSF-/- mice. Although the absence of GM-CSF has profound effects on AMs, peritoneal macrophages seem to have normal bactericidal activities in GM-CSF-/- mice. Defects in AM function may be related to diminished levels of IFN-gamma and TNF-alpha postinfection. Thus, GM-CSF-/- mice are more susceptible to lung infection with P. aeruginosa as a result of impaired AM function.
Collapse
MESH Headings
- Animals
- Chemokines, CXC/metabolism
- Colony Count, Microbial
- Cytokines/metabolism
- Eicosanoids/metabolism
- Granulocyte-Macrophage Colony-Stimulating Factor/genetics
- Granulocyte-Macrophage Colony-Stimulating Factor/immunology
- Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
- Leukocytes/immunology
- Leukocytes/metabolism
- Leukocytes/microbiology
- Lung/immunology
- Lung/metabolism
- Lung/microbiology
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/metabolism
- Macrophages, Alveolar/microbiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Phagocytosis
- Pneumonia, Bacterial/immunology
- Pneumonia, Bacterial/metabolism
- Pneumonia, Bacterial/microbiology
- Pseudomonas Infections/immunology
- Pseudomonas Infections/metabolism
- Pseudomonas Infections/microbiology
- Pseudomonas aeruginosa/isolation & purification
Collapse
Affiliation(s)
- Megan N Ballinger
- Immunology Graduate Program, Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI 48109-0642, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Ader F, Le Berre R, Faure K, Gosset P, Epaulard O, Toussaint B, Polack B, Nowak E, Viget NB, Kipnis E, Guery BP. Alveolar response to Pseudomonas aeruginosa: role of the type III secretion system. Infect Immun 2005; 73:4263-71. [PMID: 15972518 PMCID: PMC1168600 DOI: 10.1128/iai.73.7.4263-4271.2005] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The type III secretion system (TTSS) is a specialized cytotoxin-translocating apparatus of gram-negative bacteria which is involved in lung injury, septic shock, and a poor patient outcome. Recent studies have attributed these effects mainly to the ExoU effector protein. However, few studies have focused on the ExoU-independent pathogenicity of the TTSS. For the present study, we compared the pathogenicities of two strains of Pseudomonas aeruginosa in a murine model of acute lung injury. We compared the CHA strain, which has a functional TTSS producing ExoS and ExoT but not ExoU, to an isogenic mutant with an inactivated exsA gene, CHA-D1, which does not express the TTSS at all. Rats challenged with CHA had significantly increased lung injury, as assessed by the wet/dry weight ratio for the lungs and the protein level in bronchoalveolar lavage fluid (BALF) at 12 h, compared to those challenged with CHA-D1. Consistent with these findings, the CHA strain was associated with increased in vitro cytotoxicity on A549 cells, as assessed by the release of lactate dehydrogenase. CHA was also associated at 12 h with a major decrease in polymorphonuclear neutrophils in BALF, with a proinflammatory response, as assessed by the amounts of tumor necrosis factor alpha and interleukin-1beta, and with decreased bacterial clearance from the lungs, ultimately leading to an increased mortality rate. These results demonstrate that the TTSS has a major role in P. aeruginosa pathogenicity independent of the role of ExoU. This report underscores the crucial roles of ExoS and ExoT or other TTSS-related virulence factors in addition to ExoU.
Collapse
Affiliation(s)
- F Ader
- Laboratoire de Recherche en Pathologie Infectieuse, Faculté de Médecine de Lille, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Sadikot RT, Blackwell TS, Christman JW, Prince AS. Pathogen-host interactions in Pseudomonas aeruginosa pneumonia. Am J Respir Crit Care Med 2005; 171:1209-23. [PMID: 15695491 PMCID: PMC2718459 DOI: 10.1164/rccm.200408-1044so] [Citation(s) in RCA: 594] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Pseudomonas aeruginosa is an important pathogen causing a wide range of acute and chronic infections. P. aeruginosa rarely causes infection in the normal host, but is an efficient opportunistic pathogen causing serious infections in patients who are mechanically ventilated, individuals who are immunocompromised, and patients with malignancies or HIV infection. Among these risk groups, the most vulnerable hosts are neutropenic and patients who are mechanically ventilated. In addition, P. aeruginosa is the most prevalent chronic infection contributing to the pathogenesis of cystic fibrosis. Because of the ubiquitous nature of P. aeruginosa and its ability to develop resistance to antibiotics, it continues to be problematic from a treatment perspective. The pathogenicity of P. aeruginosa is largely caused by multiple bacterial virulence factors and genetic flexibility enabling it to survive in varied environments. Lung injury associated with P. aeruginosa infection results from both the direct destructive effects of the organism on the lung parenchyma and exuberant host immune responses. This article focuses on the major bacterial virulence factors and important aspects of the host immunity that are involved in the pathogenesis of serious P. aeruginosa infection. In addition to antibiotic therapy, strategies directed toward enhancing host defense and/or limiting excessive inflammation could be important to improve outcome in P. aeruginosa lung infections.
Collapse
Affiliation(s)
- Ruxana T Sadikot
- Department of Veterans Affairs Medical Center, Nashville, Tennessee, USA.
| | | | | | | |
Collapse
|
33
|
Jensen PØ, Moser C, Kobayashi O, Hougen HP, Kharazmi A, Høiby N. Faster activation of polymorphonuclear neutrophils in resistant mice during early innate response to Pseudomonas aeruginosa lung infection. Clin Exp Immunol 2004; 137:478-85. [PMID: 15320896 PMCID: PMC1809146 DOI: 10.1111/j.1365-2249.2004.02554.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Polymorphonuclear neutrophils (PMNs) are crucial for the outcome of Pseudomonas aeruginosa lung infection in patients with cystic fibrosis. We compared PMNs and inflammatory cytokines in the lungs and blood from susceptible BALB/c and resistant C3H/HeN mice 1 and 2 days after intratracheal challenge with alginate embedded P. aeruginosa. These parameters were correlated with the quantitative bacteriology and histopathology of the lungs. After challenge, the content of granulocyte colony-stimulating factor (G-CSF) and macrophage inflammatory protein-2 (MIP-2) was increased in the lungs and the sera and the percentage of PMNs was increased in the blood. However, 2 days after challenge the concentration of G-CSF and MIP-2 was higher in the lungs and sera of BALB/c mice. CD11b expression was higher on the PMNs of the C3H/HeN mice. The expression of CD62L on PMNs of both strains of mice was decreased 1 day after bacterial challenge, whereas the expression was increased after 2 days of challenge on PMNs of C3H/HeN mice only. These changes were accompanied by a more severe lung inflammation in BALB/c mice and faster clearance of the bacteria in C3H/HeN mice. In conclusion, the rapid early bacterial clearance in the lungs of C3H/HeN mice could be explained by faster activation of the PMNs, as indicated by the higher up-regulation of CD11b. The severe lung inflammation in BALB/c mice may be caused by the early higher content of G-CSF in the sera mobilizing PMNs from the bone marrow and the persistent chemotactic gradient provided by MIP-2 in the lungs.
Collapse
Affiliation(s)
- P Ø Jensen
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark.
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is a common organism associated with bacterial keratitis, especially in those who use extended wear contact lenses. Recent advances in our understanding of host innate and adaptive immune responses to experimental infection have been made using a variety of animal models, including inbred murine models that are classed as resistant (cornea heals) vs. susceptible (cornea perforates). Evidence has been provided that sustained IL-12-driven IFN-gamma production in dominant Th1 responder strains such as C57BL/6 (B6) contributes to corneal destruction and perforation, while IL-18-driven production of IFN-gamma in the absence of IL-12 is associated with bacterial killing and less corneal destruction in dominant Th2 responder strains such as BALB/c. The critical role of IL-1 and chemotactic cytokines such as MIP-2 in PMN recruitment and the critical role of this cell in the innate immune response to bacterial infection is reviewed. Regulation of PMN persistence is also discussed and evidence provided that persistence of PMN in B6 cornea is regulated by CD4+ T cells, while macrophages regulate PMN number in the cornea of BALB/c mice. The studies provide a better understanding of the inflammatory mechanisms that are operative in the cornea after P. aeruginosa challenge and are consistent with long-term goals of providing targets for alternative or adjunctive treatment for this disease. Future studies will be aimed at better defining the role of Toll receptors, neuropeptides (as unconventional modulators of the immune response) and exploitation of disease control by new techniques, such as RNA silencing.
Collapse
Affiliation(s)
- Linda Dondero Hazlett
- Department of Anatomy and Cell Biology, Wayne State University, 540 E Canfield, Room 7341, Detroit, MI 48201, USA.
| |
Collapse
|
35
|
Ritchie AJ, Yam AOW, Tanabe KM, Rice SA, Cooley MA. Modification of in vivo and in vitro T- and B-cell-mediated immune responses by the Pseudomonas aeruginosa quorum-sensing molecule N-(3-oxododecanoyl)-L-homoserine lactone. Infect Immun 2003; 71:4421-31. [PMID: 12874321 PMCID: PMC165988 DOI: 10.1128/iai.71.8.4421-4431.2003] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
N-3-(oxododecanoyl)-L-homoserine lactone (OdDHL), a quorum-sensing molecule of Pseudomonas aeruginosa, plays an important role in the pathogenesis of the organism through its control of virulence factor expression. Several reports have suggested that OdDHL can also directly modulate host immune responses. However, the nature of the modulation is controversial, with different reports suggesting promotion of either humoral (Th2-mediated) or inflammatory (Th1-mediated) responses. This report describes a series of studies which demonstrate for the first time that in vivo administration of OdDHL can modulate the course of an antibody response, with an increase in ovalbumin (OVA)-specific immunogloblulin G1 (IgG1) but not IgG2a in OdDHL-treated OVA-immunized BALB/c mice compared to levels for controls. In vitro stimulation of lymphocytes from both Th1-biased C57Bl/6 and T-cell receptor transgenic mice and Th2-biased BALB/c mice in the presence of OdDHL demonstrated that OdDHL inhibits in vitro cytokine production in response to both mitogen and antigen, with gamma interferon (IFN-gamma) tending to be more inhibited than interleukin-4 (IL-4). In vitro mitogen or antigen restimulation of cells from mice treated with OdDHL in vivo shows effects on cytokine production which depend on the underlying immune bias of the mouse strain used, with a relative increase of IFN-gamma in Th1-biased C57Bl/6 mice and a relative increase of IL-4 in Th2-biased BALB/c mice. Thus, the mode of action of OdDHL on T-cell cytokine production is likely to be a relatively nonspecific one which accentuates an underlying immune response bias rather than one which specifically targets either Th1 or Th2 responses.
Collapse
Affiliation(s)
- Adam J Ritchie
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia 2052
| | | | | | | | | |
Collapse
|
36
|
Venkatesh J, Kumar P, Krishna PSM, Manjunath R, Varshney U. Importance of uracil DNA glycosylase in Pseudomonas aeruginosa and Mycobacterium smegmatis, G+C-rich bacteria, in mutation prevention, tolerance to acidified nitrite, and endurance in mouse macrophages. J Biol Chem 2003; 278:24350-8. [PMID: 12679366 DOI: 10.1074/jbc.m302121200] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Uracil DNA glycosylase (Ung (or UDG)) initiates the excision repair of an unusual base, uracil, in DNA. Ung is a highly conserved protein found in all organisms. Paradoxically, loss of this evolutionarily conserved enzyme has not been seen to result in severe growth phenotypes in the cellular life forms. In this study, we chose G+C-rich genome containing bacteria (Pseudomonas aeruginosa and Mycobacterium smegmatis) as model organisms to investigate the biological significance of ung. Ung deficiency was created either by expression of a highly specific inhibitor protein, Ugi, and/or by targeted disruption of the ung gene. We show that abrogation of Ung activity in P. aeruginosa and M. smegmatis confers upon them an increased mutator phenotype and sensitivity to reactive nitrogen intermediates generated by acidified nitrite. Also, in a mouse macrophage infection model, P. aeruginosa (Ung-) shows a significant decrease in its survival. Infections of the macrophages with M. smegmatis show an initial increase in the bacterial counts that remain for up to 48 h before a decline. Interestingly, abrogation of Ung activity in M. smegmatis results in nearly a total abolition of their multiplication and a much-decreased residency in macrophages stimulated with interferon gamma. These observations suggest Ung as a useful target to control growth of G+C-rich bacteria.
Collapse
Affiliation(s)
- Jeganathan Venkatesh
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, 560 012 India
| | | | | | | | | |
Collapse
|
37
|
Huang X, McClellan SA, Barrett RP, Hazlett LD. IL-18 contributes to host resistance against infection with Pseudomonas aeruginosa through induction of IFN-gamma production. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:5756-63. [PMID: 12023376 DOI: 10.4049/jimmunol.168.11.5756] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Pseudomonas aeruginosa keratitis destroys the cornea in susceptible (B6), but not resistant (BALB/c) mice. To determine mechanisms mediating resistance, the role of IFN-gamma, IL-12, and IL-18 was tested in BALB/c mice. RT-PCR analysis detected IFN-gamma mRNA expression levels in cornea that were significantly increased at 1-7 days postinfection. IL-18 mRNA was detected constitutively in cornea and, at 1-7 days postinfection, levels were elevated significantly, while no IL-12 mRNA was similarly detected. To test whether IL-18 contributed to IFN-gamma production, mice were treated with anti-IL-18 mAb. Treatment decreased corneal IFN-gamma mRNA levels, and bacterial load and disease increased/worsened, compared with IgG-treated mice. To stringently examine the role of IFN-gamma in bacterial killing, knockout (-/-) vs wild-type (wt) mice also were tested. All corneas perforated, and bacterial load was increased significantly in -/- vs wt mice. Because disease severity was increased in IFN-gamma(-/-) vs IL-18-neutralized mice, and since IL-18 also induces production of TNF, we tested for TNF-alpha in both groups. ELISA analysis demonstrated significantly elevated corneal TNF-alpha protein levels in IFN-gamma(-/-) vs wt mice after infection. In contrast, RT-PCR analysis of IL-18-neutralized vs IgG-treated infected mice revealed decreased corneal TNF-alpha mRNA expression. Next, to resolve whether TNF was required for bacterial killing, TNF-alpha was neutralized in BALB/c mice. No difference in corneal bacterial load was detected in neutralized vs IgG-treated mice. These data provide evidence that IL-18 contributes to the resistance response by induction of IFN-gamma and that IFN-gamma is required for bacterial killing.
Collapse
Affiliation(s)
- Xi Huang
- Department of Anatomy/Cell Biology, Wayne State University School of Medicine, Detroit, MI 48201
| | | | | | | |
Collapse
|
38
|
Moser C, Jensen PO, Kobayashi O, Hougen HP, Song Z, Rygaard J, Kharazmi A, H by N. Improved outcome of chronic Pseudomonas aeruginosa lung infection is associated with induction of a Th1-dominated cytokine response. Clin Exp Immunol 2002; 127:206-13. [PMID: 11876741 PMCID: PMC1906339 DOI: 10.1046/j.1365-2249.2002.01731.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Repeated challenge with antigen is involved in the pathogenesis of a variety of pulmonary diseases. Patients with cystic fibrosis (CF) experience recurrent pulmonary colonization with Pseudomonas aeruginosa before establishment of chronic lung infection. To mimic recurrent lung infections in CF patients, the lungs of susceptible BALB/c mice were re-infected with P. aeruginosa 14 days after the initial infection. Singly-infected BALB/c mice, as well as non-infected mice, were used as controls. Decreased mortality and milder lung inflammation in re-infected BALB/c mice, as well as a tendency for improved clearance of bacteria, was observed when compared with singly-infected mice. The improved outcome in re-infected mice correlated with changes in CD4 cell numbers. Surface expression of LFA-1 on pulmonary CD4 cells was increased in re-infected compared with singly-infected mice. Moreover, resistance to re-infection was paralleled by a shift towards a Th1-dominated response and increased IL-12 production. No significant increase in serum IgG was observed in the re-infected mice. In conclusion, these results indicate a protective role for a Th1-dominated response, independent of antibody production, in chronic P. aeruginosa lung infection in CF.
Collapse
Affiliation(s)
- C Moser
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Shime N, Sawa T, Fujimoto J, Faure K, Allmond LR, Karaca T, Swanson BL, Spack EG, Wiener-Kronish JP. Therapeutic administration of anti-PcrV F(ab')(2) in sepsis associated with Pseudomonas aeruginosa. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:5880-6. [PMID: 11698464 DOI: 10.4049/jimmunol.167.10.5880] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The effects of rabbit-derived polyclonal Ab against PcrV, a protein involved in the translocation of type III secreted toxins of Pseudomonas aeruginosa, was investigated in two animal models of P. aeruginosa sepsis. In a mouse survival study, the i.v. administration of anti-PcrV IgG after the airspace instillation of a lethal dose of P. aeruginosa resulted in the complete survival of the animals. In a rabbit model of septic shock associated with Pseudomonas-induced lung injury, animals treated with anti-PcrV IgG intratracheally or i.v. had significant decreases in lung injury, bacteremia, and plasma TNF-alpha and significant improvement in the hemodynamic parameters associated with shock compared with animals treated in a similar manner with nonspecific control IgG. The administration of anti-PcrV F(ab')(2) showed protective effects comparable to those of whole anti-PcrV IgG. These results document that the therapeutic administration of anti-PcrV IgG blocks the type III secretion system-mediated virulence of P. aeruginosa and prevents septic shock and death, and that these protective effects are largely Fc independent. We conclude that Ab therapy neutralizing the type III secretion system has significant potential against lethal P. aeruginosa infections.
Collapse
Affiliation(s)
- N Shime
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
McMorran BJ, Palmer JS, Lunn DP, Oceandy D, Costelloe EO, Thomas GR, Hume DA, Wainwright BJ. G551D CF mice display an abnormal host response and have impaired clearance of Pseudomonas lung disease. Am J Physiol Lung Cell Mol Physiol 2001; 281:L740-7. [PMID: 11504703 DOI: 10.1152/ajplung.2001.281.3.l740] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Several cystic fibrosis (CF) mouse models demonstrate an increased susceptibility to Pseudomonas aeruginosa lung infection, characterized by excessive inflammation and high rates of mortality. Here we developed a model of chronic P. aeruginosa lung disease in mice homozygous for the murine CF transmembrane conductance regulator G551D mutation that provides an excellent model for CF lung disease. After 3 days of infection with mucoid P. aeruginosa entrapped in agar beads, the G551D animals lost substantially more body weight than non-CF control animals and were less able to control the infection, harboring over 40-fold more bacteria in the lung. The airways of infected G551D animals contained altered concentrations of the inflammatory mediators tumor necrosis factor-alpha, KC/N51, and macrophage inflammatory protein-2 during the first 2 days of infection, suggesting that an ineffective inflammatory response is partly responsible for the clearance defect.
Collapse
Affiliation(s)
- B J McMorran
- Institute for Molecular Biosciences, University of Queensland, St. Lucia, Queensland 4072, Australia
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Chroneos ZC, Wert SE, Livingston JL, Hassett DJ, Whitsett JA. Role of cystic fibrosis transmembrane conductance regulator in pulmonary clearance of Pseudomonas aeruginosa in vivo. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:3941-50. [PMID: 11034402 DOI: 10.4049/jimmunol.165.7.3941] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cystic fibrosis (CF)2 is a fatal genetic disease caused by mutations in the CF transmembrane conductance regulator (CFTR) that is commonly associated with chronic pulmonary infections with mucoid Pseudomonas aeruginosa (PA). To test the hypothesis that CFTR plays a direct role in PA adhesion and clearance, we have used mouse lines expressing varying levels of human (h) or mouse (m) CFTR. A subacute intratracheal dose of 3 x 10(6) bacteria was cleared with similar kinetics in control wild-type (WT) and transgenic mice overexpressing hCFTR in the lung from the surfactant protein C (SP-C) promoter (SP-C-hCFTR+/-). In a second series of experiments, the clearance of an acute intratracheal dose of 1.5 x 10(7) PA bacteria was also similar in WT, hemizygous SP-C-hCFTR+/-, and bitransgenic gut-corrected FABP-hCFTR+/+-mCFTR-/-, the latter lacking expression of mCFTR in the lung. However, a small but significant decrease in bacterial killing was observed in lungs of homozygote SP-C-hCFTR+/+ mice. Lung pathology in both WT and SP-C-hCFTR+/+ mice was marked by neutrophilic inflammation and bacterial invasion of perivascular and subepithelial compartments. Bacteria were associated primarily with leukocytes and were not associated with alveolar type II or bronchiolar epithelial cells, the cellular sites of SP-C-hCFTR+/+ transgene expression. The results indicate that there is no direct correlation between levels of CFTR expression and bacterial clearance or association of bacteria with epithelial cells in vivo.
Collapse
MESH Headings
- Animals
- Bacterial Adhesion/genetics
- Bacterial Adhesion/immunology
- Cystic Fibrosis Transmembrane Conductance Regulator/biosynthesis
- Cystic Fibrosis Transmembrane Conductance Regulator/deficiency
- Cystic Fibrosis Transmembrane Conductance Regulator/genetics
- Cystic Fibrosis Transmembrane Conductance Regulator/physiology
- Interleukin-1/metabolism
- Intubation, Intratracheal
- Lung/immunology
- Lung/metabolism
- Lung/microbiology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Inbred Strains
- Mice, Transgenic
- Pneumonia, Bacterial/genetics
- Pneumonia, Bacterial/metabolism
- Pneumonia, Bacterial/microbiology
- Pneumonia, Bacterial/pathology
- Proteolipids/biosynthesis
- Proteolipids/genetics
- Pseudomonas Infections/genetics
- Pseudomonas Infections/metabolism
- Pseudomonas Infections/microbiology
- Pseudomonas Infections/pathology
- Pseudomonas aeruginosa/physiology
- Pulmonary Surfactants/biosynthesis
- Pulmonary Surfactants/genetics
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Z C Chroneos
- Children's Hospital Medical Center, Division of Pulmonary Biology, Cincinnati, OH 45229, USA
| | | | | | | | | |
Collapse
|
42
|
Thomas LD, Dunkley ML, Moore R, Reynolds S, Bastin DA, Kyd JM, Cripps AW. Catalase immunization from Pseudomonas aeruginosa enhances bacterial clearance in the rat lung. Vaccine 2000; 19:348-57. [PMID: 10930690 DOI: 10.1016/s0264-410x(00)00146-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Pseudomonas aeruginosa is a common cause of infection in immunocompromised patients and is the major contributor to morbidity in individuals with cystic fibrosis (CF). The antibiotic resistance shown by this pathogen and morbidity in patients with chronic infection has encouraged investigations into the development of a vaccine. This study reports the purification of a 60 kDa protein, isolated from a mucoid strain of P. aeruginosa, identified by amino acid sequence analysis as the catalase protein (KatA). A rat model of acute P. aeruginosa respiratory infection was used to investigate the immunogenicity of KatA and determine the potential of mucosal immunization with KatA to protect against infection. Immunization regimens compared a single intra-Peyer's patch (IPP) immunization with an IPP primary inoculation followed by an intratracheal boost to the lungs. Mucosal immunization with KatA resulted in significant pulmonary clearance of both homologous (p<0.001) and heterologous (p<0.05) strains of P. aeruginosa. Both immunization regimens enhanced bacterial clearance, increased the rate of recruitment of phagocytes to the bronchoalveoli and induced KatA-specific antibody. However, the regimen that included a boost induced a more effective immune response that also resulted in better clearance of P. aeruginosa from the lungs. Mucosal immunization induced KatA- specific antibodies in the serum and the bronchoalveolar lavage, and KatA-specific lymphocyte proliferation in vitro in cells isolated from the mesenteric lymph nodes of immunized rats. The data presented suggests that KatA has the potential to afford a protective immune response against pulmonary infection by P. aeruginosa
Collapse
Affiliation(s)
- L D Thomas
- Gadi Research Centre, Division of Science and Design, University of Canberra, Australia
| | | | | | | | | | | | | |
Collapse
|
43
|
St-Denis A, Caouras V, Gervais F, Descoteaux A. Role of Protein Kinase C-α in the Control of Infection by Intracellular Pathogens in Macrophages. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.10.5505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
The protein kinase C (PKC) family regulates macrophage function involved in host defense against infection. In this study, we investigated the role of macrophage PKC-α in the uptake and subsequent fate of Leishmania donovani promastigotes and Legionella pneumophila infections. To this end, we used clones of the murine macrophage cell line RAW 264.7 overexpressing a dominant-negative (DN) mutant of PKC-α. While phagocytosis of L. donovani promastigotes was not affected by DN PKC-α overexpression, their intracellular survival was enhanced by 10- to 20-fold at 48 h postinfection. Intracellular survival of a L. donovani mutant defective in lipophosphoglycan repeating units synthesis, which normally is rapidly degraded in phagolysosomes, was enhanced by 100-fold at 48 h postinfection. However, IFN-γ-induced leishmanicidal activity was not affected by DN PKC-α overexpression. Similar to macrophages from genetically resistant C57BL/6 mice, control RAW 264.7 cells were not permissive for the intracellular replication of Legionella pneumophila. In contrast, DN PKC-α-overexpressing RAW 264.7 clones were phenotypically similar to macrophages from genetically susceptible A/J mice, as they allowed intracellular replication of L. pneumophila. Permissiveness to L. pneumophila was not the consequence of a general defect in the microbicidal capacities because killing of a temperature-sensitive mutant of Pseudomonas aeruginosa was normal in DN PKC-α-overexpressing RAW 264.7 clones. Collectively, these results support a role for PKC-α in the regulation of innate macrophage functions involved in the control of infection by intracellular parasites.
Collapse
Affiliation(s)
- Anik St-Denis
- *INRS-Institut Armand-Frappier, Université du Québec, Laval, Québec, Canada; and
| | - Vassiliki Caouras
- †Centre for the Study of Host Resistance, McGill University, Montréal, Québec, Canada
| | - Francine Gervais
- †Centre for the Study of Host Resistance, McGill University, Montréal, Québec, Canada
| | - Albert Descoteaux
- *INRS-Institut Armand-Frappier, Université du Québec, Laval, Québec, Canada; and
| |
Collapse
|
44
|
Tam M, Snipes GJ, Stevenson MM. Characterization of chronic bronchopulmonary Pseudomonas aeruginosa infection in resistant and susceptible inbred mouse strains. Am J Respir Cell Mol Biol 1999; 20:710-9. [PMID: 10101003 DOI: 10.1165/ajrcmb.20.4.3223] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Chronic bronchopulmonary Pseudomonas aeruginosa infection, initiated by intratracheal instillation of 1 to 2 x 10(5) colony-forming units of a mucoid strain of bacteria trapped in agar beads, was characterized in resistant BALB/c mice and susceptible C57BL/6 (B6) mice through 28 d postinfection. B6 mice experienced a more severe infection than BALB/c mice as evidenced by significantly higher mortality and significantly greater weight loss during the first 14 d. Furthermore, B6 mice had significantly higher numbers of bacteria in the lungs through 21 d after infection. Overall, only 22% of these hosts cleared the infection. In contrast, 67% of BALB/c mice cleared the infection. These differences between resistant and susceptible mice were found to correlate with histopathologic differences in the type of inflammation and the extent of tissue damage. An acute, predominantly neutrophilic inflammation and extensive tissue damage were apparent in the lungs of susceptible B6 mice, whereas chronic, granulomatous inflammation and little or no tissue damage were visible in resistant BALB/c mice. The finding of acute inflammation in the lungs of infected B6 mice was confirmed by fluorescence-activated cell sorter (FACS) analyses, which demonstrated that these mice had significantly greater proportions of polymorphonuclear neutrophils in the lungs on Days 7 and 14 after infection than did BALB/c mice. FACS analyses also revealed significant and similar increases in CD3(+) lung cells in both strains as the infection progressed. The CD4/CD8 ratio was significantly greater in BALB/c mice by 21 d after infection when the majority of these animals, but not B6 mice, had cleared the infection.
Collapse
Affiliation(s)
- M Tam
- Centre for the Study of Host Resistance, Montreal General Hospital Research Institute; and Department of Neuropathology, Montreal, Canada
| | | | | |
Collapse
|
45
|
King BK, Kudsk KA, Li J, Wu Y, Renegar KB. Route and type of nutrition influence mucosal immunity to bacterial pneumonia. Ann Surg 1999; 229:272-8. [PMID: 10024110 PMCID: PMC1191641 DOI: 10.1097/00000658-199902000-00016] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To develop a model of established respiratory immunity against Pseudomonas aeruginosa pneumonia and to investigate the effects of route and type of nutrition on this immunity. SUMMARY BACKGROUND DATA Diet influences the ability of gut-associated lymphoid tissue (GALT) to maintain mucosal immunity. Complex enteral diets and chow maintain normal GALT populations against established IgA-mediated antiviral respiratory immunity. Both intravenous and intragastric total parenteral nutrition (TPN) produce GALT atrophy, but only intragastric TPN preserves established antiviral immunity. The authors hypothesized that both GALT-depleting diets (intragastric and intravenous TPN) would impair immunity against bacterial pneumonia. METHODS P. aeruginosa was administered intratracheally to determine the mortality rate at increasing doses, and liposomes containing P. aeruginosa antigens were used to generate effective respiratory immunization. In the final experiment, mice received liposomes containing P. aeruginosa antigens to establish immunity and then were randomized to chow, complex enteral diets, intragastric TPN, or intravenous TPN. After 5 days of diet, mice received live intratracheal P. aeruginosa, and the death rate was recorded at 24 and 48 hours. RESULTS The LD50 and LD100 were 9 x 10(7) and 12 x 10(7), respectively. Immunization reduced the mortality rate from 66% to 12%. This immunization was maintained in mice fed chow or a complex enteral diet and was lost in animals receiving intravenous TPN. Intragastric TPN partially preserved this respiratory immunity. CONCLUSIONS Protection against bacterial pneumonia can be induced by prior antigenic immunization. This protection is lost with intravenous TPN, partially preserved with a chemically defined enteral diet, and completely preserved with chow or complex enteral diets. Both route and type of nutrition influence antibacterial respiratory tract immunity.
Collapse
Affiliation(s)
- B K King
- Department of Surgery, The University of Tennessee, Memphis, USA
| | | | | | | | | |
Collapse
|
46
|
Sapru K, Stotland PK, Stevenson MM. Quantitative and qualitative differences in bronchoalveolar inflammatory cells in Pseudomonas aeruginosa-resistant and -susceptible mice. Clin Exp Immunol 1999; 115:103-9. [PMID: 9933427 PMCID: PMC1905184 DOI: 10.1046/j.1365-2249.1999.00762.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The difference in severity of Pseudomonas aeruginosa-induced chronic lung infection may be determined by differences in host inflammatory responses. In the present study we investigate this possibility using BALB/c and C57Bl/6 mice, resistant and susceptible, respectively, to chronic lung infection with P. aeruginosa. Following intratracheal inoculation of P. aeruginosa-impregnated agar beads, C57Bl/6 mice mounted a stronger inflammatory response with significantly higher total cell numbers in the bronchoalveolar lavage fluid compared with BALB/c mice. While polymorphonuclear leucocytes were the predominant cell in C57Bl/6 mice, macrophages constituted the majority in BALB/c mice at day 7 post-infection. Alveolar macrophages from C57Bl/6 mice showed significantly higher spontaneous production of nitric oxide (NO) at day 7 post-infection compared with BALB/c mice. Following in vitro stimulation with heat-killed Pseudomonas antigen, these cells produced significantly higher NO compared with cells from BALB/c mice at day 21 post-infection. Production of tumour necrosis factor-alpha (TNF-alpha) by alveolar macrophages was significantly higher at day 7 in BALB/c mice compared with C57Bl/6 mice, which showed significantly higher levels at day 28 post-infection. Taken together, these results suggest that defects in the host inflammatory process contribute to the variable outcome of chronic lung infection with P. aeruginosa. An exaggerated inflammatory response dominated by polymorphonuclear cells correlates with susceptibility to infection, whilst a modest inflammatory response dominated by macrophages correlates with resistance. Moreover, the quantity and timing of production of NO and TNF-alpha by alveolar macrophages may modulate the course and outcome of infection.
Collapse
Affiliation(s)
- K Sapru
- Centre for the Study of Host Resistance, McGill University, Montreal, Quebec, Canada
| | | | | |
Collapse
|
47
|
Baker DG. Natural pathogens of laboratory mice, rats, and rabbits and their effects on research. Clin Microbiol Rev 1998; 11:231-66. [PMID: 9564563 PMCID: PMC106832 DOI: 10.1128/cmr.11.2.231] [Citation(s) in RCA: 226] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Laboratory mice, rats, and rabbits may harbor a variety of viral, bacterial, parasitic, and fungal agents. Frequently, these organisms cause no overt signs of disease. However, many of the natural pathogens of these laboratory animals may alter host physiology, rendering the host unsuitable for many experimental uses. While the number and prevalence of these pathogens have declined considerably, many still turn up in laboratory animals and represent unwanted variables in research. Investigators using mice, rats, and rabbits in biomedical experimentation should be aware of the profound effects that many of these agents can have on research.
Collapse
Affiliation(s)
- D G Baker
- Division of Laboratory Animal Medicine, School of Veterinary Medicine, Louisiana State University, Baton Rouge 70810, USA.
| |
Collapse
|
48
|
Yu H, Hanes M, Chrisp CE, Boucher JC, Deretic V. Microbial pathogenesis in cystic fibrosis: pulmonary clearance of mucoid Pseudomonas aeruginosa and inflammation in a mouse model of repeated respiratory challenge. Infect Immun 1998; 66:280-8. [PMID: 9423869 PMCID: PMC107888 DOI: 10.1128/iai.66.1.280-288.1998] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Chronic endobronchiolitis compounded by recurring Pseudomonas aeruginosa infections is the major cause of morbidity and mortality in patients with cystic fibrosis (CF). In this study, a mouse model of repeated respiratory exposure to P. aeruginosa was established to facilitate investigations of factors contributing to P. aeruginosa persistence and associated inflammatory processes in the lung. While a single exposure to P. aeruginosa aerosols resulted in only mild histopathological changes, repeated exposure caused significant lung pathology in C57BL/6J mice. The peak of histopathological changes and inflammation in C57BL/6J mice was characterized by subacute lymphohistiocytic bronchopneumonia and persistent elevation of tumor necrosis factor alpha and macrophage inflammatory protein 2 in the lung but not in the serum. When isogenic nonmucoid (mucA+) and mucoid (mucA22) P. aeruginosa strains were compared, the mucoid cells were cleared several-fold less efficiently than the parental nonmucoid strain during the initial stages of the aerosol exposure regimen. However, the microscopic pathology findings and proinflammatory cytokine levels were similar in mice exposed to nonmucoid and mucoid P. aeruginosa throughout the infection. We also tested lung histopathology and proinflammatory cytokines in interleukin 10 (IL-10)-deficient transgenic (IL-10T) mice. Significant mortality was seen in IL-10T mice on initial challenge with P. aeruginosa, although no histopathological differences could be observed in the lungs of C57BL/6J and surviving IL-10T mice after a single exposure. However, increased pathology was detected in IL-10T mice relative to C57BL/6J after repeated challenge with P. aeruginosa. This observation supports the proposals that anti-inflammatory cytokines may play a role in suppressing P. aeruginosa-induced tissue damage during chronic infection.
Collapse
Affiliation(s)
- H Yu
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor 48109, USA
| | | | | | | | | |
Collapse
|
49
|
Telford G, Wheeler D, Williams P, Tomkins PT, Appleby P, Sewell H, Stewart GS, Bycroft BW, Pritchard DI. The Pseudomonas aeruginosa quorum-sensing signal molecule N-(3-oxododecanoyl)-L-homoserine lactone has immunomodulatory activity. Infect Immun 1998; 66:36-42. [PMID: 9423836 PMCID: PMC107855 DOI: 10.1128/iai.66.1.36-42.1998] [Citation(s) in RCA: 297] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Diverse gram-negative bacterial cells communicate with each other by using diffusible N-acyl homoserine lactone (AHL) signal molecules to coordinate gene expression with cell population density. Accumulation of AHLs above a threshold concentration renders the population "quorate," and the appropriate target gene is activated. In pathogenic bacteria, such as Pseudomonas aeruginosa, AHL-mediated quorum sensing is involved in the regulation of multiple virulence determinants. We therefore sought to determine whether the immune system is capable of responding to these bacterial signal molecules. Consequently the immunomodulatory properties of the AHLs N-(3-oxododecanoyl)-L-homoserine lactone (OdDHL) and N-(3-oxohexanoyl)-L-homoserine lactone (OHHL) were evaluated in murine and human leukocyte immunoassays in vitro. OdDHL, but not OHHL, inhibited lymphocyte proliferation and tumor necrosis factor alpha production by lipopolysaccharide-stimulated macrophages. Furthermore, OdDHL simultaneously and potently down-regulated the production of IL-12, a Th-1-supportive cytokine. At high concentrations (>7 x 10(-5) M) OdDHL inhibited antibody production by keyhole limpet hemocyanin-stimulated spleen cells, but at lower concentrations (<7 x 10(-5) M), antibody production was stimulated, apparently by increasing the proportion of the immunoglobulin G1 (IgG1) isotype. OdDHL also promoted IgE production by interleukin-4-stimulated human peripheral blood mononuclear cells. These data indicate that OdDHL may influence the Th-1-Th-2 balance in the infected host and suggest that, in addition to regulating the expression of virulence determinants, OdDHL may contribute to the pathogenesis of P. aeruginosa infections by functioning as a virulence determinant per se.
Collapse
Affiliation(s)
- G Telford
- Department of Life Science, University of Nottingham, University Park, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Boucher JC, Yu H, Mudd MH, Deretic V. Mucoid Pseudomonas aeruginosa in cystic fibrosis: characterization of muc mutations in clinical isolates and analysis of clearance in a mouse model of respiratory infection. Infect Immun 1997; 65:3838-46. [PMID: 9284161 PMCID: PMC175548 DOI: 10.1128/iai.65.9.3838-3846.1997] [Citation(s) in RCA: 208] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
A distinguishing feature of Pseudomonas aeruginosa isolates from cystic fibrosis (CF) patients is their mucoid, exopolysaccharide alginate-overproducing phenotype. One mechanism of conversion to mucoidy is based on mutations in the algU mucABCD cluster, encoding the stress sigma factor AlgU and its regulators. However, conversion to mucoidy in laboratory strains can be achieved via mutations in other chromosomal sites. Here, we investigated mechanisms of the emergence of mucoid P. aeruginosa in CF by analyzing the status of mucA in a collection of mucoid P. aeruginosa isolates from 53 CF patients. This negative regulator of algU, when inactivated under laboratory conditions, causes conversion to mucoidy. The overall frequency of mucA alterations in mucoid CF isolates was 84%. Nucleotide sequence analyses revealed that the majority of the alterations caused premature termination of the mucA coding sequence. Comparison of paired nonmucoid and mucoid P. aeruginosa isolates from three CF patients indicated the presence of mucA mutations only in the mucoid strains. Interestingly, mucoid P. aeruginosa isolates from urinary tract infections also had mutations in the mucA gene. Clearance of CF isolates from the murine lung was investigated in an aerosol infection model with C57BL/6J, BALB/c, and DBA/2NHsd mice. Two CF strains, selected for further study based on the dependence of their alginate production on the concentration of salt in the medium, were used to examine the effects of mucoidy on pulmonary clearance. Statistically significant improvement in recovery from the murine lung of viable mucoid P. aeruginosa cells relative to the nonmucoid bacteria was observed in the majority of mouse strains tested. Collectively, the results reported here suggest that mucA is most likely the preferential site for conversion to mucoidy in CF and that alginate overproduction in mucA-mutant P. aeruginosa improves its resistance to the innate clearance mechanisms in the lung.
Collapse
Affiliation(s)
- J C Boucher
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor 48109-0620, USA
| | | | | | | |
Collapse
|