1
|
Nasreen M, Ellis D, Hosmer J, Essilfie AT, Fantino E, Sly P, McEwan AG, Kappler U. The DmsABC S-oxide reductase is an essential component of a novel, hypochlorite-inducible system of extracellular stress defense in Haemophilus influenzae. Front Microbiol 2024; 15:1359513. [PMID: 38638903 PMCID: PMC11024254 DOI: 10.3389/fmicb.2024.1359513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/11/2024] [Indexed: 04/20/2024] Open
Abstract
Defenses against oxidative damage to cell components are essential for survival of bacterial pathogens during infection, and here we have uncovered that the DmsABC S-/N-oxide reductase is essential for virulence and in-host survival of the human-adapted pathogen, Haemophilus influenzae. In several different infection models, H. influenzae ΔdmsA strains showed reduced immunogenicity as well as lower levels of survival in contact with host cells. Expression of DmsABC was induced in the presence of hypochlorite and paraquat, closely linking this enzyme to defense against host-produced antimicrobials. In addition to methionine sulfoxide, DmsABC converted nicotinamide- and pyrimidine-N-oxide, precursors of NAD and pyrimidine for which H. influenzae is an auxotroph, at physiologically relevant concentrations, suggesting that these compounds could be natural substrates for DmsABC. Our data show that DmsABC forms part of a novel, periplasmic system for defense against host-induced S- and N-oxide stress that also comprises the functionally related MtsZ S-oxide reductase and the MsrAB peptide methionine sulfoxide reductase. All three enzymes are induced following exposure of the bacteria to hypochlorite. MsrAB is required for physical resistance to HOCl and protein repair. In contrast, DmsABC was required for intracellular colonization of host cells and, together with MtsZ, contributed to resistance to N-Chlorotaurine. Our work expands and redefines the physiological role of DmsABC and highlights the importance of different types of S-oxide reductases for bacterial virulence.
Collapse
Affiliation(s)
- Marufa Nasreen
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, QLD, Australia
| | - Daniel Ellis
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, QLD, Australia
| | - Jennifer Hosmer
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, QLD, Australia
| | | | | | - Peter Sly
- Child Health Research Centre, South Brisbane, QLD, Australia
| | - Alastair G. McEwan
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, QLD, Australia
| | - Ulrike Kappler
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, QLD, Australia
| |
Collapse
|
2
|
Zahid A, Wilson JC, Grice ID, Peak IR. Otitis media: recent advances in otitis media vaccine development and model systems. Front Microbiol 2024; 15:1345027. [PMID: 38328427 PMCID: PMC10847372 DOI: 10.3389/fmicb.2024.1345027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/08/2024] [Indexed: 02/09/2024] Open
Abstract
Otitis media is an inflammatory disorder of the middle ear caused by airways-associated bacterial or viral infections. It is one of the most common childhood infections as globally more than 80% of children are diagnosed with acute otitis media by 3 years of age and it is a common reason for doctor's visits, antibiotics prescriptions, and surgery among children. Otitis media is a multifactorial disease with various genetic, immunologic, infectious, and environmental factors predisposing children to develop ear infections. Streptococcus pneumoniae, Haemophilus influenzae, and Moraxella catarrhalis are the most common culprits responsible for acute otitis media. Despite the massive global disease burden, the pathogenesis of otitis media is still unclear and requires extensive future research. Antibiotics are the preferred treatment to cure middle ear infections, however, the antimicrobial resistance rate of common middle ear pathogens has increased considerably over the years. At present, pneumococcal and influenza vaccines are administered as a preventive measure against otitis media, nevertheless, these vaccines are only beneficial in preventing carriage and/or disease caused by vaccine serotypes. Otitis media caused by non-vaccine serotype pneumococci, non-typeable H. influenza, and M. catarrhalis remain an important healthcare burden. The development of multi-species vaccines is an arduous process but is required to reduce the global burden of this disease. Many novel vaccines against S. pneumoniae, non-typeable H. influenza, and M. catarrhalis are in preclinical trials. It is anticipated that these vaccines will lower the disease burden and provide better protection against otitis media. To study disease pathology the rat, mouse, and chinchilla are commonly used to induce experimental acute otitis media to test new therapeutics, including antibiotics and vaccines. Each of these models has its advantages and disadvantages, yet there is still a need to develop an improved animal model providing a better correlated mechanistic understanding of human middle ear infections, thereby underpinning the development of more effective otitis media therapeutics. This review provides an updated summary of current vaccines against otitis media, various animal models of otitis media, their limitations, and some future insights in this field providing a springboard in the development of new animal models and novel vaccines for otitis media.
Collapse
Affiliation(s)
- Ayesha Zahid
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Jennifer C. Wilson
- School of Pharmacy and Medical Science, Griffith University, Gold Coast, QLD, Australia
| | - I. Darren Grice
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy and Medical Science, Griffith University, Gold Coast, QLD, Australia
| | - Ian R. Peak
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy and Medical Science, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
3
|
Lee H, Edgar RJ, Lichtenstein IJ, Velarde JJ, Korotkova N, Wessels MR. Streptococcus pyogenes can support or inhibit growth of Haemophilus influenzae by supplying or restricting extracellular NAD+. PLoS One 2022; 17:e0270697. [PMID: 36170255 PMCID: PMC9518897 DOI: 10.1371/journal.pone.0270697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/15/2022] [Indexed: 11/19/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential co-factor for cellular metabolism and serves as a substrate in enzymatic processes. NAD+ is produced by de novo synthesis or salvage pathways in nearly all bacterial species. Haemophilus influenzae lacks the capacity for de novo synthesis, so it is dependent on import of NAD+ from the external environment or salvage biosynthetic pathways for recycling of NAD+ precursors and breakdown products. However, the actual sources of NAD+ utilized by H. influenzae in the respiratory tract are not well defined. In this study, we found that a variety of bacteria, including species found in the upper airway of humans, released NAD+ that was readily detectable in extracellular culture fluid, and which supported growth of H. influenzae in vitro. By contrast, certain strains of Streptococcus pyogenes (group A streptococcus or GAS) inhibited growth of H. influenzae in vitro by secreting NAD+-glycohydrolase (NADase), which degraded extracellular NAD+. Conversely, GAS strains that lacked enzymatically active NADase released extracellular NAD+, which could support H. influenzae growth. Our results suggest that many bacterial species, including normal flora of the upper airway, release NAD+ into the environment. GAS is distinctive in its ability to both release and degrade NAD+. Thus, colonization of the airway with H. influenzae may be promoted or restricted by co-colonization with GAS in a strain-specific manner that depends, respectively, on release of NAD+ or secretion of active NADase. We suggest that, in addition to its role as a cytotoxin for host cells, NADase may serve a separate function by restricting growth of H. influenzae in the human respiratory tract.
Collapse
Affiliation(s)
- Hyunju Lee
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Pediatrics, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Rebecca J. Edgar
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Ian J. Lichtenstein
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jorge J. Velarde
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Natalia Korotkova
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, Kentucky, United States of America
| | - Michael R. Wessels
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
4
|
Nasreen M, Nair RP, McEwan AG, Kappler U. The Peptide Methionine Sulfoxide Reductase (MsrAB) of Haemophilus influenzae Repairs Oxidatively Damaged Outer Membrane and Periplasmic Proteins Involved in Nutrient Acquisition and Virulence. Antioxidants (Basel) 2022; 11:antiox11081557. [PMID: 36009276 PMCID: PMC9404787 DOI: 10.3390/antiox11081557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/06/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Sulfoxide-damage repair mechanisms are emerging as essential for the virulence of bacterial pathogens, and in the human respiratory pathogen Haemophilus influenzae the periplasmic MsrAB peptide methionine sulfoxide reductase is necessary for resistance to reactive chlorine species such as hypochlorite. Additionally, this enzyme has a role in modulating the host immune response to infection. Here, we have analysed the enzymatic properties of MsrAB, which revealed that both domains of the protein are catalytically active, with the turnover number of the MsrA domain being 50% greater than that for the MsrB domain. MsrAB was active with small molecular sulfoxides as well as oxidised calmodulin, and maximal activity was observed at 30°C, a temperature close to that found in the natural niche of H. influenzae, the nasopharynx. Analyses of differential methionine oxidation identified 29 outer membrane and periplasmic proteins that are likely substrates for MsrAB. These included the LldD lactate dehydrogenase and the lipoprotein eP4 that is involved in NAD and hemin metabolism in H. influenzae. Subsequent experiments showed that H. influenzae MsrAB can repair oxidative damage to methionines in purified eP4 with up to 100% efficiency. Our work links MsrAB to the maintenance of different adhesins and essential metabolic processes in the H. influenzae, such as NAD metabolism and access to L-lactate, which is a key growth substrate for H. influenzae during infection.
Collapse
|
5
|
Hosmer J, Nasreen M, Dhouib R, Essilfie AT, Schirra HJ, Henningham A, Fantino E, Sly P, McEwan AG, Kappler U. Access to highly specialized growth substrates and production of epithelial immunomodulatory metabolites determine survival of Haemophilus influenzae in human airway epithelial cells. PLoS Pathog 2022; 18:e1010209. [PMID: 35085362 PMCID: PMC8794153 DOI: 10.1371/journal.ppat.1010209] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 12/14/2021] [Indexed: 11/18/2022] Open
Abstract
Haemophilus influenzae (Hi) infections are associated with recurring acute exacerbations of chronic respiratory diseases in children and adults including otitis media, pneumonia, chronic obstructive pulmonary disease and asthma. Here, we show that persistence and recurrence of Hi infections are closely linked to Hi metabolic properties, where preferred growth substrates are aligned to the metabolome of human airway epithelial surfaces and include lactate, pentoses, and nucleosides, but not glucose that is typically used for studies of Hi growth in vitro. Enzymatic and physiological investigations revealed that utilization of lactate, the preferred Hi carbon source, required the LldD L-lactate dehydrogenase (conservation: 98.8% of strains), but not the two redox-balancing D-lactate dehydrogenases Dld and LdhA. Utilization of preferred substrates was directly linked to Hi infection and persistence. When unable to utilize L-lactate or forced to rely on salvaged guanine, Hi showed reduced extra- and intra-cellular persistence in a murine model of lung infection and in primary normal human nasal epithelia, with up to 3000-fold attenuation observed in competitive infections. In contrast, D-lactate dehydrogenase mutants only showed a very slight reduction compared to the wild-type strain. Interestingly, acetate, the major Hi metabolic end-product, had anti-inflammatory effects on cultured human tissue cells in the presence of live but not heat-killed Hi, suggesting that metabolic endproducts also influence HI-host interactions. Our work provides significant new insights into the critical role of metabolism for Hi persistence in contact with host cells and reveals for the first time the immunomodulatory potential of Hi metabolites.
Collapse
Affiliation(s)
- Jennifer Hosmer
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, Australia
| | - Marufa Nasreen
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, Australia
| | - Rabeb Dhouib
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, Australia
| | | | | | - Anna Henningham
- Child Health Research Centre, The University of Queensland, South Brisbane, Australia
| | - Emmanuelle Fantino
- Child Health Research Centre, The University of Queensland, South Brisbane, Australia
| | - Peter Sly
- Child Health Research Centre, The University of Queensland, South Brisbane, Australia
| | - Alastair G. McEwan
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, Australia
| | - Ulrike Kappler
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, Australia
| |
Collapse
|
6
|
Dhouib R, Nasreen M, Othman DSMP, Ellis D, Lee S, Essilfie AT, Hansbro PM, McEwan AG, Kappler U. The DmsABC Sulfoxide Reductase Supports Virulence in Non-typeable Haemophilus influenzae. Front Microbiol 2021; 12:686833. [PMID: 34367088 PMCID: PMC8340005 DOI: 10.3389/fmicb.2021.686833] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 06/21/2021] [Indexed: 11/15/2022] Open
Abstract
Although molybdenum-containing enzymes are well-established as having a key role in bacterial respiration, it is increasingly recognized that some may also support bacterial virulence. Here, we show that DmsABC, a putative dimethylsulfoxide (DMSO) reductase, is required for fitness of the respiratory pathogen Haemophilus influenzae (Hi) in different models of infection. Expression of the dmsABC operon increased with decreasing oxygen availability, but despite this, a Hi2019Δd msA strain did not show any defects in anaerobic growth on chemically defined medium (CDM), and viability was also unaffected. Although Hi2019Δd msA exhibited increased biofilm formation in vitro and greater resistance to hypochlorite killing compared to the isogenic wild-type strain, its survival in contact with primary human neutrophils, in infections of cultured tissue cells, or in a mouse model of lung infection was reduced compared to Hi2019WT. The tissue cell infection model revealed a two-fold decrease in intracellular survival, while in the mouse model of lung infection Hi2019Δd msA was strongly attenuated and below detection levels at 48 h post-inoculation. While Hi2019WT was recovered in approximately equal numbers from bronchoalveolar lavage fluid (BALF) and lung tissue, survival of Hi2019Δd msA was reduced in lung tissue compared to BALF samples, indicating that Hi2019Δd msA had reduced access to or survival in the intracellular niche. Our data clearly indicate for the first time a role for DmsABC in H. influenzae infection and that the conditions under which DmsABC is required in this bacterium are closely linked to interactions with the host.
Collapse
Affiliation(s)
- Rabeb Dhouib
- School of Chemistry and Molecular Biosciences, Australian Infectious Disease Research Centre, The University of Queensland, St Lucia, QLD, Australia
| | - Marufa Nasreen
- School of Chemistry and Molecular Biosciences, Australian Infectious Disease Research Centre, The University of Queensland, St Lucia, QLD, Australia
| | - Dk Seti Maimonah Pg Othman
- School of Chemistry and Molecular Biosciences, Australian Infectious Disease Research Centre, The University of Queensland, St Lucia, QLD, Australia
| | - Daniel Ellis
- School of Chemistry and Molecular Biosciences, Australian Infectious Disease Research Centre, The University of Queensland, St Lucia, QLD, Australia
| | - Simon Lee
- School of Chemistry and Molecular Biosciences, Australian Infectious Disease Research Centre, The University of Queensland, St Lucia, QLD, Australia
| | | | - Philip M. Hansbro
- Centre for Inflammation, Centenary Institute, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Alastair G. McEwan
- School of Chemistry and Molecular Biosciences, Australian Infectious Disease Research Centre, The University of Queensland, St Lucia, QLD, Australia
| | - Ulrike Kappler
- School of Chemistry and Molecular Biosciences, Australian Infectious Disease Research Centre, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
7
|
Nasreen M, Fletcher A, Hosmer J, Zhong Q, Essilfie AT, McEwan AG, Kappler U. The Alternative Sigma Factor RpoE2 Is Involved in the Stress Response to Hypochlorite and in vivo Survival of Haemophilus influenzae. Front Microbiol 2021; 12:637213. [PMID: 33643271 PMCID: PMC7907618 DOI: 10.3389/fmicb.2021.637213] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/07/2021] [Indexed: 11/29/2022] Open
Abstract
Extracytoplasmic function (ECF) sigma factors underpin the ability of bacteria to adapt to changing environmental conditions, a process that is particularly relevant in human pathogens that inhabit niches where human immune cells contribute to high levels of extracellular stress. Here, we have characterized the previously unstudied RpoE2 ECF sigma factor from the human respiratory pathogen H. influenzae (Hi) and its role in hypochlorite-induced stress. Exposure of H. influenzae to oxidative stress (HOCl, H2O2) increased rpoE2 gene expression, and the activity of RpoE2 was controlled by a cytoplasmic 67-aa anti-sigma factor, HrsE. RpoE2 regulated the expression of the periplasmic MsrAB peptide methionine sulfoxide reductase that, in H. influenzae, is required for HOCl resistance, thus linking RpoE2 to HOCl stress. Interestingly, a HiΔrpoE2 strain had wild-type levels of resistance to oxidative stress in vitro, but HiΔrpoE2 survival was reduced 26-fold in a mouse model of lung infection, demonstrating the relevance of this sigma factor for H. influenzae pathogenesis. The HiRpoE2 system has some similarity to the ECF sigma factors described in Streptomyces and Neisseria sp. that also control the expression of msr genes. However, HiRpoE2 regulation extended to genes encoding other periplasmic damage repair proteins, an operon containing a DoxX-like protein, and also included selected OxyR-controlled genes. Based on our results, we propose that the highly conserved HiRpoE2 sigma factor is a key regulator of H. influenzae responses to oxidative damage in the cell envelope region that controls a variety of target genes required for survival in the host.
Collapse
Affiliation(s)
- Marufa Nasreen
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia
| | - Aidan Fletcher
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia
| | - Jennifer Hosmer
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia
| | - Qifeng Zhong
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia
| | | | - Alastair G McEwan
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia
| | - Ulrike Kappler
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia
| |
Collapse
|
8
|
Exploring the Impact of Ketodeoxynonulosonic Acid in Host-Pathogen Interactions Using Uptake and Surface Display by Nontypeable Haemophilus influenzae. mBio 2021; 12:mBio.03226-20. [PMID: 33468699 PMCID: PMC7845648 DOI: 10.1128/mbio.03226-20] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
All cells in vertebrates are coated with a dense array of glycans often capped with sugars called sialic acids. Sialic acids have many functions, including serving as a signal for recognition of “self” cells by the immune system, thereby guiding an appropriate immune response against foreign “nonself” and/or damaged cells. Surface expression of the common vertebrate sialic acid (Sia) N-acetylneuraminic acid (Neu5Ac) by commensal and pathogenic microbes appears structurally to represent “molecular mimicry” of host sialoglycans, facilitating multiple mechanisms of host immune evasion. In contrast, ketodeoxynonulosonic acid (Kdn) is a more ancestral Sia also present in prokaryotic glycoconjugates that are structurally quite distinct from vertebrate sialoglycans. We detected human antibodies against Kdn-terminated glycans, and sialoglycan microarray studies found these anti-Kdn antibodies to be directed against Kdn-sialoglycans structurally similar to those on human cell surface Neu5Ac-sialoglycans. Anti-Kdn-glycan antibodies appear during infancy in a pattern similar to those generated following incorporation of the nonhuman Sia N-glycolylneuraminic acid (Neu5Gc) onto the surface of nontypeable Haemophilus influenzae (NTHi), a human commensal and opportunistic pathogen. NTHi grown in the presence of free Kdn took up and incorporated the Sia into its lipooligosaccharide (LOS). Surface display of the Kdn within NTHi LOS blunted several virulence attributes of the pathogen, including Neu5Ac-mediated resistance to complement and whole blood killing, complement C3 deposition, IgM binding, and engagement of Siglec-9. Upper airway administration of Kdn reduced NTHi infection in human-like Cmah null (Neu5Gc-deficient) mice that express a Neu5Ac-rich sialome. We propose a mechanism for the induction of anti-Kdn antibodies in humans, suggesting that Kdn could be a natural and/or therapeutic “Trojan horse” that impairs colonization and virulence phenotypes of free Neu5Ac-assimilating human pathogens.
Collapse
|
9
|
Heo M, Chenon G, Castrillon C, Bibette J, Bruhns P, Griffiths AD, Baudry J, Eyer K. Deep phenotypic characterization of immunization-induced antibacterial IgG repertoires in mice using a single-antibody bioassay. Commun Biol 2020; 3:614. [PMID: 33106526 PMCID: PMC7589517 DOI: 10.1038/s42003-020-01296-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Antibodies with antibacterial activity need to bind to the bacterial surface with affinity, specificity, and sufficient density to induce efficient elimination. To characterize the anti-bacterial antibody repertoire, we developed an in-droplet bioassay with single-antibody resolution. The assay not only allowed us to identify whether the secreted antibodies recognized a bacterial surface antigen, but also to estimate the apparent dissociation constant (KD app) of the interaction and the density of the recognized epitope on the bacteria. Herein, we found substantial differences within the KD app/epitope density profiles in mice immunized with various species of heat-killed bacteria. The experiments further revealed a high cross-reactivity of the secreted IgG repertoires, binding to even unrelated bacteria with high affinity. This application confirmed the ability to quantify the anti-bacterial antibody repertoire and the utility of the developed bioassay to study the interplay between bacteria and the humoral response.
Collapse
Affiliation(s)
- Millie Heo
- 'Laboratoire Colloïdes et Matériaux Divisés' (LCMD), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France
| | - Guilhem Chenon
- 'Laboratoire Colloïdes et Matériaux Divisés' (LCMD), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France
| | - Carlos Castrillon
- Unit of Antibodies in Therapy and Pathology, Institute Pasteur, UMR1222 INSERM, F-75015, Paris, France
- 'Laboratoire de Biochimie' (LBC), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France
| | - Jérôme Bibette
- 'Laboratoire Colloïdes et Matériaux Divisés' (LCMD), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France
| | - Pierre Bruhns
- Unit of Antibodies in Therapy and Pathology, Institute Pasteur, UMR1222 INSERM, F-75015, Paris, France
| | - Andrew D Griffiths
- 'Laboratoire de Biochimie' (LBC), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France
| | - Jean Baudry
- 'Laboratoire Colloïdes et Matériaux Divisés' (LCMD), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France
| | - Klaus Eyer
- 'Laboratoire Colloïdes et Matériaux Divisés' (LCMD), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France.
- Laboratory for Functional Immune Repertoire Analysis, Institute of Pharmaceutical Sciences, D-CHAB, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
10
|
Nasreen M, Dhouib R, Hosmer J, Wijesinghe HGS, Fletcher A, Mahawar M, Essilfie AT, Blackall PJ, McEwan AG, Kappler U. Peptide Methionine Sulfoxide Reductase from Haemophilus influenzae Is Required for Protection against HOCl and Affects the Host Response to Infection. ACS Infect Dis 2020; 6:1928-1939. [PMID: 32492342 DOI: 10.1021/acsinfecdis.0c00242] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Peptide methionine sulfoxide reductases (Msrs) are enzymes that repair ROS-damage to sulfur-containing amino acids such as methionine, ensuring functional integrity of cellular proteins. Here we have shown that unlike the majority of pro- and eukaryotic Msrs, the peptide methionine sulfoxide reductase (MsrAB) from the human pathobiont Haemophilus influenzae (Hi) is required for the repair of hypochlorite damage to cell envelope proteins, but more importantly, we were able to demonstrate that MsrAB plays a role in modulating the host immune response to Hi infection. Loss of MsrAB resulted in >1000-fold increase in sensitivity of Hi to HOCl-mediated killing, and also reduced biofilm formation and in-biofilm survival. Expression of msrAB was also induced by hydrogen peroxide and paraquat, but a Hi2019ΔmsrAB strain was not susceptible to killing by these ROS in vitro. Hi2019ΔmsrAB fitness in infection models was low, with a 3-fold reduction in intracellular survival in bronchial epithelial cells, increased susceptibility to neutrophil killing, and a 10-fold reduction in survival in a mouse model of lung infection. Interestingly, infection with Hi2019ΔmsrAB led to specific changes in the antibacterial response of human host cells, with genes encoding antimicrobial peptides (BPI, CAMP) upregulated between 4 and 9 fold compared to infection with Hi2019WT, and reduction in expression of two proteins with antiapoptotic functions (BIRC3, XIAP). Modulation of host immune responses is a novel role for an enzyme of this type and provides first insights into mechanisms by which MsrAB supports Hi survival in vivo.
Collapse
Affiliation(s)
- Marufa Nasreen
- Australian Infectious Disease Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Rabeb Dhouib
- Australian Infectious Disease Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Jennifer Hosmer
- Australian Infectious Disease Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Hewa Godage Sithija Wijesinghe
- Australian Infectious Disease Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Aidan Fletcher
- Australian Infectious Disease Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Manish Mahawar
- Australian Infectious Disease Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland 4072, Australia
- Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India
| | - Ama-Tawiah Essilfie
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland 4006, Australia
| | - Patrick J. Blackall
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Alastair G. McEwan
- Australian Infectious Disease Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Ulrike Kappler
- Australian Infectious Disease Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland 4072, Australia
| |
Collapse
|
11
|
Muda NM, Nasreen M, Dhouib R, Hosmer J, Hill J, Mahawar M, Schirra HJ, McEwan AG, Kappler U. Metabolic analyses reveal common adaptations in two invasive Haemophilus influenzae strains. Pathog Dis 2020; 77:5420469. [PMID: 30915434 DOI: 10.1093/femspd/ftz015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 03/19/2019] [Indexed: 01/22/2023] Open
Abstract
Non-typeable Haemophilus influenzae (NTHi) is a major pathogen in upper and lower respiratory tract infections in humans, and is increasingly also associated with invasive disease. We have examined two unrelated NTHi invasive disease isolates, R2866 and C188, in order to identify metabolic and physiological properties that distinguish them from respiratory tract disease isolates such as Hi2019. While the general use of the Hi metabolic network was similar across all three strains, the two invasive isolates secreted increased amounts of succinate, which can have anti-inflammatory properties. In addition, they showed a common shift in their carbon source utilization patterns, with strongly enhanced metabolism of nucleoside substrates, glucose and sialic acid. The latter two are major compounds present in blood and cerebrospinal fluid (CSF). Interestingly, C188 and R2866 also shared a reduced ability to invade or survive intracellularly in 16HBE14 bronchial epithelial cells relative to Hi2019 (4-fold (4 h), 25-fold (24 h) reduction). Altered metabolic properties, such as the ones observed here, could arise from genomic adaptations that NTHi undergo during infection. Together these data indicate that shifts in substrate preferences in otherwise conserved metabolic pathways may underlie strain niche specificity and thus have the potential to alter the outcomes of host-NTHi interactions.
Collapse
Affiliation(s)
- Noor Marian Muda
- School of Chemistry and Molecular Biosciences, Australian Infectious Disease Research Centre, Centre for Metals in Biology, The University of Queensland, St. Lucia QLD 4072, Australia
| | - Marufa Nasreen
- School of Chemistry and Molecular Biosciences, Australian Infectious Disease Research Centre, Centre for Metals in Biology, The University of Queensland, St. Lucia QLD 4072, Australia
| | - Rabeb Dhouib
- School of Chemistry and Molecular Biosciences, Australian Infectious Disease Research Centre, Centre for Metals in Biology, The University of Queensland, St. Lucia QLD 4072, Australia
| | - Jennifer Hosmer
- School of Chemistry and Molecular Biosciences, Australian Infectious Disease Research Centre, Centre for Metals in Biology, The University of Queensland, St. Lucia QLD 4072, Australia
| | - Julian Hill
- School of Chemistry and Molecular Biosciences, Australian Infectious Disease Research Centre, Centre for Metals in Biology, The University of Queensland, St. Lucia QLD 4072, Australia
| | - Manish Mahawar
- School of Chemistry and Molecular Biosciences, Australian Infectious Disease Research Centre, Centre for Metals in Biology, The University of Queensland, St. Lucia QLD 4072, Australia.,Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India
| | - Horst Joachim Schirra
- Centre for Advanced Imaging, The University of Queensland, St. Lucia QLD 4072, Australia
| | - Alastair G McEwan
- School of Chemistry and Molecular Biosciences, Australian Infectious Disease Research Centre, Centre for Metals in Biology, The University of Queensland, St. Lucia QLD 4072, Australia
| | - Ulrike Kappler
- School of Chemistry and Molecular Biosciences, Australian Infectious Disease Research Centre, Centre for Metals in Biology, The University of Queensland, St. Lucia QLD 4072, Australia
| |
Collapse
|
12
|
Nontypeable Haemophilus influenzae Has Evolved Preferential Use of N-Acetylneuraminic Acid as a Host Adaptation. mBio 2019; 10:mBio.00422-19. [PMID: 31064827 PMCID: PMC6509186 DOI: 10.1128/mbio.00422-19] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Host-adapted bacterial pathogens such as NTHi cannot survive out of their host environment and have evolved host-specific mechanisms to obtain nutrients and evade the immune response. Relatively few of these host adaptations have been characterized at the molecular level. NTHi utilizes sialic acid as a nutrient and also incorporates this sugar into LOS, which is important in biofilm formation and immune evasion. In the present study, we showed that NTHi has evolved to preferentially utilize the Neu5Ac form of sialic acid. This adaptation is due to the substrate preference of the enzyme CMP-Neu5Ac synthetase, which synthesizes the activated form of Neu5Ac for macromolecule biosynthesis. This adaptation allows NTHi to evade killing by a human antibody response against the nonhuman sialic acid Neu5Gc. Nontypeable Haemophilus influenzae (NTHi) is a Gram-negative bacterial pathogen that is adapted exclusively to human hosts. NTHi utilizes sialic acid from the host as a carbon source and as a terminal sugar on the outer membrane glycolipid lipooligosaccharide (LOS). Sialic acid expressed on LOS is critical in NTHi biofilm formation and immune evasion. There are two major forms of sialic acids in most mammals, N-acetylneuraminic acid (Neu5Ac) and N-glycolylneuraminic acid (Neu5Gc), the latter of which is derived from Neu5Ac. Humans lack the enzyme to convert Neu5Ac to Neu5Gc and do not express Neu5Gc in normal tissues; instead, Neu5Gc is recognized as a foreign antigen. A recent study showed that dietary Neu5Gc can be acquired by NTHi colonizing humans and then presented on LOS, which acts as an antigen for the initial induction of anti-Neu5Gc antibodies. Here we examined Neu5Gc uptake and presentation on NTHi LOS. We show that, although Neu5Gc and Neu5Ac are utilized equally well as sole carbon sources, Neu5Gc is not incorporated efficiently into LOS. When equal amounts of Neu5Gc and Neu5Ac are provided in culture media, there is ∼4-fold more Neu5Ac incorporated into LOS, suggesting a bias in a step of the LOS biosynthetic pathway. CMP-Neu5Ac synthetase (SiaB) was shown to have ∼4,000-fold-higher catalytic efficiency for Neu5Ac than for Neu5Gc. These data suggest that NTHi has adapted preferential utilization of Neu5Ac, thus avoiding presentation of the nonhuman Neu5Gc in the bacterial cell surface. The selective pressure for this adaptation may represent the human antibody response to the Neu5Gc xenoantigen.
Collapse
|
13
|
Micoli F, Costantino P, Adamo R. Potential targets for next generation antimicrobial glycoconjugate vaccines. FEMS Microbiol Rev 2018; 42:388-423. [PMID: 29547971 PMCID: PMC5995208 DOI: 10.1093/femsre/fuy011] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/13/2018] [Indexed: 12/21/2022] Open
Abstract
Cell surface carbohydrates have been proven optimal targets for vaccine development. Conjugation of polysaccharides to a carrier protein triggers a T-cell-dependent immune response to the glycan moiety. Licensed glycoconjugate vaccines are produced by chemical conjugation of capsular polysaccharides to prevent meningitis caused by meningococcus, pneumococcus and Haemophilus influenzae type b. However, other classes of carbohydrates (O-antigens, exopolysaccharides, wall/teichoic acids) represent attractive targets for developing vaccines. Recent analysis from WHO/CHO underpins alarming concern toward antibiotic-resistant bacteria, such as the so called ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter spp.) and additional pathogens such as Clostridium difficile and Group A Streptococcus. Fungal infections are also becoming increasingly invasive for immunocompromised patients or hospitalized individuals. Other emergencies could derive from bacteria which spread during environmental calamities (Vibrio cholerae) or with potential as bioterrorism weapons (Burkholderia pseudomallei and mallei, Francisella tularensis). Vaccination could aid reducing the use of broad-spectrum antibiotics and provide protection by herd immunity also to individuals who are not vaccinated. This review analyzes structural and functional differences of the polysaccharides exposed on the surface of emerging pathogenic bacteria, combined with medical need and technological feasibility of corresponding glycoconjugate vaccines.
Collapse
Affiliation(s)
- Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena
| | | | | |
Collapse
|
14
|
Dhouib R, Othman DSMP, Lin V, Lai XJ, Wijesinghe HGS, Essilfie AT, Davis A, Nasreen M, Bernhardt PV, Hansbro PM, McEwan AG, Kappler U. A Novel, Molybdenum-Containing Methionine Sulfoxide Reductase Supports Survival of Haemophilus influenzae in an In vivo Model of Infection. Front Microbiol 2016; 7:1743. [PMID: 27933034 PMCID: PMC5122715 DOI: 10.3389/fmicb.2016.01743] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 10/18/2016] [Indexed: 01/07/2023] Open
Abstract
Haemophilus influenzae is a host adapted human mucosal pathogen involved in a variety of acute and chronic respiratory tract infections, including chronic obstructive pulmonary disease and asthma, all of which rely on its ability to efficiently establish continuing interactions with the host. Here we report the characterization of a novel molybdenum enzyme, TorZ/MtsZ that supports interactions of H. influenzae with host cells during growth in oxygen-limited environments. Strains lacking TorZ/MtsZ showed a reduced ability to survive in contact with epithelial cells as shown by immunofluorescence microscopy and adherence/invasion assays. This included a reduction in the ability of the strain to invade human epithelial cells, a trait that could be linked to the persistence of H. influenzae. The observation that in a murine model of H. influenzae infection, strains lacking TorZ/MtsZ were almost undetectable after 72 h of infection, while ∼3.6 × 103 CFU/mL of the wild type strain were measured under the same conditions is consistent with this view. To understand how TorZ/MtsZ mediates this effect we purified and characterized the enzyme, and were able to show that it is an S- and N-oxide reductase with a stereospecificity for S-sulfoxides. The enzyme converts two physiologically relevant sulfoxides, biotin sulfoxide and methionine sulfoxide (MetSO), with the kinetic parameters suggesting that MetSO is the natural substrate of this enzyme. TorZ/MtsZ was unable to repair sulfoxides in oxidized Calmodulin, suggesting that a role in cell metabolism/energy generation and not protein repair is the key function of this enzyme. Phylogenetic analyses showed that H. influenzae TorZ/MtsZ is only distantly related to the Escherichia coli TorZ TMAO reductase, but instead is a representative of a new, previously uncharacterized clade of molybdenum enzyme that is widely distributed within the Pasteurellaceae family of pathogenic bacteria. It is likely that MtsZ/TorZ has a similar role in supporting host/pathogen interactions in other members of the Pasteurellaceae, which includes both human and animal pathogens.
Collapse
Affiliation(s)
- Rabeb Dhouib
- Centre for Metals in Biology/Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St. LuciaQLD, Australia
| | - Dk. Seti Maimonah Pg Othman
- Centre for Metals in Biology/Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St. LuciaQLD, Australia
| | - Victor Lin
- Centre for Metals in Biology/Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St. LuciaQLD, Australia
| | - Xuanjie J. Lai
- Centre for Metals in Biology/Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St. LuciaQLD, Australia
| | - Hewa G. S. Wijesinghe
- Centre for Metals in Biology/Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St. LuciaQLD, Australia
| | - Ama-Tawiah Essilfie
- Centre for Asthma and Respiratory Diseases, Hunter Medical Research Institute, The University of Newcastle, New LambtonNSW, Australia
| | - Amanda Davis
- Centre for Metals in Biology/Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St. LuciaQLD, Australia
- Department of Chemistry and Biochemistry, The University of Arizona, TucsonAZ, USA
| | - Marufa Nasreen
- Centre for Metals in Biology/Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St. LuciaQLD, Australia
| | - Paul V. Bernhardt
- Centre for Metals in Biology/Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St. LuciaQLD, Australia
| | - Philip M. Hansbro
- Centre for Asthma and Respiratory Diseases, Hunter Medical Research Institute, The University of Newcastle, New LambtonNSW, Australia
| | - Alastair G. McEwan
- Centre for Metals in Biology/Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St. LuciaQLD, Australia
| | - Ulrike Kappler
- Centre for Metals in Biology/Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St. LuciaQLD, Australia
| |
Collapse
|
15
|
Hartwig SM, Ketterer M, Apicella MA, Varga SM. Non-typeable Haemophilus influenzae protects human airway epithelial cells from a subsequent respiratory syncytial virus challenge. Virology 2016; 498:128-135. [PMID: 27573069 DOI: 10.1016/j.virol.2016.08.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 07/27/2016] [Accepted: 08/19/2016] [Indexed: 02/03/2023]
Abstract
Respiratory syncytial virus (RSV) and the common commensal and opportunistic pathogen, non-typeable Haemophilus influenzae (NTHi) both serve as a frequent cause of respiratory infection in children. Although it is well established that some respiratory viruses can increase host susceptibility to secondary bacterial infections, few studies have examined how commensal bacteria could influence a secondary viral response. Here, we examined the impact of NTHi exposure on a subsequent RSV infection of human bronchial epithelial cells (16HBE14o-). Co-culture of 16HBE14o- cells with NTHi resulted in inhibition of viral gene expression following RSV infection. 16HBE14o- cells co-cultured with heat-killed NTHi failed to protect against an RSV infection, indicating that protection requires live bacteria. However, NTHi did not inhibit influenza A virus replication, indicating that NTHi-mediated protection was RSV-specific. Our data demonstrates that prior exposure to a commensal bacterium such as NTHi can elicit protection against a subsequent RSV infection.
Collapse
Affiliation(s)
- Stacey M Hartwig
- Department of Microbiology, University of Iowa, Iowa City, IA, United States
| | - Margaret Ketterer
- Department of Microbiology, University of Iowa, Iowa City, IA, United States
| | - Michael A Apicella
- Department of Microbiology, University of Iowa, Iowa City, IA, United States
| | - Steven M Varga
- Department of Microbiology, University of Iowa, Iowa City, IA, United States; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States; Department of Pathology, University of Iowa, Iowa City, IA, United States.
| |
Collapse
|
16
|
Ketterer MR, Rice PA, Gulati S, Kiel S, Byerly L, Fortenberry JD, Soper DE, Apicella MA. Desialylation of Neisseria gonorrhoeae Lipooligosaccharide by Cervicovaginal Microbiome Sialidases: The Potential for Enhancing Infectivity in Men. J Infect Dis 2016; 214:1621-1628. [PMID: 27471322 DOI: 10.1093/infdis/jiw329] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 07/07/2016] [Indexed: 12/22/2022] Open
Abstract
Previous studies have demonstrated that Neisseria gonorrhoeae sialylates the terminal N-acetyllactosamine present on its lipooligosaccharide (LOS) by acquiring CMP-N-acetyl-5-neuraminic acid upon entering human cells during infection. This renders the organism resistant to killing by complement in normal human serum. N-acetyllactosamine residues on LOS must be free of N-acetyl-5-neuraminc acid (Neu5Ac; also known as "sialic acid") in order for organisms to bind to and enter urethral epithelial cells during infection in men. This raises the question of how the gonococcus infects men if N-acetyllactosamine residues are substituted by Neu5Ac during infection in women. Here, we demonstrate that women with gonococcal infections have levels of sialidases present in cervicovaginal secretions that can result in desialylation of (sialylated) gonococcal LOS. The principle sialidases responsible for this desialylation appear to be bacterial in origin. These studies suggest that members of the cervicovaginal microbiome can modify N. gonorrhoeae, which will enhance successful transmission to men.
Collapse
Affiliation(s)
- Margaret R Ketterer
- Department of Microbiology, University of Iowa Carver College of Medicine, Iowa City
| | - Peter A Rice
- Department of Medicine/Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester
| | - Sunita Gulati
- Department of Medicine/Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester
| | - Steven Kiel
- Department of Microbiology, University of Iowa Carver College of Medicine, Iowa City
| | - Luke Byerly
- Department of Microbiology, University of Iowa Carver College of Medicine, Iowa City
| | | | - David E Soper
- Division of Obstetric and Gynecologic Specialists, Medical College of South Carolina Hospital, Charleston
| | - Michael A Apicella
- Department of Microbiology, University of Iowa Carver College of Medicine, Iowa City
| |
Collapse
|
17
|
Khan MN, Ren D, Kaur R, Basha S, Zagursky R, Pichichero ME. Developing a vaccine to prevent otitis media caused by nontypeable Haemophilus influenzae. Expert Rev Vaccines 2016; 15:863-78. [PMID: 26894630 DOI: 10.1586/14760584.2016.1156539] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Nontypeable Haemophilus influenzae (NTHi) is a predominant organism of the upper respiratory nasopharyngeal microbiota. Its disease spectrum includes otitis media, sinusitis, non-bacteremic pneumonia and invasive infections. Protein-based vaccines to prevent NTHi infections are needed to alleviate these infections in children and vulnerable populations such as the elderly and those with chronic obstructive pulmonary disease (COPD). One NTHi protein is included in a pneumococcal conjugate vaccine and has been shown to provide efficacy. Our lab has been interested in understanding the immunogenicity of NTHi vaccine candidates P6, protein D and OMP26 for preventing acute otitis media in young children. We expect that continued investigation and progress in the development of an efficacious protein based vaccine against NTHi infections is achievable in the near future.
Collapse
Affiliation(s)
- M Nadeem Khan
- a Center for Infectious Disease and Immunology , Rochester General Hospital Research Institute , Rochester , NY , USA
| | - Dabin Ren
- a Center for Infectious Disease and Immunology , Rochester General Hospital Research Institute , Rochester , NY , USA
| | - Ravinder Kaur
- a Center for Infectious Disease and Immunology , Rochester General Hospital Research Institute , Rochester , NY , USA
| | - Saleem Basha
- a Center for Infectious Disease and Immunology , Rochester General Hospital Research Institute , Rochester , NY , USA
| | - Robert Zagursky
- a Center for Infectious Disease and Immunology , Rochester General Hospital Research Institute , Rochester , NY , USA
| | - Michael E Pichichero
- a Center for Infectious Disease and Immunology , Rochester General Hospital Research Institute , Rochester , NY , USA
| |
Collapse
|
18
|
Dhouib R, Pg Othman DSM, Essilfie AT, Hansbro PM, Hanson JO, McEwan AG, Kappler U. Maturation of molybdoenzymes and its influence on the pathogenesis of non-typeable Haemophilus influenzae. Front Microbiol 2015; 6:1219. [PMID: 26594204 PMCID: PMC4633490 DOI: 10.3389/fmicb.2015.01219] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 10/19/2015] [Indexed: 01/08/2023] Open
Abstract
Mononuclear molybdenum enzymes of the dimethylsulfoxide (DMSO) reductase family occur exclusively in prokaryotes, and a loss of some these enzymes has been linked to a loss of bacterial virulence in several cases. The MobA protein catalyzes the final step in the synthesis of the molybdenum guanine dinucleotide (MGD) cofactor that is exclusive to enzymes of the DMSO reductase family. MobA has been proposed as a potential target for control of virulence since its inhibition would affect the activities of all molybdoenzymes dependent upon MGD. Here, we have studied the phenotype of a mobA mutant of the host-adapted human pathogen Haemophilus influenzae. H. influenzae causes and contributes to a variety of acute and chronic diseases of the respiratory tract, and several enzymes of the DMSO reductase family are conserved and highly expressed in this bacterium. The mobA mutation caused a significant decrease in the activities of all Mo-enzymes present, and also resulted in a small defect in anaerobic growth. However, we did not detect a defect in in vitro biofilm formation nor in invasion and adherence to human epithelial cells in tissue culture compared to the wild-type. In a murine in vivo model, the mobA mutant showed only a mild attenuation compared to the wild-type. In summary, our data show that MobA is essential for the activities of molybdenum enzymes, but does not appear to affect the fitness of H. influenzae. These results suggest that MobA is unlikely to be a useful target for antimicrobials, at least for the purpose of treating H. influenzae infections.
Collapse
Affiliation(s)
- Rabeb Dhouib
- Centre for Metals in Biology, Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland St. Lucia, QLD, Australia
| | - Dk S M Pg Othman
- Centre for Metals in Biology, Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland St. Lucia, QLD, Australia
| | - Ama-Tawiah Essilfie
- Centre for Asthma and Respiratory Diseases and Hunter Medical Research Institute, The University of Newcastle Newcastle, NSW, Australia
| | - Phil M Hansbro
- Centre for Asthma and Respiratory Diseases and Hunter Medical Research Institute, The University of Newcastle Newcastle, NSW, Australia
| | - Jeffrey O Hanson
- School of Biological Sciences, The University of Queensland St. Lucia, QLD, Australia
| | - Alastair G McEwan
- Centre for Metals in Biology, Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland St. Lucia, QLD, Australia
| | - Ulrike Kappler
- Centre for Metals in Biology, Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland St. Lucia, QLD, Australia
| |
Collapse
|
19
|
Post DMB, Held JM, Ketterer MR, Phillips NJ, Sahu A, Apicella MA, Gibson BW. Comparative analyses of proteins from Haemophilus influenzae biofilm and planktonic populations using metabolic labeling and mass spectrometry. BMC Microbiol 2014; 14:329. [PMID: 25551439 PMCID: PMC4302520 DOI: 10.1186/s12866-014-0329-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/16/2014] [Indexed: 11/25/2022] Open
Abstract
Background Non-typeable H. influenzae (NTHi) is a nasopharyngeal commensal that can become an opportunistic pathogen causing infections such as otitis media, pneumonia, and bronchitis. NTHi is known to form biofilms. Resistance of bacterial biofilms to clearance by host defense mechanisms and antibiotic treatments is well-established. In the current study, we used stable isotope labeling by amino acids in cell culture (SILAC) to compare the proteomic profiles of NTHi biofilm and planktonic organisms. Duplicate continuous-flow growth chambers containing defined media with either “light” (L) isoleucine or “heavy” (H) 13C6-labeled isoleucine were used to grow planktonic (L) and biofilm (H) samples, respectively. Bacteria were removed from the chambers, mixed based on weight, and protein extracts were generated. Liquid chromatography-mass spectrometry (LC-MS) was performed on the tryptic peptides and 814 unique proteins were identified with 99% confidence. Results Comparisons of the NTHi biofilm to planktonic samples demonstrated that 127 proteins showed differential expression with p-values ≤0.05. Pathway analysis demonstrated that proteins involved in energy metabolism, protein synthesis, and purine, pyrimidine, nucleoside, and nucleotide processes showed a general trend of downregulation in the biofilm compared to planktonic organisms. Conversely, proteins involved in transcription, DNA metabolism, and fatty acid and phospholipid metabolism showed a general trend of upregulation under biofilm conditions. Selected reaction monitoring (SRM)-MS was used to validate a subset of these proteins; among these were aerobic respiration control protein ArcA, NAD nucleotidase and heme-binding protein A. Conclusions The present proteomic study indicates that the NTHi biofilm exists in a semi-dormant state with decreased energy metabolism and protein synthesis yet is still capable of managing oxidative stress and in acquiring necessary cofactors important for biofilm survival. Electronic supplementary material The online version of this article (doi:10.1186/s12866-014-0329-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Deborah M B Post
- The Buck Institute for Research on Aging, Novato, CA, 94945, USA.
| | - Jason M Held
- Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | | | - Nancy J Phillips
- The University of California San Francisco, San Francisco, CA, 94143, USA.
| | - Alexandria Sahu
- The Buck Institute for Research on Aging, Novato, CA, 94945, USA.
| | | | - Bradford W Gibson
- The Buck Institute for Research on Aging, Novato, CA, 94945, USA. .,The University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
20
|
Wu S, Li X, Gunawardana M, Maguire K, Guerrero-Given D, Schaudinn C, Wang C, Baum MM, Webster P. Beta- lactam antibiotics stimulate biofilm formation in non-typeable haemophilus influenzae by up-regulating carbohydrate metabolism. PLoS One 2014; 9:e99204. [PMID: 25007395 PMCID: PMC4090067 DOI: 10.1371/journal.pone.0099204] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 05/12/2014] [Indexed: 12/31/2022] Open
Abstract
Non-typeable Haemophilus influenzae (NTHi) is a common acute otitis media pathogen, with an incidence that is increased by previous antibiotic treatment. NTHi is also an emerging causative agent of other chronic infections in humans, some linked to morbidity, and all of which impose substantial treatment costs. In this study we explore the possibility that antibiotic exposure may stimulate biofilm formation by NTHi bacteria. We discovered that sub-inhibitory concentrations of beta-lactam antibiotic (i.e., amounts that partially inhibit bacterial growth) stimulated the biofilm-forming ability of NTHi strains, an effect that was strain and antibiotic dependent. When exposed to sub-inhibitory concentrations of beta-lactam antibiotics NTHi strains produced tightly packed biofilms with decreased numbers of culturable bacteria but increased biomass. The ratio of protein per unit weight of biofilm decreased as a result of antibiotic exposure. Antibiotic-stimulated biofilms had altered ultrastructure, and genes involved in glycogen production and transporter function were up regulated in response to antibiotic exposure. Down-regulated genes were linked to multiple metabolic processes but not those involved in stress response. Antibiotic-stimulated biofilm bacteria were more resistant to a lethal dose (10 µg/mL) of cefuroxime. Our results suggest that beta-lactam antibiotic exposure may act as a signaling molecule that promotes transformation into the biofilm phenotype. Loss of viable bacteria, increase in biofilm biomass and decreased protein production coupled with a concomitant up-regulation of genes involved with glycogen production might result in a biofilm of sessile, metabolically inactive bacteria sustained by stored glycogen. These biofilms may protect surviving bacteria from subsequent antibiotic challenges, and act as a reservoir of viable bacteria once antibiotic exposure has ended.
Collapse
Affiliation(s)
- Siva Wu
- Life Sciences Division, University of California, Berkeley, California, United States of America
| | - Xiaojin Li
- Molecular Diagnostic Laboratory, ApolloGen Inc., Irvine, California, United States of America
| | - Manjula Gunawardana
- Oak Crest Institute of Science, Pasadena, California, United States of America
| | - Kathleen Maguire
- University of California San Diego, San Diego, California, United States of America
| | | | | | - Charles Wang
- Center for Genomics and Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, United States of America
| | - Marc M. Baum
- Oak Crest Institute of Science, Pasadena, California, United States of America
| | - Paul Webster
- Oak Crest Institute of Science, Pasadena, California, United States of America
- Center for Electron Microscopy and Microanalysis, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
21
|
Othman DSMP, Schirra H, McEwan AG, Kappler U. Metabolic versatility in Haemophilus influenzae: a metabolomic and genomic analysis. Front Microbiol 2014; 5:69. [PMID: 24624122 PMCID: PMC3941224 DOI: 10.3389/fmicb.2014.00069] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 02/09/2014] [Indexed: 12/11/2022] Open
Abstract
Haemophilus influenzae is a host adapted human pathogen known to contribute to a variety of acute and chronic diseases of the upper and lower respiratory tract as well as the middle ear. At the sites of infection as well as during growth as a commensal the environmental conditions encountered by H. influenzae will vary significantly, especially in terms of oxygen availability, however, the mechanisms by which the bacteria can adapt their metabolism to cope with such changes have not been studied in detail. Using targeted metabolomics the spectrum of metabolites produced during growth of H. influenzae on glucose in RPMI-based medium was found to change from acetate as the main product during aerobic growth to formate as the major product during anaerobic growth. This change in end-product is likely caused by a switch in the major route of pyruvate degradation. Neither lactate nor succinate or fumarate were major products of H. influenzae growth under any condition studied. Gene expression studies and enzyme activity data revealed that despite an identical genetic makeup and very similar metabolite production profiles, H. influenzae strain Rd appeared to favor glucose degradation via the pentose phosphate pathway, while strain 2019, a clinical isolate, showed higher expression of enzymes involved in glycolysis. Components of the respiratory chain were most highly expressed during microaerophilic and anaerobic growth in both strains, but again clear differences existed in the expression of genes associated e.g., with NADH oxidation, nitrate and nitrite reduction in the two strains studied. Together our results indicate that H. influenzae uses a specialized type of metabolism that could be termed “respiration assisted fermentation” where the respiratory chain likely serves to alleviate redox imbalances caused by incomplete glucose oxidation, and at the same time provides a means of converting a variety of compounds including nitrite and nitrate that arise as part of the host defence mechanisms.
Collapse
Affiliation(s)
| | - Horst Schirra
- Centre for Advanced Imaging, The University of Queensland St. Lucia, QLD, Australia
| | - Alastair G McEwan
- School of Chemistry and Molecular Biosciences, The University of Queensland St. Lucia, QLD, Australia
| | - Ulrike Kappler
- School of Chemistry and Molecular Biosciences, The University of Queensland St. Lucia, QLD, Australia
| |
Collapse
|
22
|
Lichtenegger S, Bina I, Roier S, Bauernfeind S, Keidel K, Schild S, Anthony M, Reidl J. Characterization of lactate utilization and its implication on the physiology of Haemophilus influenzae. Int J Med Microbiol 2014; 304:490-8. [PMID: 24674911 PMCID: PMC4012139 DOI: 10.1016/j.ijmm.2014.02.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 02/10/2014] [Accepted: 02/20/2014] [Indexed: 01/03/2023] Open
Abstract
Haemophilus influenzae is a Gram-negative bacillus and a frequent commensal of the human nasopharynx. Earlier work demonstrated that in H. influenzae type b, l-lactate metabolism is associated with serum resistance and in vivo survival of the organism. To further gain insight into lactate utilization of the non-typeable (NTHi) isolate 2019 and laboratory prototype strain Rd KW20, deletion mutants of the l-lactate dehydrogenase (lctD) and permease (lctP) were generated and characterized. It is shown, that the apparent KM of l-lactate uptake is 20.1 μM as determined for strain Rd KW20. Comparison of the COPD isolate NTHi 2019-R with the corresponding lctP knockout strain for survival in human serum revealed no lactate dependent serum resistance. In contrast, we observed a 4-fold attenuation of the mutant strain in a murine model of nasopharyngeal colonization. Characterization of lctP transcriptional control shows that the lactate utilization system in H. influenzae is not an inductor inducible system. Rather negative feedback regulation was observed in the presence of l-lactate and this is dependent on the ArcAB regulatory system. Additionally, for 2019 it was found that lactate may have signaling function leading to increased cell growth in late log phase under conditions where no l-lactate is metabolized. This effect seems to be ArcA independent and was not observed in strain Rd KW20. We conclude that l-lactate is an important carbon-source and may act as host specific signal substrate which fine tunes the globally acting ArcAB regulon and may additionally affect a yet unknown signaling system and thus may contribute to enhanced in vivo survival.
Collapse
Affiliation(s)
- Sabine Lichtenegger
- Institute of Molecular Biosciences, University of Graz, Humboldtstr. 50, 8010 Graz, Austria
| | - Isabelle Bina
- Institute of Molecular Biosciences, University of Graz, Humboldtstr. 50, 8010 Graz, Austria
| | - Sandro Roier
- Institute of Molecular Biosciences, University of Graz, Humboldtstr. 50, 8010 Graz, Austria
| | - Stilla Bauernfeind
- Insitute of Hygiene and Microbiology, University of Wuerzburg, Josef-Schneider Str. 2 E1, Wuerzburg 97080, Germany
| | - Kristina Keidel
- Insitute of Hygiene and Microbiology, University of Wuerzburg, Josef-Schneider Str. 2 E1, Wuerzburg 97080, Germany
| | - Stefan Schild
- Institute of Molecular Biosciences, University of Graz, Humboldtstr. 50, 8010 Graz, Austria
| | - Mark Anthony
- Department of Paediatrics, Oxford University Hospitals, Headington, Oxford OX3 9DU, United Kingdom
| | - Joachim Reidl
- Institute of Molecular Biosciences, University of Graz, Humboldtstr. 50, 8010 Graz, Austria.
| |
Collapse
|
23
|
Clementi EA, Wilhelm KR, Schleucher J, Morozova-Roche LA, Hakansson AP. A complex of equine lysozyme and oleic acid with bactericidal activity against Streptococcus pneumoniae. PLoS One 2013; 8:e80649. [PMID: 24260444 PMCID: PMC3832479 DOI: 10.1371/journal.pone.0080649] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 10/06/2013] [Indexed: 12/31/2022] Open
Abstract
HAMLET and ELOA are complexes consisting of oleic acid and two homologous, yet functionally different, proteins with cytotoxic activities against mammalian cells, with HAMLET showing higher tumor cells specificity, possibly due to the difference in propensity for oleic acid binding, as HAMLET binds 5-8 oleic acid molecules per protein molecule and ELOA binds 11-48 oleic acids. HAMLET has been shown to possess bactericidal activity against a number of bacterial species, particularly those with a respiratory tropism, with Streptococcus pneumoniae displaying the greatest degree of sensitivity. We show here that ELOA also displays bactericidal activity against pneumococci, which at lower concentrations shows mechanistic similarities to HAMLET’s bactericidal activity. ELOA binds to S. pneumoniae and causes perturbations of the plasma membrane, including depolarization and subsequent rupture, and activates an influx of calcium into the cells. Selective inhibition of calcium channels and sodium/calcium exchange activity significantly diminished ELOA’s bactericidal activity, similar to what we have observed with HAMLET. Finally, ELOA-induced death was also accompanied by DNA fragmentation into high molecular weight fragments – an apoptosis-like morphological phenotype that is seen during HAMLET-induced death. Thus, in contrast to different mechanisms of eukaryote cell death induced by ELOA and HAMLET, these complexes are characterized by rather similar activities towards bacteria. Although the majority of these events could be mimicked using oleic acid alone, the concentrations of oleic acid required were significantly higher than those present in the ELOA complex, and for some assays, the results were not identical between oleic acid alone and the ELOA complex. This indicates that the lipid, as a common denominator in both complexes, is an important component for the complexes’ bactericidal activities, while the proteins are required both to solubilize and/or present the lipid at the bacterial membrane and likely to confer other and separate functions during the bacterial death.
Collapse
Affiliation(s)
- Emily A. Clementi
- Department of Microbiology and Immunology, University at Buffalo (SUNY), Buffalo, New York, United States of America
| | - Kristina R. Wilhelm
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
| | - Jürgen Schleucher
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
| | | | - Anders P. Hakansson
- Department of Microbiology and Immunology, University at Buffalo (SUNY), Buffalo, New York, United States of America
- The Witebsky Center for Microbial Pathogenesis and Immunology, Buffalo, New York, United States of America
- The New York Center of Excellence for Bioinformatics and Life Sciences, University at Buffalo (SUNY), Buffalo, New York, United States of America
- * E-mail: (AH); (LMR)
| |
Collapse
|
24
|
Clark SE, Eichelberger KR, Weiser JN. Evasion of killing by human antibody and complement through multiple variations in the surface oligosaccharide of Haemophilus influenzae. Mol Microbiol 2013; 88:603-18. [PMID: 23577840 DOI: 10.1111/mmi.12214] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2013] [Indexed: 11/29/2022]
Abstract
The lipopolysaccharide (LPS) of H. influenzae is highly variable. Much of the structural diversity is derived from phase variation, or high frequency on-off switching, of molecules attached during LPS biosynthesis. In this study, we examined the dynamics of LPS phase variation following exposure to human serum as a source of antibody and complement in multiple H. influenzae isolates. We show that lic2A, lgtC and lex2A switch from phase-off to phase-on following serial passage in human serum. These genes, which control attachment of a galα1-4gal di-galactoside structure (lic2A and lgtC phase-on) or an alternative glucose extension (lex2A phase-on) from the same hexose moiety, reduce binding of bactericidal antibody to conserved inner core LPS structures. The effects of the di-galactoside and alternative glucose extension were also examined in the context of the additional LPS phase variable structures phosphorylcholine (ChoP) and sialic acid. We found that di-galactoside, the alternative glucose extension, ChoP, and sialic acid each contribute independently to bacterial survival in the presence of human complement, and have an additive effect in combination. We propose that LPS phase variable extensions serve to shield conserved inner core structures from recognition by host immune components encountered during infection.
Collapse
Affiliation(s)
- Sarah E Clark
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA
| | | | | |
Collapse
|
25
|
Vitiazeva V, Li J, Hood DW, Moxon ER, Schweda EKH. A novel branching pattern in the lipopolysaccharide expressed by non-typeable Haemophilus influenzae strain 1232. Carbohydr Res 2013; 378:114-22. [PMID: 23582099 DOI: 10.1016/j.carres.2013.03.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 03/12/2013] [Accepted: 03/14/2013] [Indexed: 11/27/2022]
Abstract
We report the novel branching pattern in lipopolysaccharide (LPS) expressed by non-typeable Haemophilus influenzae (NTHi) strain 1232. The strain expressed the β-d-Glcp-(1→4)-[α-d-Galp-(1→4)-β-d-Galp-(1→7)]-d-α-d-Hepp-(1→6)-β-d-Glcp chain linked to the proximal heptose (HepI) of the conserved triheptosyl inner-core moiety of NTHi LPS: l-α-d-HepIIIp-(1→2)-[PEtn→6]-l-α-d-HepIIp-(1→3)-l-α-d-HepIp-(1→5)-[PPEtn→4]-α-Kdop-(2→6)-lipid A. The structure has been elucidated using NMR spectroscopy, electrospray ionization mass spectrometry (ESI-MS) and capillary electrophoresis coupled to electrospray ionization tandem mass spectrometry (CE-ESI-MS(n)) on O-deacylated LPS and core oligosaccharide (OS) materials, as well as HPLC-ESI-MS(n) on permethylated, dephosphorylated OS. It was also found that a tetrasaccharide unit bearing sialic acid [α-Neu5Ac-(2→3)-β-d-Galp-(1→4)-β-d-GlcNAcp-(1→3)-β-d-Galp-(1→] could substitute O-4 of the β-d-Glcp linked to HepI. In addition, the distal heptose (HepIII) was substituted by PCho→6-β-d-Galp-(1→ at the O-2 position.
Collapse
Affiliation(s)
- Varvara Vitiazeva
- Clinical Research Centre, Karolinska Institutet, NOVUM, S-141 86 Huddinge, Sweden
| | | | | | | | | |
Collapse
|
26
|
Characterization of a ferrous iron-responsive two-component system in nontypeable Haemophilus influenzae. J Bacteriol 2012; 194:6162-73. [PMID: 22961857 DOI: 10.1128/jb.01465-12] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Nontypeable Haemophilus influenzae (NTHI), an opportunistic pathogen that is commonly found in the human upper respiratory tract, has only four identified two-component signal transduction systems. One of these, an ortholog to the QseBC (quorum-sensing Escherichia coli) system, was characterized. This system, designated firRS, was found to be transcribed in an operon with a gene encoding a small, predicted periplasmic protein with an unknown function, ygiW. The ygiW-firRS operon exhibited a unique feature with an attenuator present between ygiW and firR that caused the ygiW transcript level to be 6-fold higher than the ygiW-firRS transcript level. FirRS induced expression of ygiW and firR, demonstrating that FirR is an autoactivator. Unlike the QseBC system of E. coli, FirRS does not respond to epinephrine or norepinephrine. FirRS signal transduction was stimulated when NTHI cultures were exposed to ferrous iron or zinc but was unresponsive to ferric iron. Notably, the ferrous iron-responsive activation only occurred when a putative iron-binding site in FirS and the key phosphorylation aspartate in FirR were intact. FirRS was also activated when cultures were exposed to cold shock. Mutants in ygiW, firR, and firS were attenuated during pulmonary infection, but not otitis media. These data demonstrate that the H. influenzae strain 2019 FirRS is a two-component regulatory system that senses ferrous iron and autoregulates its own operon.
Collapse
|
27
|
Intranasal immunization with nontypeable Haemophilus influenzae outer membrane vesicles induces cross-protective immunity in mice. PLoS One 2012; 7:e42664. [PMID: 22880074 PMCID: PMC3411803 DOI: 10.1371/journal.pone.0042664] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 07/10/2012] [Indexed: 01/29/2023] Open
Abstract
Haemophilus influenzae is a Gram-negative human-restricted bacterium that can act as a commensal and a pathogen of the respiratory tract. Especially nontypeable H. influenzae (NTHi) is a major threat to public health and is responsible for several infectious diseases in humans, such as pneumonia, sinusitis, and otitis media. Additionally, NTHi strains are highly associated with exacerbations in patients suffering from chronic obstructive pulmonary disease. Currently, there is no licensed vaccine against NTHi commercially available. Thus, this study investigated the utilization of outer membrane vesicles (OMVs) as a potential vaccine candidate against NTHi infections. We analyzed the immunogenic and protective properties of OMVs derived from various NTHi strains by means of nasopharyngeal immunization and colonization studies with BALB/c mice. The results presented herein demonstrate that an intranasal immunization with NTHi OMVs results in a robust and complex humoral and mucosal immune response. Immunoprecipitation revealed the most important immunogenic proteins, such as the heme utilization protein, protective surface antigen D15, heme binding protein A, and the outer membrane proteins P1, P2, P5 and P6. The induced immune response conferred not only protection against colonization with a homologous NTHi strain, which served as an OMV donor for the immunization mixtures, but also against a heterologous NTHi strain, whose OMVs were not part of the immunization mixtures. These findings indicate that OMVs derived from NTHi strains have a high potential to act as a vaccine against NTHi infections.
Collapse
|
28
|
Abstract
Sialic acids are a diverse family of monosaccharides widely expressed on all cell surfaces of vertebrates and so-called "higher" invertebrates, and on certain bacteria that interact with vertebrates. This overview surveys examples of biological roles of sialic acids in immunity, with emphasis on an evolutionary perspective. Given the breadth of the subject, the treatment of individual topics is brief. Subjects discussed include biophysical effects regulation of factor H; modulation of leukocyte trafficking via selectins; Siglecs in immune cell activation; sialic acids as ligands for microbes; impact of microbial and endogenous sialidases on immune cell responses; pathogen molecular mimicry of host sialic acids; Siglec recognition of sialylated pathogens; bacteriophage recognition of microbial sialic acids; polysialic acid modulation of immune cells; sialic acids as pathogen decoys or biological masks; modulation of immunity by sialic acid O-acetylation; sialic acids as antigens and xeno-autoantigens; antisialoglycan antibodies in reproductive incompatibility; and sialic-acid-based blood groups.
Collapse
Affiliation(s)
- Ajit Varki
- Glycobiology Research and Training Center, Department of Medicine, University of California at San Diego, La Jolla, 92093-0687, USA.
| | | |
Collapse
|
29
|
Arrevillaga G, Gaona J, Sánchez C, Rosales V, Gómez B. Respiratory syncytial virus persistence in macrophages downregulates intercellular adhesion molecule-1 expression and reduces adhesion of non-typeable Haemophilus influenzae. Intervirology 2012; 55:442-50. [PMID: 22572178 DOI: 10.1159/000335548] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 11/30/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Persistence of respiratory syncytial virus (RSV) has been associated with episodes of chronic obstructive pulmonary disease (COPD); furthermore, co-infection of RSV with non-typeable Haemophilus influenzae (NTHi) is increasingly recognized as a cause of exacerbations of COPD. OBJECTIVE To study whether RSV persistence in a macrophage (Mφ)-like cell line alters NTHi uptake (adhesion and ingestion). METHODS A murine Mφ-like cell line persistently infected with RSV (MφP) was used. The effects of RSV persistence on NTHi uptake by MφP and mock-infected Mφ (MφN) were determined by flow-cytometric assays with NTHi labelled with either ethidium bromide or FITC. Expression of intercellular adhesion molecule-1 (ICAM-1), a ligand for NTHi, was determined by measuring mRNA through real-time PCR and protein by Western blot assays. RESULTS RSV persistence reduced both the capacity of Mφ to take up bacteria and the expression of ICAM-1 mRNA and protein. Furthermore, when ICAM-1 was blocked with anti-ICAM-1 antibody, the adhesion capacity of NTHi was significantly reduced for MφN, whereas for MφP the effect was less evident, implying that ICAM-1 participates in NTHi adhesion to Mφ. CONCLUSION RSV persistence in murine Mφ diminishes their capacity to adhere and ingest NTHi through downregulation of ICAM-1 expression at the transcriptional level.
Collapse
Affiliation(s)
- Gerardo Arrevillaga
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad Universitaria, México D.F., México
| | | | | | | | | |
Collapse
|
30
|
Phosphorylcholine allows for evasion of bactericidal antibody by Haemophilus influenzae. PLoS Pathog 2012; 8:e1002521. [PMID: 22396641 PMCID: PMC3291618 DOI: 10.1371/journal.ppat.1002521] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 12/20/2011] [Indexed: 11/19/2022] Open
Abstract
The human pathogen Haemophilus influenzae has the ability to quickly adapt to different host environments through phase variation of multiple structures on its lipooligosaccharide (LPS), including phosphorylcholine (ChoP). During colonization with H. influenzae, there is a selection for ChoP+ phase variants. In a murine model of nasopharyngeal colonization, this selection is lost in the absence of adaptive immunity. Based on previous data highlighting the importance of natural antibody in limiting H. influenzae colonization, the effect of ChoP expression on antibody binding and its bactericidal activity was investigated. Flow cytometric analysis revealed that ChoP+ phase variants had decreased binding of antibody to LPS epitopes compared to ChoP- phase variants. This difference in antibody binding correlated with increased survival of ChoP+ phase variants in the presence of antibody-dependent, complement-mediated killing. ChoP+ phase variants were also more resistant to trypsin digestion, suggesting a general effect on the physical properties of the outer membrane. Moreover, ChoP-mediated protection against antibody binding correlated with increased resilience of outer membrane integrity. Collectively, these data suggest that ChoP expression provides a selective advantage during colonization through ChoP-mediated effects on the accessibility of bactericidal antibody to the cell surface.
Collapse
|
31
|
Infant rat infection modifies phenotypic properties of an invasive nontypeable Haemophilus influenzae. Microbes Infect 2011; 14:509-16. [PMID: 22222846 DOI: 10.1016/j.micinf.2011.12.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 12/16/2011] [Accepted: 12/21/2011] [Indexed: 11/21/2022]
Abstract
Enhancing the virulence trait of a specific bacterium in an animal model is often performed prior to the use of the strain for ex vivo human studies, such as reactivity with complement and antibody, or with phagocytic cells. For example, in Streptococcus pneumoniae mouse passage is used to enhance capsule production. While investigating an unusual serum-resistant unencapsulated Haemophilus influenzae (R2866), we found that animal passage yielded an isolate (R3392) which had decreased resistance to human serum, but increased virulence in Chang conjunctival cell monolayers, but with less invasion and transcytosis of polar H292 cells. We examined 90 colonies recovered from three infant rats for phase variants of LPS biosynthetic genes. In 88 colonies lgtC was OFF due to tetrameric repeat mediated slipped-strand mispairing at the time of DNA replication, while there was no variation in lic1A, lic2A, lic3A, lexA and oaf A. With lgtC OFF the LPS lacks Galα1-4βGal, an epitope mimicking the human p(k) blood group, and molecular mimicry is lost. Selection for strain susceptible to NHS in the infant rat was not antibody mediated. We conclude that the passage of pathogens virulent in humans and animals may select for phenotypes only relevant for the animal species used.
Collapse
|
32
|
Johnston JW. An improved counterselection cassette for use in Haemophilus influenzae. Gene 2011; 492:325-8. [PMID: 22037605 DOI: 10.1016/j.gene.2011.10.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2011] [Revised: 09/06/2011] [Accepted: 10/04/2011] [Indexed: 11/19/2022]
Abstract
Counterselectable cassettes are extremely useful in molecular biology and allow for the creation of unmarked deletion mutants or the introduction of point mutations. I have constructed an inducible sacB cassette, using the tetracycline repressor. When used in tandem with a kanamycin-resistance marker, the cassette was successful in creating unmarked mutants in Haemophilus influenzae. The inducible nature of the cassette avoids some of the common problems associated with the utilization of sacB in counterselection.
Collapse
Affiliation(s)
- Jason W Johnston
- Department of Microbiology, Immunology, & Molecular Genetics, University of Kentucky, MS-423 Chandler Medical Center, Lexington, KY 40536-0298, USA.
| |
Collapse
|
33
|
Twelkmeyer B, Burström PK, Li J, Richard Moxon E, Hood DW, Schweda EK. Expression of a new disialyllacto structure in the lipopolysaccharide of non-typeable Haemophilus influenzae. Carbohydr Res 2011; 346:1885-97. [DOI: 10.1016/j.carres.2011.05.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 05/17/2011] [Accepted: 05/19/2011] [Indexed: 10/18/2022]
|
34
|
Engskog MKR, Deadman M, Li J, Hood DW, Schweda EKH. Detailed structural features of lipopolysaccharide glycoforms in nontypeable Haemophilus influenzae strain 2019. Carbohydr Res 2011; 346:1241-9. [PMID: 21571259 DOI: 10.1016/j.carres.2011.04.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Revised: 04/13/2011] [Accepted: 04/19/2011] [Indexed: 11/16/2022]
Abstract
We have investigated the structure of the lipopolysaccharide (LPS) of nontypeable Haemophilus influenzae (NTHi) strain 2019, a prototype strain that is used for studies of NTHi biology and disease. Analysis of LPS from wild type and lex2B, lpt3 and pgm mutant strains using NMR techniques and ESI-MS on O-deacylated LPS and core oligosaccharide material (OS), as well as ESI-MS(n) on permethylated dephosphorylated OS, confirmed the previously established structure in which lactose is linked to the proximal heptose (HepI) of the conserved triheptosyl inner-core moiety, l-α-D-Hepp-(1→2)-[PEtn→6]-l-α-D-Hepp-(1→3)-l-α-D-Hepp-(1→5)-[PPEtn→4]-α-Kdo-(2→6)-lipid A. Importantly, our data provide further structural detail whereby extensions from the middle heptose (HepII) are now characterized as β-D-Galp-(1→4)-β-D-Glcp-(1→4)-α-D-Glcp-(1→3 and truncated versions thereof. PEtn substitutes O-3 of the distal heptose (HepIII) of the inner-core moiety. This PEtn substituent was absent in the lpt3 mutant indicating that Lpt3 is the transferase required to add PEtn to the distal heptose. Interestingly, in the lex2B mutant strain HepIII was found to be substituted at O-2 by β-D-Glcp which, in turn, can be further extended. Contrary to previous findings, LPS of the pgm mutant strain contained minor glycoforms having β-D-Glcp linked to O-4 of HepI and also glycoforms with an additional PEtn which could be assigned to HepIII. Acetate groups and one glycine residue further substitute HepIII in NTHi 2019.
Collapse
Affiliation(s)
- Mikael K R Engskog
- Clinical Research Centre, University College of South Stockholm, Karolinska Institutet, Stockholm, NOVUM, S-141 86 Huddinge, Sweden
| | | | | | | | | |
Collapse
|
35
|
Hakansson AP, Roche-Hakansson H, Mossberg AK, Svanborg C. Apoptosis-like death in bacteria induced by HAMLET, a human milk lipid-protein complex. PLoS One 2011; 6:e17717. [PMID: 21423701 PMCID: PMC3053380 DOI: 10.1371/journal.pone.0017717] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Accepted: 02/11/2011] [Indexed: 11/20/2022] Open
Abstract
Background Apoptosis is the primary means for eliminating unwanted cells in multicellular organisms in order to preserve tissue homeostasis and function. It is characterized by distinct changes in the morphology of the dying cell that are orchestrated by a series of discrete biochemical events. Although there is evidence of primitive forms of programmed cell death also in prokaryotes, no information is available to suggest that prokaryotic death displays mechanistic similarities to the highly regulated programmed death of eukaryotic cells. In this study we compared the characteristics of tumor and bacterial cell death induced by HAMLET, a human milk complex of alpha-lactalbumin and oleic acid. Methodology/Principal Findings We show that HAMLET-treated bacteria undergo cell death with mechanistic and morphologic similarities to apoptotic death of tumor cells. In Jurkat cells and Streptococcus pneumoniae death was accompanied by apoptosis-like morphology such as cell shrinkage, DNA condensation, and DNA degradation into high molecular weight fragments of similar sizes, detected by field inverse gel electrophoresis. HAMLET was internalized into tumor cells and associated with mitochondria, causing a rapid depolarization of the mitochondrial membrane and bound to and induced depolarization of the pneumococcal membrane with similar kinetic and magnitude as in mitochondria. Membrane depolarization in both systems required calcium transport, and both tumor cells and bacteria were found to require serine protease activity (but not caspase activity) to execute cell death. Conclusions/Significance Our results suggest that many of the morphological changes and biochemical responses associated with apoptosis are present in prokaryotes. Identifying the mechanisms of bacterial cell death has the potential to reveal novel targets for future antimicrobial therapy and to further our understanding of core activation mechanisms of cell death in eukaryote cells.
Collapse
Affiliation(s)
- Anders P Hakansson
- Department of Microbiology and Immunology, University at Buffalo, State University of New York, Buffalo, New York, United States of America.
| | | | | | | |
Collapse
|
36
|
Nontypeable Haemophilus influenzae initiates formation of neutrophil extracellular traps. Infect Immun 2010; 79:431-8. [PMID: 20956567 DOI: 10.1128/iai.00660-10] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Nontypeable Haemophilus influenzae (NTHI) is a leading cause of otitis media infections, which are often chronic and/or recurrent in nature. NTHI and other bacterial species persist in vivo within biofilms during otitis media and other persistent infections. These biofilms have a significant host component that includes neutrophil extracellular traps (NETs). These NETs do not mediate clearance of NTHI, which survives within NET structures by means of specific subpopulations of lipooligosaccharides on the bacterial surface that are determinants of biofilm formation in vitro. In this study, the ability of NTHI and NTHI components to initiate NET formation was examined using an in vitro model system. Both viable and nonviable NTHI strains were shown to promote NET formation, as did preparations of bacterial DNA, outer membrane proteins, and lipooligosaccharide (endotoxin). However, only endotoxin from a parental strain of NTHI exhibited equivalent potency in NET formation to that of NTHI. Additional studies showed that NTHI entrapped within NET structures is resistant to both extracellular killing within NETs and phagocytic killing by incoming neutrophils, due to oligosaccharide moieties within the lipooligosaccharides. Thus, we concluded that NTHI elicits NET formation by means of multiple pathogen-associated molecular patterns (most notably endotoxin) and is highly resistant to killing within NET structures. These data support the conclusion that, for NTHI, formation of NET structures may be a persistence determinant by providing a niche within the middle-ear chamber.
Collapse
|
37
|
Johnston JW, Shamsulddin H, Miller AF, Apicella MA. Sialic acid transport and catabolism are cooperatively regulated by SiaR and CRP in nontypeable Haemophilus influenzae. BMC Microbiol 2010; 10:240. [PMID: 20843349 PMCID: PMC2946308 DOI: 10.1186/1471-2180-10-240] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Accepted: 09/15/2010] [Indexed: 11/12/2022] Open
Abstract
Background The transport and catabolism of sialic acid, a critical virulence factor for nontypeable Haemophilus influenzae, is regulated by two transcription factors, SiaR and CRP. Results Using a mutagenesis approach, glucosamine-6-phosphate (GlcN-6P) was identified as a co-activator for SiaR. Evidence for the cooperative regulation of both the sialic acid catabolic and transport operons suggested that cooperativity between SiaR and CRP is required for regulation. cAMP was unable to influence the expression of the catabolic operon in the absence of SiaR but was able to induce catabolic operon expression when both SiaR and GlcN-6P were present. Alteration of helical phasing supported this observation by uncoupling SiaR and CRP regulation. The insertion of one half-turn of DNA between the SiaR and CRP operators resulted in the loss of SiaR-mediated repression of the transport operon while eliminating cAMP-dependent induction of the catabolic operon when GlcN-6P was present. SiaR and CRP were found to bind to their respective operators simultaneously and GlcN-6P altered the interaction of SiaR with its operator. Conclusions These results suggest multiple novel features for the regulation of these two adjacent operons. SiaR functions as both a repressor and an activator and SiaR and CRP interact to regulate both operons from a single set of operators.
Collapse
Affiliation(s)
- Jason W Johnston
- Department of Microbiology, Immunology, and Molecular Genetics, The University of Kentucky, Lexington, KY, USA.
| | | | | | | |
Collapse
|
38
|
Ghaderi D, Taylor RE, Padler-Karavani V, Diaz S, Varki A. Implications of the presence of N-glycolylneuraminic acid in recombinant therapeutic glycoproteins. Nat Biotechnol 2010; 28:863-7. [PMID: 20657583 DOI: 10.1038/nbt.1651] [Citation(s) in RCA: 268] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Accepted: 05/24/2010] [Indexed: 12/22/2022]
Abstract
Recombinant glycoprotein therapeutics produced in nonhuman mammalian cell lines and/or with animal serum are often modified with the nonhuman sialic acid N-glycolylneuraminic acid (Neu5Gc; refs. 1,2). This documented contamination has generally been ignored in drug development because healthy individuals were not thought to react to Neu5Gc (ref. 2). However, recent findings indicate that all humans have Neu5Gc-specific antibodies, sometimes at high levels. Working with two monoclonal antibodies in clinical use, we demonstrate the presence of covalently bound Neu5Gc in cetuximab (Erbitux) but not panitumumab (Vectibix). Anti-Neu5Gc antibodies from healthy humans interact with cetuximab in a Neu5Gc-specific manner and generate immune complexes in vitro. Mice with a human-like defect in Neu5Gc synthesis generate antibodies to Neu5Gc after injection with cetuximab, and circulating anti-Neu5Gc antibodies can promote drug clearance. Finally, we show that the Neu5Gc content of cultured human and nonhuman cell lines and their secreted glycoproteins can be reduced by adding a human sialic acid to the culture medium. Our findings may be relevant to improving the half-life, efficacy and immunogenicity of glycoprotein therapeutics.
Collapse
Affiliation(s)
- Darius Ghaderi
- Glycobiology Research and Training Center, Department of Medicine and Cellular & Molecular Medicine, University of California, San Diego, La Jolla, California, USA
| | | | | | | | | |
Collapse
|
39
|
Taylor RE, Gregg CJ, Padler-Karavani V, Ghaderi D, Yu H, Huang S, Sorensen RU, Chen X, Inostroza J, Nizet V, Varki A. Novel mechanism for the generation of human xeno-autoantibodies against the nonhuman sialic acid N-glycolylneuraminic acid. ACTA ACUST UNITED AC 2010; 207:1637-46. [PMID: 20624889 PMCID: PMC2916132 DOI: 10.1084/jem.20100575] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The nonhuman sialic acid N-glycolylneuraminic acid (Neu5Gc) is metabolically incorporated into human tissues from certain mammalian-derived foods, and this occurs in the face of an anti-Neu5Gc “xeno-autoantibody” response. Given evidence that this process contributes to chronic inflammation in some diseases, it is important to understand when and how these antibodies are generated in humans. We show here that human anti-Neu5Gc antibodies appear during infancy and correlate with weaning and exposure to dietary Neu5Gc. However, dietary Neu5Gc alone cannot elicit anti-Neu5Gc antibodies in mice with a humanlike Neu5Gc deficiency. Other postnatally appearing anti-carbohydrate antibodies are likely induced by bacteria expressing these epitopes; however, no microbe is known to synthesize Neu5Gc. Here, we show that trace exogenous Neu5Gc can be incorporated into cell surface lipooligosaccharides (LOS) of nontypeable Haemophilus influenzae (NTHi), a human-specific commensal/pathogen. Indeed, infant anti-Neu5Gc antibodies appear coincident with antibodies against NTHi. Furthermore, NTHi that express Neu5Gc-containing LOS induce anti-Neu5Gc antibodies in Neu5Gc-deficient mice, without added adjuvant. Finally, Neu5Gc from baby food is taken up and expressed by NTHi. As the flora residing in the nasopharynx of infants can be in contact with ingested food, we propose a novel model for how NTHi and dietary Neu5Gc cooperate to generate anti-Neu5Gc antibodies in humans.
Collapse
Affiliation(s)
- Rachel E Taylor
- Department of Medicine, Skaggs School of Pharmacy and PharmaceuticalSciences, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Schweda EKH, Richards JC. Profiling LPS glycoforms of non-typeable Haemophilus influenzae by multiple-stage tandem mass spectrometry. Methods Mol Biol 2010; 600:79-92. [PMID: 19882122 DOI: 10.1007/978-1-60761-454-8_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Non-typeable (acapsular) Haemophilus influenzae (NTHi) is a major cause of otitis media accounting for 25-30% of all cases of the disease. Lipopolysaccharide (LPS) is an essential and exposed component of the H. influenzae cell wall. A characteristic feature of H. influenzae LPS is the extensive inter-strain and intra-strain heterogeneity of glycoform structure which is key to the role of the molecule in both commensal and disease-causing behavior of the bacterium. However, to characterize LPS structure unambiguously is a major challenge due to the extreme heterogeneity of glycoforms that certain strains express. A powerful tool for obtaining sequence and branching information is multiple-stage tandem ESI-MS (ESI-MS( n )) performed on dephosphorylated and permethylated oligosaccharide material using an ESI-quadrupole ion trap mass spectrometer. In general, permethylation increases the MS response by several orders of magnitude and sequence information is readily obtained since methyl tagging allows the distinction between fragment ions generated by cleavage of a single glycosidic bond and inner fragments resulting from the rupture of two glycosidic linkages. Using this approach we are now able to identify all isomeric glycoforms in very heterogeneous LPS preparations.
Collapse
Affiliation(s)
- Elke K H Schweda
- Clinical Research Centre, Karolinska Institute and Södertörn University, Huddinge, Sweden
| | | |
Collapse
|
41
|
LuxS promotes biofilm maturation and persistence of nontypeable haemophilus influenzae in vivo via modulation of lipooligosaccharides on the bacterial surface. Infect Immun 2009; 77:4081-91. [PMID: 19564381 DOI: 10.1128/iai.00320-09] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Nontypeable Haemophilus influenzae (NTHI) is an extremely common airway commensal which can cause opportunistic infections that are usually localized to airway mucosal surfaces. During many of these infections, NTHI forms biofilm communities that promote persistence in vivo. For many bacterial species, density-dependent quorum-signaling networks can affect biofilm formation and/or maturation. Mutation of luxS, a determinant of the autoinducer 2 (AI-2) quorum signal pathway, increases NTHI virulence in the chinchilla model for otitis media infections. For example, bacterial counts in middle-ear fluids and the severity of the host inflammatory response were increased in luxS mutants compared with parental strains. As these phenotypes are consistent with those that we have observed for biofilm-defective NTHI mutants, we hypothesized that luxS may affect NTHI biofilms. A luxS mutant was generated using the well-characterized NTHI 86-028NP strain and tested to determine the effects of the mutation on biofilm phenotypes in vitro and bacterial persistence and disease severity during experimental otitis media. Quantitation of the biofilm structure by confocal microscopy and COMSTAT analysis revealed significantly reduced biomass for NTHI 86-028NP luxS biofilms, which was restored by a soluble mediator in NTHI 86-028NP supernatants. Analysis of lipooligosaccharide moieties using an enzyme-linked immunosorbent assay and immunoblotting showed decreased levels of biofilm-associated glycoforms in the NTHI 86-028NP luxS strain. Infection studies showed that NTHI 86-028NP luxS had a significant persistence defect in vivo during chronic otitis media infection. Based on these data, we concluded that a luxS-dependent soluble mediator modulates the composition of the NTHI lipooligosaccharides, resulting in effects on biofilm maturation and bacterial persistence in vivo.
Collapse
|
42
|
Hong W, Juneau RA, Pang B, Swords WE. Survival of bacterial biofilms within neutrophil extracellular traps promotes nontypeable Haemophilus influenzae persistence in the chinchilla model for otitis media. J Innate Immun 2009; 1:215-24. [PMID: 20375579 DOI: 10.1159/000205937] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2008] [Accepted: 11/24/2008] [Indexed: 01/09/2023] Open
Abstract
Nontypeable Haemophilus influenzae (NTHi) is a leading cause of acute and chronic otitis media, which are a major public health problem worldwide. The persistence of NTHi during chronic and recurrent otitis media infections involves multicellular biofilm communities formed within the middle-ear chamber. Bacterial biofilms resist immune clearance and antibiotic therapy due in part to encasement within a polymeric matrix. In this study, the contribution of biofilms to bacterial persistence in vivo and composition of the NTHi biofilm matrix during experimental otitis media were investigated. The presence of biofilms within the chinchilla middle-ear chamber was significantly correlated with increased bacterial load in middle-ear effusions and tissue. Examination of thin sections revealed polymorphonuclear cells within a DNA lattice containing elastase and histones, which is consistent with the definition of neutrophil extracellular traps. Viable multicellular biofilm communities with biofilm phenotypes were found within the DNA lattice throughout the biofilm. Further, NTHi was resistant to both phagocytic and extracellular neutrophil killing in vitro by means of lipooligosaccharide moieties that promote biofilm formation. These data support the conclusion that NTHi subverts neutrophil extracellular traps to persist in vivo. These data also indicate that a more inclusive definition for biofilms may be warranted.
Collapse
Affiliation(s)
- Wenzhou Hong
- Department of Microbiology and Immunology, Wake Forest University Health Sciences, Winston-Salem, N.C., USA
| | | | | | | |
Collapse
|
43
|
Engskog MKR, Yildirim HH, Li J, Richards JC, Deadman M, Hood DW, Schweda EKH. A dual role for the lex2 locus: identification of galactosyltransferase activity in non-typeable Haemophilus influenzae strains 1124 and 2019. Carbohydr Res 2009; 344:632-41. [PMID: 19211098 DOI: 10.1016/j.carres.2009.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Accepted: 01/08/2009] [Indexed: 10/21/2022]
Abstract
Lipopolysaccharide (LPS) of Haemophilus influenzae comprises a conserved tri-l-glycero-d-manno-heptosyl inner-core moiety (l-alpha-d-Hepp-(1-->2)-[PEtn-->6]-l-alpha-d-Hepp-(1-->3)-[beta-d-GlcIp-(1-->4)]-l-alpha-d-Hepp-(1-->5)-alpha-Kdop) to which addition of beta-d-Glcp to O-4 of GlcI in serotype b strains is controlled by the gene lex2B. In non-typeable H. influenzae strains 1124 and 2019, however, a beta-d-Galp is linked to O-4 of GlcI. In order to test the hypothesis that the lex2 locus is involved in the expression of beta-d-Galp-(1-->4-beta-d-Glcp-(1--> from HepI, lex2B was inactivated in strains 1124 and 2019, and LPS glycoform populations from the resulting mutant strains were investigated. Detailed structural analyses using NMR techniques and electrospray-ionisation mass spectrometry (ESIMS) on O-deacylated LPS and core oligosaccharide material (OS), as well as ESIMS(n) on permethylated dephosphorylated OS, indicated both lex2B mutant strains to express only beta-d-Glcp extensions from HepI. This provides strong evidence that Lex2B functions as a galactosyltransferase adding a beta-d-Galp to O-4 of GlcI in these strains, indicating that allelic polymorphisms in the lex2B sequence direct alternative functions of the gene product.
Collapse
Affiliation(s)
- Mikael K R Engskog
- Clinical Research Centre, Karolinska Institutet and Södertörn University, NOVUM, S-141 86 Huddinge, Sweden
| | | | | | | | | | | | | |
Collapse
|
44
|
Tracy E, Ye F, Baker BD, Munson RS. Construction of non-polar mutants in Haemophilus influenzae using FLP recombinase technology. BMC Mol Biol 2008; 9:101. [PMID: 19014437 PMCID: PMC2625361 DOI: 10.1186/1471-2199-9-101] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2008] [Accepted: 11/11/2008] [Indexed: 05/25/2023] Open
Abstract
Background Nontypeable Haemophilus influenzae (NTHi) is a gram-negative bacterium that causes otitis media in children as well as other infections of the upper and lower respiratory tract in children and adults. We are employing genetic strategies to identify and characterize virulence determinants in NTHi. NTHi is naturally competent for transformation and thus construction of most mutants by common methodologies is relatively straightforward. However, new methodology was required in order to construct unmarked non-polar mutations in poorly expressed genes whose products are required for transformation. We have adapted the lambda red/FLP-recombinase-mediated strategy used in E. coli for use in NTHi. Results A cassette containing a spectinomycin resistance gene and an rpsL gene flanked by FRT sites was constructed. A PCR amplicon containing 50 base pairs of DNA homologous to the 5' and 3' ends of the gene to be disrupted and the cassette was generated, then recombineered into the target NTHi gene, cloned on a plasmid, using the lambda recombination proteins expressed in E. coli DY380. Thus, the gene of interest was replaced by the cassette. The construct was then transformed into a streptomycin resistant NTHi strain and mutants were selected on spectinomycin-containing growth media. A plasmid derived from pLS88 with a temperature sensitive replicon expressing the FLP recombinase gene under the control of the tet operator/repressor was constructed. This plasmid was electroporated into the NTHi mutant at the permissive temperature and FLP expression was induced using anhydrotetracycline. The recombinase recognizes the FRT sites and eliminates the antibiotic cassette by site-specific recombination, creating the unmarked non-polar mutation. The plasmid is cured by growth of cells at the restrictive temperature. Conclusion The products of the genes in the NTHi pilABCD operon are required for type IV pilus biogenesis and have a role in transformation. We demonstrated the utility of our methodology by the construction of a non-polar pilA mutation in NTHi strain 2019 and complementation of the mutation with a plasmid containing the pilA gene. Utilization of this approach allowed us to readily generate unmarked non-polar mutations in NTHi genes.
Collapse
Affiliation(s)
- Erin Tracy
- Center for Microbial Pathogenesis in The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.
| | | | | | | |
Collapse
|
45
|
Pang B, Hong W, West-Barnette SL, Kock ND, Swords WE. Diminished ICAM-1 expression and impaired pulmonary clearance of nontypeable Haemophilus influenzae in a mouse model of chronic obstructive pulmonary disease/emphysema. Infect Immun 2008; 76:4959-67. [PMID: 18794286 PMCID: PMC2573371 DOI: 10.1128/iai.00664-08] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2008] [Revised: 07/02/2008] [Accepted: 09/03/2008] [Indexed: 11/20/2022] Open
Abstract
The airways of patients with chronic obstructive pulmonary disease (COPD) are continually colonized with bacterial opportunists like nontypeable Haemophilus influenzae (NTHi), and a wealth of evidence indicates that changes in bacterial populations within the lung can influence the severity of COPD. In this study, we used a murine model for COPD/emphysema to test the hypothesis that COPD affects pulmonary clearance. Mice were treated with a pulmonary bolus of elastase, and as reported previously, the lungs of these mice were pathologically similar to those with COPD/emphysema at approximately 1 month posttreatment. Pulmonary clearance of NTHi was significantly impaired in elastase-treated versus mock-treated mice. While histopathologic analysis revealed minimal differences in localized lung inflammation between the two groups, lower levels of intercellular adhesion molecule 1 (ICAM-1) were observed for the airway epithelial surface of elastase-treated mice than for those of control mice. Following infection, elastase-treated mice had lung pathology consistent with pneumonia for as long as 72 h postinfection, whereas at the same time point, mock-treated mice had cleared NTHi and showed little apparent pathology. Large aggregates of bacteria were observed within damaged lung tissue of the elastase-treated mice, whereas sparse individual bacteria were observed in lungs of mock-treated mice at the same time point postinfection. Additional infection studies showed that NTHi mutants with biofilm defects were less persistent in the elastase-treated mice than the parent strain. These findings establish a model for COPD-related infections and support the hypotheses that ICAM-1 promotes clearance of NTHi. Furthermore, the data indicate that NTHi may form biofilms within the context of COPD-related infections.
Collapse
Affiliation(s)
- Bing Pang
- Departments of Microbiology and Immunology, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | | | | | | | | |
Collapse
|
46
|
Li J, Deadman ME, Hood DW, Moxon ER, Schweda EKH. Structural Analysis of the Lipopolysaccharide from Nontypeable Haemophilus influenzae Strain R2846. Biochemistry 2008; 47:6025-38. [DOI: 10.1021/bi702510b] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jianjun Li
- Clinical Research Centre, Karolinska Institutet and University College of South Stockholm, Novum, S-14186 Huddinge, Sweden, Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario, Canada, K1A 0R6, and Molecular Infectious Diseases Group, University of Oxford, Department of Paediatrics, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford OX3 9DS, U.K
| | - Mary E. Deadman
- Clinical Research Centre, Karolinska Institutet and University College of South Stockholm, Novum, S-14186 Huddinge, Sweden, Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario, Canada, K1A 0R6, and Molecular Infectious Diseases Group, University of Oxford, Department of Paediatrics, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford OX3 9DS, U.K
| | - Derek W. Hood
- Clinical Research Centre, Karolinska Institutet and University College of South Stockholm, Novum, S-14186 Huddinge, Sweden, Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario, Canada, K1A 0R6, and Molecular Infectious Diseases Group, University of Oxford, Department of Paediatrics, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford OX3 9DS, U.K
| | - E. Richard Moxon
- Clinical Research Centre, Karolinska Institutet and University College of South Stockholm, Novum, S-14186 Huddinge, Sweden, Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario, Canada, K1A 0R6, and Molecular Infectious Diseases Group, University of Oxford, Department of Paediatrics, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford OX3 9DS, U.K
| | - Elke K. H. Schweda
- Clinical Research Centre, Karolinska Institutet and University College of South Stockholm, Novum, S-14186 Huddinge, Sweden, Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario, Canada, K1A 0R6, and Molecular Infectious Diseases Group, University of Oxford, Department of Paediatrics, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford OX3 9DS, U.K
| |
Collapse
|
47
|
Pang B, Winn D, Johnson R, Hong W, West-Barnette S, Kock N, Swords WE. Lipooligosaccharides containing phosphorylcholine delay pulmonary clearance of nontypeable Haemophilus influenzae. Infect Immun 2008; 76:2037-43. [PMID: 18347044 PMCID: PMC2346676 DOI: 10.1128/iai.01716-07] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Revised: 01/25/2008] [Accepted: 03/06/2008] [Indexed: 11/20/2022] Open
Abstract
Nontypeable Haemophilus influenzae (NTHi) causes pulmonary infections in patients with chronic obstructive pulmonary disease and other mucociliary clearance defects. Like many bacteria inhabiting mucosal surfaces, NTHi produces lipooligosaccharide (LOS) endotoxins that lack the O side chain. Persistent NTHi populations express a discrete subset of LOS glycoforms, including those containing phosphorylcholine (PCho). In this study, we compared two NTHi strains with isogenic mutants lacking PCho for clearance from mice following pulmonary infection. Consistent with data from other model systems, populations of the strains NTHi 2019 and NTHi 86-028NP recovered from mouse lung contained an increased proportion of PCho+ variants compared to that in the inocula. PCho- mutants were more rapidly cleared. Serial passage of NTHi increased both PCho content and bacterial resistance to clearance, and no such increases were observed for PCho- mutants. Increased PCho content was also observed in NTHi populations within non-endotoxin-responsive C3H/HeJ and Toll-like receptor 4 null (TLR4-/-) mice, albeit at later times postinfection. Changes in bacterial subpopulations and clearance were unaffected in TLR2-/- mice compared to the subpopulations in and clearance from mice of the parental strain. The clearance of PCho- mutants occurred at earlier time points in both strain backgrounds and in all types of mice. Comparison of bacterial populations in lung tissue cryosections by immunofluorescent staining showed sparse bacteria within the air spaces of C57BL/6 mice and large bacterial aggregates within the lungs of MyD88-/- mice. These results indicate that PCho promotes bacterial resistance to pulmonary clearance early in infection in a manner that is at least partially independent of the TLR4 pathway.
Collapse
Affiliation(s)
- Bing Pang
- Department of Microbiology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Johnston JW, Coussens NP, Allen S, Houtman JC, Turner KH, Zaleski A, Ramaswamy S, Gibson BW, Apicella MA. Characterization of the N-Acetyl-5-neuraminic Acid-binding Site of the Extracytoplasmic Solute Receptor (SiaP) of Nontypeable Haemophilus influenzae Strain 2019. J Biol Chem 2008; 283:855-65. [DOI: 10.1074/jbc.m706603200] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
49
|
Johnston JW, Zaleski A, Allen S, Mootz JM, Armbruster D, Gibson BW, Apicella MA, Munson RS. Regulation of sialic acid transport and catabolism in Haemophilus influenzae. Mol Microbiol 2007; 66:26-39. [PMID: 17880422 DOI: 10.1111/j.1365-2958.2007.05890.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Virulence of nontypeable Haemophilus influenzae (NTHi) is dependent on the decoration of lipooligosaccharide with sialic acid. This sugar must be derived from the host, as NTHi cannot synthesize sialic acids. NTHi can also use sialic acid as a carbon source. The genes encoding the sialic acid transporter and the genes encoding the catabolic activities are localized to two divergently transcribed operons, the siaPT operon and the nan operon respectively. In this study, we identified SiaR as a repressor of sialic acid transport and catabolism in NTHi. Inactivation of siaR resulted in the unregulated expression of the genes in both operons. Unregulated catabolism of sialic acid in the siaR mutant resulted in the reduction of surface sialylation and an increase in serum sensitivity. In addition to SiaR-mediated repression, CRP, the cAMP receptor protein, was shown to activate expression of the siaPT operon but not the nan operon. We describe a model in which SiaR and CRP work to modulate intracellular sialic acid levels. Our results demonstrate the importance of SiaR-mediated regulation to balance the requirement of surface sialylation and the toxic accumulation of intracellular sialic acid.
Collapse
Affiliation(s)
- Jason W Johnston
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Subinhibitory concentrations of azithromycin decrease nontypeable Haemophilus influenzae biofilm formation and Diminish established biofilms. Antimicrob Agents Chemother 2007; 52:137-45. [PMID: 17954687 DOI: 10.1128/aac.00607-07] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Nontypeable Haemophilus influenzae (NTHi) commonly causes otitis media, chronic bronchitis in emphysema, and early airway infections in cystic fibrosis. Long-term, low-dose azithromycin has been shown to improve clinical outcomes in chronic lung diseases, although the mechanism of action remains unclear. The inhibition of bacterial biofilms by azithromycin has been postulated to be one mechanism mediating these effects. We hypothesized that subinhibitory concentrations of azithromycin would affect NTHi biofilm formation. Laboratory strains of NTHi expressing green fluorescent protein and azithromycin-resistant clinical isolates were grown in flow-cell and static-culture biofilm models. Using a range of concentrations of azithromycin and gentamicin, we measured the degree to which these antibiotics inhibited biofilm formation and persistence. Large biofilms formed over 2 to 4 days in a flow cell, displaying complex structures, including towers and channels. Subinhibitory concentrations of azithromycin significantly decreased biomass and maximal thickness in both forming and established NTHi biofilms. In contrast, subinhibitory concentrations of gentamicin had no effect on biofilm formation. Furthermore, established NTHi biofilms became resistant to gentamicin at concentrations far above the MIC. Biofilm formation of highly resistant clinical NTHi isolates (azithromycin MIC of > 64 microg/ml) was similarly decreased at subinhibitory azithromycin concentrations. Clinically obtainable azithromycin concentrations inhibited biofilms in all but the most highly resistant isolates. These data show that subinhibitory concentrations of azithromycin have antibiofilm properties, provide mechanistic insights, and supply an additional rationale for the use of azithromycin in chronic biofilm infections involving H. influenzae.
Collapse
|